link
stringlengths
57
71
question
stringlengths
428
3.74k
opa
stringlengths
6
200
opb
stringlengths
6
213
opc
stringlengths
3
177
opd
stringlengths
4
183
diagnosis
stringlengths
6
302
answer_idx
stringclasses
4 values
answer
stringlengths
3
213
explanation
stringlengths
1.52k
8.11k
field
stringclasses
10 values
case
stringlengths
405
3.72k
clinical_question
stringclasses
15 values
normalized_question
stringclasses
4 values
opa_shuffled
stringlengths
6
174
opb_shuffled
stringlengths
4
213
opc_shuffled
stringlengths
3
170
opd_shuffled
stringlengths
6
193
answer_idx_shuffled
stringclasses
4 values
test_image
int64
0
1
test_lab
int64
0
1
test_other
int64
0
1
figure
int64
0
1
gender
stringclasses
3 values
pregnancy
int64
0
1
woman_health
int64
0
1
age
float64
0
95
age_group
stringclasses
10 values
ethnicity
stringclasses
17 values
case_id
int64
1
1.52k
version
stringclasses
1 value
https://jamanetwork.com/journals/jamadermatology/fullarticle/2737326
A man in his 80s presented with an 18-month history of an irregularly linear, rusty-brown, atrophic, streaklike plaque extending from the tip of his right shoulder to the right biceps, where it ended in arborizing purple plaque with scattered, slightly indurated, irregular papules within it (Figure, A and B). There was no warmth, thrills, or pulsations across the affected area. The patient complained about a mild tingling sensation within the lesion. The patient could not think of any triggers or causes for his condition and denied any aggravating or alleviating factors as well as any treatment attempts. Complete review of systems yielded negative results except for easy bruising. He was taking finasteride, montelukast, enalapril, and metoprolol. His medical history was significant for right total reverse shoulder replacement 5 years prior. Apart from the previously described lesion, his physical examination was unremarkable, and he had full range of motion of his right shoulder. A punch biopsy of the slightly indurated papule from the involved area on the right biceps was completed (Figure, C and D).A and B, An irregularly linear, rusty-brown, atrophic, streaklike plaque extends from the tip of the right shoulder to the right biceps, where it ends in arborizing purple plaque with scattered, slightly indurated, irregular papules within it. C, Hematoxylin-eosin staining demonstrates a dermal infiltrate arranged in nodules with clefting between nodules; some inflammatory aggregates are also seen within dilated vessels. D, Immunohistochemical staining for CD68 antigen reveals that the infiltrate is composed mostly of histiocytes. What Is Your Diagnosis?
Linear morphea
Serpentine supravenous hyperpigmentation
Intraoperative cutaneous electrical injury (Lichtenberg figure)
Intralymphatic histiocytosis
D. Intralymphatic histiocytosis
D
Intralymphatic histiocytosis
Microscopic findings from the punch biopsy showed a dense, nodular infiltrate of lymphocytes and histiocytes (highlighted by immunohistochemical stain for CD68 antigen) throughout the dermis and within thin-walled lymphatic vessels (highlighted by immunohistochemical stain for D2-40 antigen). These are diagnostic findings of intralymphatic histiocytosis.Intralymphatic histiocytosis is a rare condition first described in 1994 as intravascular histiocytosis.1 It usually presents on limbs as ill-defined erythematous or reticulated plaques resembling livedo reticularis, and it may be associated with rheumatoid arthritis, malignant tumors (Merkel cell carcinoma, incidental finding at the periphery of the excision specimen2; breast carcinoma, in the mastectomy scar2; and lung adenocarcinoma, in the dermis overlying the soft tissue metastasis3), and orthopedic metal implants (such as in the present case).2-5 However, it may also occur without any other associated conditions.2 The pathogenesis is unclear, but it is hypothesized to be a nonspecific reaction to an injury such as trauma, including surgery; inflammation; or infiltrative cancers, all of which may cause impaired lymphatic flow.6 There is an attempt to include intralymphatic histiocytosis with other peculiar histiocytic proliferations occurring in various body sites under a unifying diagnosis of histiocytosis with raisinoid nuclei. All of these proliferations are benign and, except for the skin lesions, asymptomatic conditions.6 Mazloom and colleagues7 make a case that intralymphatic histiocytosis is a distinct entity from intravascular histiocytosis, which they include in the group of reactive angiomatoses.Linear morphea would not have arborizing appearance and would have entirely different histopathologic findings with dense collagen in the dermis, decreased adnexal structures, and sparse inflammation in the lower dermis and upper subcutaneous fat. Serpentine supravenous hyperpigmentation is a cutaneous adverse effect of chemotherapy infusion with fluorouracil, vinorelbine, fotemustine, and docetaxel.8 Histopathologic findings of the condition are very different from intralymphatic histiocytosis and may show interface dermatitis with isolated necrotic keratinocytes, papillary dermal edema, and superficial perivascular inflammatory infiltrates.9 Clinically, serpentine supravenous hyperpigmentation may look very similar to intralymphatic histiocytosis; however, it is self-limiting and resolves within several months. Intraoperative cutaneous electrical injury (Lichtenberg figure) could look similar to intralymphatic histiocytosis, but history of such injury is lacking in the present case, and histologic results were different in that one would expect to see necrosis of the epidermis and dermis, with epidermal and dermal cell nuclear elongation and homogenization of dermal collagen with vascular dilatation, congestion, hemorrhage, and thrombosis.A skin biopsy is needed to establish the diagnosis of intralymphatic histiocytosis with a characteristic histologic picture, as described in this case. In cases without clear association with rheumatoid arthritis or joint replacement, an underlying malignant tumor should be considered.2,3 Reported treatment options include topical and systemic steroids, intralesional triamcinolone acetonide and pressure bandaging,10 radiotherapy, pentoxifylline, cyclophosphamide, and arthrocentesis, all with variable success.2The patient in the present case declined any treatment, and his condition at last follow-up remained unchanged. The condition, though chronic and usually resistant to treatments, is deemed harmless.2
Dermatology
A man in his 80s presented with an 18-month history of an irregularly linear, rusty-brown, atrophic, streaklike plaque extending from the tip of his right shoulder to the right biceps, where it ended in arborizing purple plaque with scattered, slightly indurated, irregular papules within it (Figure, A and B). There was no warmth, thrills, or pulsations across the affected area. The patient complained about a mild tingling sensation within the lesion. The patient could not think of any triggers or causes for his condition and denied any aggravating or alleviating factors as well as any treatment attempts. Complete review of systems yielded negative results except for easy bruising. He was taking finasteride, montelukast, enalapril, and metoprolol. His medical history was significant for right total reverse shoulder replacement 5 years prior. Apart from the previously described lesion, his physical examination was unremarkable, and he had full range of motion of his right shoulder. A punch biopsy of the slightly indurated papule from the involved area on the right biceps was completed (Figure, C and D).A and B, An irregularly linear, rusty-brown, atrophic, streaklike plaque extends from the tip of the right shoulder to the right biceps, where it ends in arborizing purple plaque with scattered, slightly indurated, irregular papules within it. C, Hematoxylin-eosin staining demonstrates a dermal infiltrate arranged in nodules with clefting between nodules; some inflammatory aggregates are also seen within dilated vessels. D, Immunohistochemical staining for CD68 antigen reveals that the infiltrate is composed mostly of histiocytes.
what is your diagnosis?
What is your diagnosis?
Linear morphea
Intralymphatic histiocytosis
Intraoperative cutaneous electrical injury (Lichtenberg figure)
Serpentine supravenous hyperpigmentation
b
0
0
1
1
male
0
0
85
81-90
null
501
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2734253
A white man in his early 40s with a medical history of chronic lymphocytic leukemia (Rai stage 0) reported a 6-month history of a growing mass in his right axilla and weight loss. Physical examination revealed palpable right axillary lymphadenopathy. The patient was an active smoker (1 pack per day) with an unremarkable family history. A positron emission tomography scan showed a hypermetabolic right axillary lymph node that measured 5 × 5 cm as well as multiple liver lesions, the largest of which measured 8 cm. The results of a core needle biopsy specimen from the right axillary lymph node were consistent with malignant neoplasm, with immunohistochemistry staining positive for S100, HMB-45, and MART-1. He began pembrolizumab therapy.After starting treatment, the patient began to notice slowly worsening painless, nonpruritic, diffuse dark gray skin, nail beds, and eye discoloration (Figure). The results of laboratory studies revealed normal electrolytes, normal kidney function, mild elevation of liver function, and mild anemia with a hemoglobin level of 12 g/dL (for SI unit conversion, multiply by 10.0 to convert to liters). Computed tomography of the chest, abdomen, and pelvis showed worsening hepatic metastases and right axillary adenopathy. What Is Your Diagnosis?
Hemochromatosis
Diffuse melanosis cutis
Addison disease
Hyperpigmentation due to sun exposure
B. Diffuse melanosis cutis
B
Diffuse melanosis cutis
Diffuse melanosis cutis (DMC) is a rare condition characterized by the hyperpigmentation of skin and mucous membranes caused by increased melanin deposition in patients with malignant melanoma. According to a systematic review,1 fewer than 100 cases have been reported. Diffuse melanosis cutis presents more commonly in males (60% of cases) and white patients (95% of cases), with a median age at the time of diagnosis of about 50 years.1 This condition is also associated with melanuria in about 77% of cases.1 Usually, DMC is seen 12 months after an initial diagnosis of metastatic melanoma. Patients present with progressive and diffuse blue-gray hyperpigmentation of the skin and mucous membranes, leading to darkening of the skin, hair, peritoneal fluid, sputum, urine, and sometimes the internal organs.2Histopathologic findings include deposition of perivascular histiocytes filled with melanin and free pigment in the dermal connective tissue. The underlying pathogenesis for this condition is not fully understood. According to one hypothesis, when metastatic melanoma undergoes lysis by ischemia, immunological responses, or oncologic treatments, cells release melanin precursors into the bloodstream that undergo auto-oxidation, become ingested by histiocytes, and are then deposited in the dermis.3 Another hypothesis suggests that tumor cells release melanocyte peptide growth factors such as α melanocyte-stimulating hormone, which might be responsible for DMC. The role immunotherapy played in the manifestation of DMC in this case is unclear. One case series of patients who developed DMC after immunotherapy initiation concluded that DMC may be a negative predictor of response to anti–PD-1 (programmed cell death 1) treatment.4 In particular, it remains unclear whether immunotherapy promotes DMC development or just cannot control DMC that would inevitably develop.The discoloration of DMC is most prominently noted in sun-exposed areas, progressing in a cephalocaudal direction.2 Diffuse melanosis cutis is an ominous sign that is associated with a worse prognosis, with a median survival of 4 to 6 months after diagnosis.1,5 According to case reports,5,6 novel therapies that target a BRAF mutation and immunotherapies have shown a considerable survival benefit.Based on the results of the biopsy specimen, the patient was diagnosed with metastatic melanoma. Metastatic melanoma should be one of the differential diagnoses in patients presenting with unusual skin discoloration and/or dark-colored bodily secretions. Given this patient’s short survival after his initial presentation, prognosis should be discussed at the time of diagnosis. Other differential diagnoses include hemochromatosis, most notably the hereditary form, which demonstrates skin hyperpigmentation in most cases. Elevated liver function levels are more likely related to worsening hepatic metastases. In this case, hemochromatosis is less likely given the patient’s unremarkable family history and the absence of other clinical features of hemochromatosis, including arthropathy or diabetes. Addison disease, also known as primary adrenal insufficiency, can cause generalized brown hyperpigmentation. However, the absence of other features of adrenal insufficiency such as hypotension, hypoglycemia, and electrolyte abnormalities makes this diagnosis less likely. Hyperpigmentation due to sun exposure would be limited to sun-exposed areas and would not cause hyperpigmentation of nail beds and eyes, as is seen in this case.This patient’s treatment was changed to the combination of pembrolizumab and talimogene laherparepvec injection. However, the disease progressed and the patient declined further treatment. He was transitioned to hospice care and died 6 months after DMC onset.
Oncology
A white man in his early 40s with a medical history of chronic lymphocytic leukemia (Rai stage 0) reported a 6-month history of a growing mass in his right axilla and weight loss. Physical examination revealed palpable right axillary lymphadenopathy. The patient was an active smoker (1 pack per day) with an unremarkable family history. A positron emission tomography scan showed a hypermetabolic right axillary lymph node that measured 5 × 5 cm as well as multiple liver lesions, the largest of which measured 8 cm. The results of a core needle biopsy specimen from the right axillary lymph node were consistent with malignant neoplasm, with immunohistochemistry staining positive for S100, HMB-45, and MART-1. He began pembrolizumab therapy.After starting treatment, the patient began to notice slowly worsening painless, nonpruritic, diffuse dark gray skin, nail beds, and eye discoloration (Figure). The results of laboratory studies revealed normal electrolytes, normal kidney function, mild elevation of liver function, and mild anemia with a hemoglobin level of 12 g/dL (for SI unit conversion, multiply by 10.0 to convert to liters). Computed tomography of the chest, abdomen, and pelvis showed worsening hepatic metastases and right axillary adenopathy.
what is your diagnosis?
What is your diagnosis?
Diffuse melanosis cutis
Hemochromatosis
Hyperpigmentation due to sun exposure
Addison disease
a
1
1
1
1
male
0
0
42
41-50
White
502
original
https://jamanetwork.com/journals/jamacardiology/fullarticle/2738099
A 44-year-old woman with dilated cardiomyopathy while receiving maximally tolerated guideline-directed medical therapy presented after ventricular fibrillation arrest and subsequently underwent an uneventful implant of a subcutaneous cardioverter defibrillator (S-ICD; EMBLEM, version A219; Boston Scientific) for secondary prevention. The device was programmed with a conditional shock zone of 200 beats per minute and a shock zone of 230 beats per minute. Postprocedure chest radiography results showed appropriate device placement. Postimplant interrogation results showed appropriate device function. The patient was discharged the day after implant. Two weeks later, she presented to the emergency department (ED) after receiving several shocks from the device during the early morning, waking her up from sleeping. There were no reported symptoms associated with shortness of breath, chest pain, or syncope. Physical examination results revealed no signs of decompensated heart failure. Device interrogation results showed shock therapies delivered postimplant on days 3, 5, 6, and 10. The surface electrogram from the events, recorded at a paper speed of 25 millimeters per second and an amplitude of 2.5 mm/mvV) is shown in Figure 1. Repeated chest radiography at the ED showed appropriate device location, lead pin, and an absence of subcutaneous air trapping at the proximal and distal sense electrodes.Surface electrogram recorded by subcutaneous implantable cardioverter defibrillator from one of the events. Recorded at a paper speed of 25 millimeters per second and an amplitude of 25 mm/mV. The shock impedance was 57 ohms and the final shock polarity was standard.Administer anticoagulation and increase the dose of the β-blocker What Would You Do Next?
Administer anticoagulation and increase the dose of the β-blocker
Subcutaneous ICD reprogramming and observation
Administer a loading dose of amiodarone
Refer the patient for catheter ablation
Inappropriate S-ICD shock from the oversensing of the electrical noise
B
Subcutaneous ICD reprogramming and observation
The surface electrogram results from all the events shared similar characteristics and revealed an inappropriate ICD shock due to an oversensing of the electrical noise. The noise has the appearance of “flutter waves,” which terminated after the shock therapies. Initially, this noise could be interpreted as ventricular or atrial arrhythmia. There are few points used to accurately differentiate between appropriate and inappropriate therapy in this case. First, the marching intrinsic QRS complexes through tracing can be identified within this electrical rhythm with varying degrees of superimposition of the flutterlike wave on the R wave. Second, the morphology and amplitude of the flutter waves are irregular and variable in cycle length, suggesting that the flutter waves are unlikely to be caused by reentry mechanism. Third, the intrinsic ventricular rate in the range of 90 to 100 beats per minute remained the same before and after the shocks were administered.The S-ICD system uses 3 sensing electrodes for detection: (1) distal sensing, (2) proximal sensing, and (3) a pulse generator. These electrodes represent 3 vector projections of cardiac conduction: primary, secondary, and alternate (Figure 2). Based on the optimal QRS:T wave ratio, one of the vectors is selected by an automatic setup algorithm for use as a configuration for sensing and to avoid inappropriate therapy because of myopotentials, multiple counting (eg, double QRS and T-wave oversensing), or noise. However, inappropriate shocks remain a substantial problem in S-ICD.1 In this case, the secondary vector was automatically selected. The inappropriate shock event occurred during sleep within 1 to 2 weeks after the device implant. There were no identified sources of electromagnetic interference and no oversensing was detected at the time of presentation to the ED. Electrical noise occurring in the early postprocedural period can be seen with incomplete electrode insertion, subcutaneous air trapping in the midline or xiphoid incision, or fluid/air trapping within the device header1-3; however, subcutaneous air trapping commonly presents within 48 hours after the procedure. Reviewing all episodes, the noise decreased or was eliminated after the shocks. The shock or a sudden change in patient posture may result in a redistribution of fluid or escaped trapped air from the header. A damaged seal plug or a slightly loosened setscrew may be associated with the characteristic of the air and fluid transition. Over several days, the redistribution of fluid and air results in repeated episodes. However, this redistribution of fluid and air escape over time eventually replaces the air in the device header and the intermittent episodic electrical noise may subside.2 Therefore, observation can be a reasonable management of this condition. Additional mitigations at time of device implant include removing the torque wrench before the electrode header tug test and a gentle insertion and removal of the torque wrench with the seal plug. In the meantime, the patient’s device is reprogrammed to switch to the primary vector, thereby disabling the circuit involving the distal sensing electrode.Subcutaneous implantable cardioverter defibrillator sensing vectors superimposed to the chest radiography. A, An electrode superior to the sternal coil (distal sense electrode). B, An electrode inferior to the sternal defibrillation coil (proximal sense electrode). C, Pulse generator. These electrodes represent 3 vector projections of cardiac electrical conduction: primary, secondary, and alternate electrocardiogram (ECG) vectors. The device image at the left corner shows the terminal connectors of sensing electrode and coil.The primary vector could be used for this patient, as the R wave amplitude was suitable. The patient had no further episodes during follow-up.
Cardiology
A 44-year-old woman with dilated cardiomyopathy while receiving maximally tolerated guideline-directed medical therapy presented after ventricular fibrillation arrest and subsequently underwent an uneventful implant of a subcutaneous cardioverter defibrillator (S-ICD; EMBLEM, version A219; Boston Scientific) for secondary prevention. The device was programmed with a conditional shock zone of 200 beats per minute and a shock zone of 230 beats per minute. Postprocedure chest radiography results showed appropriate device placement. Postimplant interrogation results showed appropriate device function. The patient was discharged the day after implant. Two weeks later, she presented to the emergency department (ED) after receiving several shocks from the device during the early morning, waking her up from sleeping. There were no reported symptoms associated with shortness of breath, chest pain, or syncope. Physical examination results revealed no signs of decompensated heart failure. Device interrogation results showed shock therapies delivered postimplant on days 3, 5, 6, and 10. The surface electrogram from the events, recorded at a paper speed of 25 millimeters per second and an amplitude of 2.5 mm/mvV) is shown in Figure 1. Repeated chest radiography at the ED showed appropriate device location, lead pin, and an absence of subcutaneous air trapping at the proximal and distal sense electrodes.Surface electrogram recorded by subcutaneous implantable cardioverter defibrillator from one of the events. Recorded at a paper speed of 25 millimeters per second and an amplitude of 25 mm/mV. The shock impedance was 57 ohms and the final shock polarity was standard.Administer anticoagulation and increase the dose of the β-blocker
what would you do next?
What would you do next?
Administer anticoagulation and increase the dose of the β-blocker
Refer the patient for catheter ablation
Administer a loading dose of amiodarone
Subcutaneous ICD reprogramming and observation
d
0
0
0
1
female
0
0
44
41-50
null
503
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2734341
A young woman presented with a 1-year history of a right tongue mass that developed after initial bite trauma. The lesion waxed and waned but never completely healed; however, it greatly increased in size over 3 weeks before presentation. She reported constant, severe stabbing pain, right otalgia, and occasional swelling that caused oral dysphagia. She reported no constitutional symptoms or any history of tobacco or alcohol use. On examination, the mass was a 3 × 2-cm, exophytic and raised, plateaulike lesion on the right dorsolateral tongue with approximately 1 to 2 cm of surrounding induration (Figure, A).A, Right lateral tongue lesion at presentation. B, Spindled-cell proliferation with palisaded areas (Verocay bodies) and an associated inflammatory infiltrate with abundant eosinophils (hematoxylin-eosin, original magnification ×20).A biopsy was obtained that demonstrated ulceration, pseudoepitheliomatous hyperplasia, and abundant acute and chronic inflammation replete with eosinophils. The differential based on the biopsy included traumatic ulcerative granuloma with stromal eosinophilia and ulceration due to drug- or infection-related sources. In addition, the possibility of an unsampled submucosal mass with overlying ulceration was suggested by the pathologist who interpreted the biopsy findings. Given our clinical concern for an unsampled neoplasm, the patient subsequently underwent resection of the mass; histologic sections of the specimen are demonstrated in Figure, B. Immunohistochemistry studies for SOX-10 revealed positive staining in the proliferating spindle-shaped cells.Schwannoma with a traumatic ulcerative granuloma with stromal eosinophilialike reaction What Is Your Diagnosis?
Human papillomavirus–related squamous cell carcinoma
Schwannoma with a traumatic ulcerative granuloma with stromal eosinophilialike reaction
Ulcerated plexiform neurofibroma
Ulcerated granular cell tumor
B. Schwannoma with a traumatic ulcerative granuloma with stromal eosinophilialike reaction
B
Schwannoma with a traumatic ulcerative granuloma with stromal eosinophilialike reaction
This case demonstrates a unique example of a lateral tongue schwannoma with an associated traumatic ulcerative granuloma with stromal eosinophilia (TUGSE)–like reaction that produced a large, exophytic mass mimicking more clinically concerning lesions, such as squamous cell carcinoma.Schwannoma is a benign, peripheral nerve sheath tumor that can occur throughout the body, but most commonly develops in the head and neck.1 An oral tongue location has been a well-documented site for this tumor. The histologic features of schwannomas in an oral tongue location parallel those at other sites and consist of a spindled cell proliferation with hypercellular and hypocellular areas that refer to specific patterns known as Antoni A and Antoni B.1 In the hypercellular areas, there are foci with nuclear palisading known as Verocay bodies. Immunohistochemistry demonstrates that the spindled cells are strongly and diffusely positive for S-100 and SOX-10 and typically negative for markers of epithelial differentiation, such as cytokeratins.1 To our knowledge, a brisk inflammatory infiltrate, such as the kind seen with TUGSE, has not been reported to occur with schwannoma.Traumatic ulcerative granuloma with stromal eosinophilia is a rare, benign condition with a strong etiologic association with repetitive trauma.2,3 In addition, most lesions develop on the dorsal or lateral, mobile tongue, which readily contacts teeth or dentures.2 Also referred to as traumatic eosinophilic granuloma, TUGSE is a self-limiting ulcer. Its duration varies from weeks to months and, rarely up to a year before it resolves.2 Its gross appearance can be alarming and often mimics that of squamous cell carcinoma. Histologically, TUGSE is characterized by a diffuse, polymorphous cellular infiltrate with abundant eosinophils that infiltrate into the deep skeletal muscle fibers.4,5 Occasional large, atypical mononuclear cells that are CD30 positive and express T-cell antigens have been identified in TUGSE.2,6 Some cases also demonstrate T-cell clonality, leading some authors to suggest that a subset of lesions diagnosed as TUGSE may be within the spectrum of CD30-positive lymphoproliferative disease.6 Others consider TUGSE a benign entity that is most likely trauma related.2-5 If a trauma-related hypothesis for TUGSE is accepted, then it is conceivable that a traumatized neoplasm could develop secondary to TUGSE-like changes. We believe that this case illustrates this concept, as this patient’s history of biting the mass likely accounts for the TUGSE-like inflammatory changes associated with the schwannoma.Although the clinical presentation and young patient age are concerning for a human papillomavirus–related squamous cell carcinoma, these tumors occur most commonly in the oropharynx rather than the oral tongue.7 Although the patient described herein did not have an alcohol or tobacco use disorder, her presentation also raised the possibility of a non–HPV-related squamous cell carcinoma of the oral cavity; in our current era and patient population, there has been an increase in the incidence of these cancers.8Plexiform neurofibromas typically have a haphazard arrangement of spindled cells with buckled-shaped nuclei.9 The stroma of neurofibromas can vary from myxoid to hyalinized. While this tumor is also positive for S-100 and SOX-10, it is typically not as diffuse as seen in schwannoma.9 In addition, the Antoni A and Antoni B patterns and Verocay bodies are characteristic of schwannoma and not a typical feature of plexiform neurofibroma. Granular cell tumors are also S-100 and SOX-10 positive but are composed of plump, polygonal cells with abundant granular cytoplasm.10 Because plexiform neurofibromas and granular cell tumors cannot be distinguished clinically, histologic assessment is required for their distinction.Two years after presentation, there has been no recurrence of the lesion after en bloc resection and primary closure. Confirmed TUGSE with underlying schwannoma was found on final pathologic examination. This case reminds us that superficial biopsy samples of ulcerated elevated lesions may not adequately sample the underlying mass. The initial biopsy in this patient only contained the ulcer, inflammation, and squamous epithelium; the submucosa was not well represented, leading to a sampling pitfall. Given the elevated nature of the lesion, superficial initial sample, and both clinical and pathologic concern for an underlying neoplasm, surgical resection was undertaken and warranted in this particular case.
General
A young woman presented with a 1-year history of a right tongue mass that developed after initial bite trauma. The lesion waxed and waned but never completely healed; however, it greatly increased in size over 3 weeks before presentation. She reported constant, severe stabbing pain, right otalgia, and occasional swelling that caused oral dysphagia. She reported no constitutional symptoms or any history of tobacco or alcohol use. On examination, the mass was a 3 × 2-cm, exophytic and raised, plateaulike lesion on the right dorsolateral tongue with approximately 1 to 2 cm of surrounding induration (Figure, A).A, Right lateral tongue lesion at presentation. B, Spindled-cell proliferation with palisaded areas (Verocay bodies) and an associated inflammatory infiltrate with abundant eosinophils (hematoxylin-eosin, original magnification ×20).A biopsy was obtained that demonstrated ulceration, pseudoepitheliomatous hyperplasia, and abundant acute and chronic inflammation replete with eosinophils. The differential based on the biopsy included traumatic ulcerative granuloma with stromal eosinophilia and ulceration due to drug- or infection-related sources. In addition, the possibility of an unsampled submucosal mass with overlying ulceration was suggested by the pathologist who interpreted the biopsy findings. Given our clinical concern for an unsampled neoplasm, the patient subsequently underwent resection of the mass; histologic sections of the specimen are demonstrated in Figure, B. Immunohistochemistry studies for SOX-10 revealed positive staining in the proliferating spindle-shaped cells.Schwannoma with a traumatic ulcerative granuloma with stromal eosinophilialike reaction
what is your diagnosis?
What is your diagnosis?
Ulcerated plexiform neurofibroma
Ulcerated granular cell tumor
Schwannoma with a traumatic ulcerative granuloma with stromal eosinophilialike reaction
Human papillomavirus–related squamous cell carcinoma
c
0
0
1
1
female
0
0
1
0-10
null
504
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2734871
A 45-year-old man was admitted to the hospital following 3 weeks of dysphagia; 1 week of nasal regurgitation, nonproductive cough, and breathy voice; and a 25-pound weight loss over the preceding 2 months. He had been diagnosed a year earlier with HIV infection and hepatic tuberculosis. Baseline CD4 count was 7 cells/μL, and following treatment with HAART (highly active antiretroviral therapy) consisting of dolutegravir, emtricitabine, and tenofovir-disoproxil-fumarate, he achieved full virological suppression; however, CD4 counts remained low at 31 cells/μL. He had completed 12 months of directly observed therapy with ethambutol, moxifloxacin, and rifabutin—a liver-sparing regimen—owing to toxic effects from the first-line regimen 1 month prior to admission. Physical examination revealed evidence of perforation of the hard palate (Figure, A). A computed tomographic scan of the chest revealed necrotic lymph nodes, and a palatine biopsy was performed (Figure, B and C).A, Clinical view of the perforation of the hard palate. B, Low-power microscopic view of a biopsy specimen from the hard palate (hematoxylin-eosin). C, Oil immersion image of the same tissue shown panel B stained with Grocott methenamine silver. What Is Your Diagnosis?
Tuberculosis of the palate
Tertiary syphilis
Histoplasmosis
Sarcoidosis
C. Histoplasmosis
C
Histoplasmosis
Histoplasmosis, caused by a soil-based fungus,1 is the most common endemic mycosis in the United States, with a reported in-hospital crude mortality rate of 5% for children and 8% for adults.2 Clinical manifestations of histoplasmosis range from asymptomatic infection to rapidly progressive fatal illness.3 Oral histoplasmosis is an important entity to recognize because it is often a manifestation of disseminated disease.4 Moreover, the appearance may mimic other local or systemically manifested oral disease resulting in misdiagnosis.5Disruption of soil by activities such as spelunking, excavation, outdoor construction, and remodeling of old buildings inhabited by birds or bats1 results in aerosolization of Histoplasma spores, which are then inhaled by human hosts causing infection. The vast majority of these primary infections are either asymptomatic or result in self-limited influenzalike illness, depending on the intensity of exposure.6 Cellular immunity develops approximately 2 weeks after infection in immunocompetent hosts, producing granulomas3 that either resolve spontaneously or persist as calcifications most commonly seen in mediastinal lymph nodes, liver, and spleen.4 In approximately 8% cases of histoplasmosis, the fungus disseminates.1 Risk of dissemination is greatest in individuals with a compromised immune system, especially those with AIDS and hematological malignant conditions, and in individuals using immunosuppressive medications.7 Dissemination can cause an acute, rapidly fatal sepsis syndrome with low blood pressure and multiorgan failure7 or a more subacute to chronically progressive disease with presence of focal lesions in various organs including the oropharynx.1Oral manifestations of histoplasmosis range from nodular, papular lesions to plaques, deep ulcers, and perforation.8 In the oral cavity, the most common locations for these lesions are the tongue, palate, buccal mucosa, gingiva, and pharynx.9 Patients can present with a wide variety of symptoms, including hoarseness, loss of weight, pain at the site of the lesion, and malaise.4 Approximately 30% to 50% of patients with disseminated histoplasmosis have these oral lesions, and sometimes these could be the initial local presentation of disseminated disease.10Oral lesions caused by Histoplasma species mimic several other disease entities such as squamous cell carcinoma, tuberculosis, syphilis, and other fungal infections like cryptococcosis, leading to morbid consequences of misdiagnosis.1 Definitive diagnosis is dependent on tissue biopsy and culture of organisms from lesions. Since culture results may take up to a month, pathological evaluation with fungal stains of biopsy material demonstrating oval to round yeast cells measuring 2 to 4 μm can be helpful in rapid identification of Histoplasma.3 Detection of Histoplasma antigen in body fluids has very high sensitivity in patients with progressive disseminated histoplasmosis and is the mainstay of diagnosis.1 Treatment is primarily dependent on the severity of infection as well as the underlying immune status of the patient. Lipid-based formulations of amphotericin B are used for severe cases, while itraconazole could be an option in less severe cases with intact immune system.1The repeatedly low CD4 cell count of the patient in the present case, even after appropriate HAART treatment, was the biggest risk factor for disseminated histoplasmosis. Despite 2 weeks of aggressive antifungal therapy with amphotericin B, he died. The granulomatous response typically present in immunocompetent patients with histoplasmosis was absent in this patient given profound immunosuppression from CD4 T-cell deficiency. Therefore, performing histochemical stains for microorganisms and ideally fungal culture in biopsied material is critically important in these patients, so as not to miss the etiologic agent of infection.
General
A 45-year-old man was admitted to the hospital following 3 weeks of dysphagia; 1 week of nasal regurgitation, nonproductive cough, and breathy voice; and a 25-pound weight loss over the preceding 2 months. He had been diagnosed a year earlier with HIV infection and hepatic tuberculosis. Baseline CD4 count was 7 cells/μL, and following treatment with HAART (highly active antiretroviral therapy) consisting of dolutegravir, emtricitabine, and tenofovir-disoproxil-fumarate, he achieved full virological suppression; however, CD4 counts remained low at 31 cells/μL. He had completed 12 months of directly observed therapy with ethambutol, moxifloxacin, and rifabutin—a liver-sparing regimen—owing to toxic effects from the first-line regimen 1 month prior to admission. Physical examination revealed evidence of perforation of the hard palate (Figure, A). A computed tomographic scan of the chest revealed necrotic lymph nodes, and a palatine biopsy was performed (Figure, B and C).A, Clinical view of the perforation of the hard palate. B, Low-power microscopic view of a biopsy specimen from the hard palate (hematoxylin-eosin). C, Oil immersion image of the same tissue shown panel B stained with Grocott methenamine silver.
what is your diagnosis?
What is your diagnosis?
Histoplasmosis
Tertiary syphilis
Sarcoidosis
Tuberculosis of the palate
a
0
1
1
1
male
0
0
45
41-50
null
505
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2734819
A man in his late 30s with a history of proliferative diabetic retinopathy presented for evaluation of sudden-onset painless blurry vision of the right eye. His ocular history included nonclearing vitreous hemorrhage in the right eye requiring pars plana vitrectomy, panretinal photocoagulation, multiple treatments with intravitreous bevacizumab, and cataract extraction with placement of an intraocular lens (IOL) into the capsular bag approximately 4 years ago. At his routine follow-up visit 2 weeks prior, the proliferative diabetic retinopathy was quiescent, and his Snellen visual acuity was 20/20 OD with manifest refraction of −1.50 + 0.25 × 180. Examination of the anterior segment at that time was unremarkable.At his current visit, visual acuity with the previous correction had been reduced to 20/150. Repeated manifest refraction of the right eye was −4.00 + 1.50 × 166, resulting in a visual acuity of 20/25. Intraocular pressure of the right eye was 10 mm Hg by Goldmann applanation tonometry. Evaluation of the posterior segment, including dilated fundus examination and optical coherence tomography (OCT) of the macula, showed stability compared with previous results. Imaging of the anterior segment with OCT is shown in Figure 1. The anterior chamber did not appear shallow, and there was no cell or flare.Slitlamp photograph of the anterior segment (A) and optical coherence tomographic imaging of the lens–capsular bag complex (B) obtained at initial presentation (arrowheads show hyperdistention of the capsular bag with turbid fluid).Perform posterior capsulotomy as part of pars plana vitrectomy What Would You Do Next?
Perform posterior capsulotomy as part of pars plana vitrectomy
Perform posterior capsulotomy with Nd:YAG laser
Exchange the IOL
Observe the patient
Capsular bag distention syndrome
B
Perform posterior capsulotomy with Nd:YAG laser
The key to diagnosis is recognizing hyperdistention of the capsular bag with a turbid milieu posterior to the IOL (Figure 1) along with a myopic shift. Anterior segment OCT confirmed the diagnosis. Performing a posterior capsulotomy would allow egress of the turbid material into the vitreous cavity and provide immediate resolution of the refractive change with minimal risk. If this was not successful, we would then perform a posterior capsulotomy during pars plana vitrectomy (choice A). Exchanging the IOL (choice C) is not preferred because decreasing the new lens power to compensate for the myopic shift would result in a hyperopic surprise, as the procedure itself would resolve the capsular bag distention syndrome (CBDS). Observation (choice D) is preferable when the patient does not want intervention; however, the myopic shift needs to be corrected with an updated manifest refraction.Capsular bag distention syndrome, also known as capsular block syndrome, is a rare complication after phacoemulsification and placement of an IOL into the capsular bag.1,2 It is characterized by the accumulation of opaque fluid within the potential space between the posterior aspect of the IOL and the intact posterior capsule. Capsular bag distention syndrome is classified as intraoperative, early-onset, or late-onset on the basis of time of onset.3 Intraoperative CBDS occurs with lens luxation following hydrodissection. Early-onset CBDS is associated with retained viscoelastic materials. Late-onset CBDS is caused by residual cortical cells secreting alpha-crystallin proteins, which creates a turbid milieu that slowly accumulates and distends the posterior capsule.4 Patients with diabetes are not at increased risk for this syndrome. Although intraoperative and early-onset forms of CBDS are often associated with high intraocular pressure and shallowing of the anterior chamber, these features may be absent in the late-onset form.5 This patient presented with late-onset CBDS, demonstrating symptomatic visual acuity decline approximately 4 years after cataract surgery.Although CBDS can be diagnosed clinically, the value of using anterior segment OCT has been previously reported.6 The combination of increased refractive index of the turbid milieu along with convex configuration of the hyperdistended capsular bag forms a plus lens effect that causes a myopic shift, as experienced by this patient.Capsular bag distention syndrome can often be treated with Nd:YAG laser–assisted posterior capsulotomy, as this quick in-office procedure evacuates the turbid milieu from the capsular bag and resolves symptoms almost immediately.5,7 When an infectious etiology, in particular Propionibacterium acnes, is suspected, use of pars plana vitrectomy is necessary to surgically remove the contents of the bag, along with possible IOL exchange and/or injection of intraocular antibiotics.8,9 In this patient, there were no clinical findings of intraocular inflammation or capsular plaques to suggest infection.This patient underwent uneventful Nd:YAG laser–assisted posterior capsulotomy of the right eye; 1 hour after the procedure, the intraocular pressure was 11 mm Hg by applanation. Within 10 minutes after the procedure, the patient reported noticeably improved distance vision in the right eye. Repeated manifest refraction showed resolution of the myopic shift. Clinical examination revealed absence of the hyperdistended capsular bag or turbid fluid, which was confirmed on repeated anterior segment OCT (Figure 2).Anterior segment optical coherence tomography image showing the lens capsule complex after Nd:YAG laser capsulotomy.
Ophthalmology
A man in his late 30s with a history of proliferative diabetic retinopathy presented for evaluation of sudden-onset painless blurry vision of the right eye. His ocular history included nonclearing vitreous hemorrhage in the right eye requiring pars plana vitrectomy, panretinal photocoagulation, multiple treatments with intravitreous bevacizumab, and cataract extraction with placement of an intraocular lens (IOL) into the capsular bag approximately 4 years ago. At his routine follow-up visit 2 weeks prior, the proliferative diabetic retinopathy was quiescent, and his Snellen visual acuity was 20/20 OD with manifest refraction of −1.50 + 0.25 × 180. Examination of the anterior segment at that time was unremarkable.At his current visit, visual acuity with the previous correction had been reduced to 20/150. Repeated manifest refraction of the right eye was −4.00 + 1.50 × 166, resulting in a visual acuity of 20/25. Intraocular pressure of the right eye was 10 mm Hg by Goldmann applanation tonometry. Evaluation of the posterior segment, including dilated fundus examination and optical coherence tomography (OCT) of the macula, showed stability compared with previous results. Imaging of the anterior segment with OCT is shown in Figure 1. The anterior chamber did not appear shallow, and there was no cell or flare.Slitlamp photograph of the anterior segment (A) and optical coherence tomographic imaging of the lens–capsular bag complex (B) obtained at initial presentation (arrowheads show hyperdistention of the capsular bag with turbid fluid).Perform posterior capsulotomy as part of pars plana vitrectomy
what would you do next?
What would you do next?
Perform posterior capsulotomy as part of pars plana vitrectomy
Observe the patient
Exchange the IOL
Perform posterior capsulotomy with Nd:YAG laser
d
0
1
1
1
male
0
0
38
31-40
null
506
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2734988
A woman in her late 60s presented with progressive, painless blurring of vision in both eyes over the past 6 months. Medical history was significant for Sjögren syndrome, for which she was taking hydroxychloroquine sulfate. Ocular history was notable for neovascular age-related macular degeneration under good control with intravitreal injections in the right eye and intermediate dry age-related macular degeneration in the left eye. Family history was negative for corneal disease.On initial presentation to the clinic, the best-corrected visual acuity of the patient was 20/150 OD and 20/40 OS. She was found to have bilateral, gray-white, irregular, poorly circumscribed, linear and nummular central and peripheral corneal opacities, confined primarily to the subepithelium and focally to the underlying anterior stroma (Figure 1A). The epithelium itself and deeper corneal stroma were spared, and the remainder of her anterior segment and fundus examination results were within normal limits. Anterior segment optical coherence tomography documented deposition of hyperreflective material in the subepithelium with trace deposition in the anterior stroma (Figure 1B). Evaluation of the patient's children showed no evidence of corneal disease.A, Right eye gray-white, irregular, linear and nummular central and peripheral corneal opacities. The left eye had similar findings. B, Hyperreflective deposits in the subepithelium (black arrowhead) with trace deposit in the anterior stroma (white arrowhead). What Would You Do Next?
Begin oral acyclovir sodium
Perform penetrating keratoplasty
Initiate topical cyclosporine
Perform superficial corneal biopsy
Paraproteinemic keratopathy from smoldering myeloma
D
Perform superficial corneal biopsy
The patient presented with bilateral subepithelial and anterior stromal corneal opacification. Antiviral agents, such as acyclovir (choice A), would not be preferred because bilaterality of the process and the lack of inflammatory response are not typical of an infectious origin. Topical cyclosporine (choice C) was not recommended because the absence of appreciable epithelial involvement suggests that keratopathy is unlikely related to Sjögren syndrome.1The subacute, noninflammatory, and bilateral nature of corneal opacification suggests corneal deposits that are either dystrophic or nondystrophic. Noninvasive genetic testing on peripheral blood for corneal epithelial-stromal dystrophies may be informative. However, although corneal dystrophies can occasionally manifest at an older age without documented familial inheritance, this type of presentation is uncommon.2 Immunoglobulin corneal deposition is an important diagnostic consideration that should prompt systemic evaluation for paraproteinemia, obviating the need for diagnostic corneal biopsy.3,4In cases in which paraproteinemic keratopathy eludes clinical diagnosis, pathological evaluation of corneal tissue is of diagnostic value.4 Penetrating keratoplasty (choice B), although it provides adequate diagnostic tissue, is an unnecessarily aggressive option for the apparently superficial corneal process. In contrast, superficial corneal biopsy (choice D) can provide valuable pathological information with minimal trauma. Lamellar keratectomy may have the advantage of therapeutic clearing of visual axis while providing the diagnostic tissue. Femto-assisted lamellar keratectomy, a relatively new corneal biopsy technique, may yield excellent optical quality with minimal refractive change, making it a viable option, if available, for patients with anterior corneal dystrophies.5Light microscopy revealed eosinophilic subepithelial and anterior stromal corneal deposits, without appreciable disruption of the Bowman membrane. The deposits did stain with Masson trichrome stain and periodic acid-Schiff stain and did not stain with Congo red stain. Immunohistochemical studies revealed immunoreactivity of deposits for IgG heavy chains and κ light chains (Figure 2). The findings were interpreted as paraproteinemic keratopathy, prompting a systemic workup. Oncologic workup demonstrated IgG κ monoclonal protein in the patient’s serum and urine and 10% κ-restricted plasma cells in the patient’s bone marrow, leading to the diagnosis of smoldering plasma cell myeloma.The immunohistochemical evaluation of superficial keratectomy demonstrates subepithelial and anterior stromal corneal deposits that immunoreact with antibodies for κ light chains (arrowheads). There is no staining for κ light chains in the intact Bowman membrane (asterisk) and in the overlying epithelium (antibody κ; original magnification ×100).Paraproteinemic keratopathy has a diverse clinical presentation, often mimicking crystalline keratopathy, corneal dystrophies, interstitial keratitis, and corneal degenerations.3,4 Systemic paraproteinemia occurs in 98% of patients with immunoglobulin corneal deposits, and it is secondary to the underlying plasma cell myeloma in 43% of patients.3,4 Clinicians should have a high index of suspicion for paraproteinemic keratopathy in older patients with bilateral, noninflammatory, progressive corneal deposits. If diagnosis is suspected, systemic workup for paraproteinemia should be considered.3,4,6In this case, superficial lamellar keratectomy was integral to establishing the diagnosis of paraproteinemic keratopathy and the patient’s underlying smoldering plasma cell myeloma. The patient is currently under the care of an oncologist, with a plan of initiation of systemic chemotherapy. She will be clinically followed up by serum protein electrophoresis and her disease will be considered quiescent once her IgG monoclonal spike is absent. She will be considered for a corneal graft if her symptoms or visual acuity worsen and when her disease is in remission. However, keratopathy may recur despite good systemic control.6
Ophthalmology
A woman in her late 60s presented with progressive, painless blurring of vision in both eyes over the past 6 months. Medical history was significant for Sjögren syndrome, for which she was taking hydroxychloroquine sulfate. Ocular history was notable for neovascular age-related macular degeneration under good control with intravitreal injections in the right eye and intermediate dry age-related macular degeneration in the left eye. Family history was negative for corneal disease.On initial presentation to the clinic, the best-corrected visual acuity of the patient was 20/150 OD and 20/40 OS. She was found to have bilateral, gray-white, irregular, poorly circumscribed, linear and nummular central and peripheral corneal opacities, confined primarily to the subepithelium and focally to the underlying anterior stroma (Figure 1A). The epithelium itself and deeper corneal stroma were spared, and the remainder of her anterior segment and fundus examination results were within normal limits. Anterior segment optical coherence tomography documented deposition of hyperreflective material in the subepithelium with trace deposition in the anterior stroma (Figure 1B). Evaluation of the patient's children showed no evidence of corneal disease.A, Right eye gray-white, irregular, linear and nummular central and peripheral corneal opacities. The left eye had similar findings. B, Hyperreflective deposits in the subepithelium (black arrowhead) with trace deposit in the anterior stroma (white arrowhead).
what would you do next?
What would you do next?
Perform superficial corneal biopsy
Initiate topical cyclosporine
Perform penetrating keratoplasty
Begin oral acyclovir sodium
a
0
0
1
1
female
0
0
68
61-70
White
507
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2735628
A boy in his teens with no significant medical history, ocular history, or recent trauma was referred by an ophthalmologist for mild photophobia and an iris lesion. The referring clinician initially suspected traumatic iridodialysis and vitreous prolapse. On ophthalmic evaluation, visual acuity was 20/20 OS, intraocular pressure was 13 mm Hg OS, and slitlamp examination of the left eye demonstrated a clear, intrinsically vascularized, cystic lesion extending into the anterior chamber from the peripheral superonasal iris (Figure 1A). The base of the lesion was surrounded by iris atrophy and corectopia was noted. The remainder of the ocular examination was unremarkable. Examination of the right eye was also unremarkable. Ultrasonographic biomicroscopy of the anterior segment demonstrated a large, cystic lesion incorporated in the iris of the left eye with a thickness of 2.2 mm and a maximum radial extent of 6.1 mm (Figure 1B).A, Slitlamp photograph of the iris cyst in the superonasal periphery of the left eye. B, Ultrasonographic biomicroscopy of the anterior segment in the left eye with longitudinal orientation at the 10-o’clock position.Given the patient’s excellent visual acuity and no documented evidence of growth, the lesion was initially monitored closely. One month after presentation, the patient developed worsening photophobia, and his visual acuity declined to 20/50 OS. The iris lesion was noted to have grown, with maximum thickness measuring 3.5 mm and with radial extension now involving the visual axis. What Would You Do Next?
Aspiration with absolute alcohol injection
Surgical resection
Continued monitoring
Laser cystotomy
Iris stromal cyst
A
Aspiration with absolute alcohol injection
Pigmented cysts arising from the iris pigment epithelium are the most common type of primary iris cyst.1 They are often of no visual consequence and typically do not require intervention.1 However, iris stromal cysts are likely to enlarge with distortion of surrounding structures and potential consequences of compromised vision, amblyopia, recurrent iridocyclitis, and glaucoma.2,3 Stromal cysts represent approximately 3% of iris tumors, and approximately 40% of iris stromal cysts are congenital.4 Congenital cysts are typically diagnosed in patients younger than 20 years.4In this case, continued observation was not ideal given the extension into the visual axis and the significant growth over the course of 1 month. Aspiration,3 alcohol sclerosis,5,6 resection,7 and laser treatment8 have all been described; however, we believe aspiration with alcohol sclerosis is the preferred initial approach given that it is less invasive than surgical resection and potentially less inflammatory and less likely to yield recurrence than laser cystotomy.Complete resection with sector iridectomy was advocated for congenital iris cysts by Capó and colleagues7 after comparison with injection of trichloroacetic acid and xenon photocoagulation. However, owing to its adverse effect profile, surgical resection is often regarded as a treatment of last resort after failure of more conservative interventions.3 Gupta and colleagues8 reported 2 cases of laser cystotomy using Nd:YAG laser to puncture the cyst wall and allow the cyst’s contents to drain into the anterior chamber. One of the 2 cases demonstrated a recurrence, which was treated successfully with Nd:YAG laser. Allowing cyst material to drain into the anterior chamber may result in inflammatory sequelae, such as peripheral anterior synechiae and epithelial downgrowth from the cyst lining.9Of the treatments mentioned above, aspiration with absolute alcohol injection to allow damage to the epithelial cell lining of the cyst, as initially described by Behrouzi and Khodadoust,6 has substantial support in the literature.5 Shields and colleagues5 described a case series of 16 iris stromal cysts treated with aspiration and absolute alcohol sclerosis. All cysts in their series had recurred after prior simple aspiration. In 15 patients with adequate follow-up, 13 cysts involuted and 1 stabilized in size. Visual acuity was stable or improved in 14 patients. Treatment failed in only 1 patient in the series, later requiring surgical resection of the cyst.5 The most common complications included transient corneal edema and anterior chamber inflammation, which resolved in association with topical corticosteroid therapy.The patient underwent aspiration and absolute alcohol injection with improvement in the size of the cyst (Figure 2).10 Initial treatment was followed by a second aspiration 2 months later with improved visual acuity to 20/20. There were no treatment complications. The cyst remained small with visual acuity 20/20 and normal intraocular pressure in the left eye after 8 months.Slitlamp photograph of the iris cyst 2 weeks after aspiration and absolute alcohol injection.
Ophthalmology
A boy in his teens with no significant medical history, ocular history, or recent trauma was referred by an ophthalmologist for mild photophobia and an iris lesion. The referring clinician initially suspected traumatic iridodialysis and vitreous prolapse. On ophthalmic evaluation, visual acuity was 20/20 OS, intraocular pressure was 13 mm Hg OS, and slitlamp examination of the left eye demonstrated a clear, intrinsically vascularized, cystic lesion extending into the anterior chamber from the peripheral superonasal iris (Figure 1A). The base of the lesion was surrounded by iris atrophy and corectopia was noted. The remainder of the ocular examination was unremarkable. Examination of the right eye was also unremarkable. Ultrasonographic biomicroscopy of the anterior segment demonstrated a large, cystic lesion incorporated in the iris of the left eye with a thickness of 2.2 mm and a maximum radial extent of 6.1 mm (Figure 1B).A, Slitlamp photograph of the iris cyst in the superonasal periphery of the left eye. B, Ultrasonographic biomicroscopy of the anterior segment in the left eye with longitudinal orientation at the 10-o’clock position.Given the patient’s excellent visual acuity and no documented evidence of growth, the lesion was initially monitored closely. One month after presentation, the patient developed worsening photophobia, and his visual acuity declined to 20/50 OS. The iris lesion was noted to have grown, with maximum thickness measuring 3.5 mm and with radial extension now involving the visual axis.
what would you do next?
What would you do next?
Aspiration with absolute alcohol injection
Laser cystotomy
Continued monitoring
Surgical resection
a
0
0
1
1
male
0
0
15
11-20
null
508
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2736416
A 72-year-old woman was referred for evaluation of a choroidal mass and subluxed crystalline lens in the right eye. The patient had a history of congestive heart failure, cleft lip surgery as an infant, and right eye amblyopia. She reported occasional floaters and flashes in the right eye but denied any ocular complaints in the left eye. On examination, her visual acuity measured 3/200 OD and 20/30 OS, stable compared with her baseline. Her intraocular pressure was 18 mm Hg OD and 14 mm Hg OS. Right eye slitlamp biomicroscopy showed an irregular pupil, aphakia, and a pigmented mass behind the temporal iris (Figure 1A). Ophthalmoscopic examination results revealed a subluxed white cataractous lens in the inferior vitreous base (Figure 1B). An evaluation of the left eye yielded normal results aside for a moderate cataract. B-scan ultrasonography results showed the 2 masses described previously; both were moderately reflective, oval, and separate from the wall of the eye. Ultrasonography biomicroscopy (UBM) confirmed that the temporal mass and the inferiorly subluxed lens were separate from the iris and ciliary body and were similar to each other in size, shape, and density.A, Anterior fundus photograph of the right eye shows a dark mass behind the pupil temporally. B, Fundoscopy of the same eye shows a white cataractous lens subluxed into the inferior vitreous base.Pars plana lensectomy to remove the subluxed lensMagnetic resonance imaging of the brain and orbits What Would You Do Next?
Plaque radiotherapy
Pars plana lensectomy to remove the subluxed lens
Magnetic resonance imaging of the brain and orbits
Biopsy of the choroidal mass
Congenital biphakia with double subluxed crystalline lenses
C
Magnetic resonance imaging of the brain and orbits
This is a case of a patient who was referred for evaluation of a likely choroidal mass. However, B-scan ultrasonography and UBM results show that the mass was adjacent to, but separate from, the ciliary body temporally, and the presence of a subluxed lens in the inferior vitreous further complicates the diagnosis.Because of the uncertainty of the etiology of the mass as to whether it is benign or malignant or even from which tissue it arose, treatment with plaque radiotherapy would not be appropriate. With the patient’s ocular history of amblyopia and poor visual prognosis, there was no visual benefit to removing the subluxed lens. Further imaging of the right eye with magnetic resonance imaging (MRI) was obtained and reaffirmed that the mass was similar in shape and density to the inferior subluxed lens (Figure 2). The MRI, along with autofluorescence, B-scan ultrasonography, and UBM, led us to diagnose congenital biphakia with 2 subluxed lenses1 and we elected observation as opposed to a biopsy.T2-weighted axial magnetic resonance imaging results of the brain and orbits with fat-saturation confirm 2 crystalline lenses (biphakia) that are subluxed in the right eye (yellow arrowheads) and a crystalline lens in normal position in the left eye (blue arrowhead).Congenital biphakia (2 crystalline lenses in the same eye) was first published in a case report by Von Graefe in 18542 and is an extremely rare condition, having only been reported in a few cases since. These cases discuss the presence of biphakia with and without other ocular abnormalities,3 such as hourglass cornea,1 uveal coloboma,4 iris coloboma,2 cleft anterior segment,5 and diplophthalmos,6 as well as its appearance alongside congenital nasal and craniocervical deformities.4,6 Notably, in our review of the literature on congenital biphakia, all cases presented unilaterally rather than bilaterally. In the aforementioned report of biphakia with congenital nasal abnormality, the patient also had a bifid eyelid and developmental delay4; by contrast, this patient’s developmental abnormalities were limited to right eye biphakia and an orofacial cleft.Although, to our knowledge, there is no known link, we question whether this patient’s biphakia and orofacial cleft may be associated because both are congenital physical anomalies that occur during early gestation. It is believed that a sporadic event, such as the duplication of the lens placode during fetal development, can result in congenital biphakia.5Unlike previously reported cases of congenital biphakia, this patient presented with double subluxed crystalline lenses, 1 brunescent and 1 white in color. The brunescent lens was confused for a choroidal neoplasm because of its dark color and location adjacent to the ciliary body. The differential diagnosis of a mass behind the iris includes uveal tumor or metastasis, iris or ciliary body cyst, inflammatory nodule, and subluxed lens. We initially dismissed the possibility of subluxed lens for this patient because of the presence of an additional, more obvious subluxed lens until further diagnostic imaging results confirmed the similarities between the mass and the subluxed lens. Although rare, this case is a good reminder that congenital defects can masquerade as other ocular abnormalities, including choroidal tumor.Comparison photographs and ultrasonography over 8 years demonstrated minimal migration of the subluxed lenses and no change in size or shape for this patient.
Ophthalmology
A 72-year-old woman was referred for evaluation of a choroidal mass and subluxed crystalline lens in the right eye. The patient had a history of congestive heart failure, cleft lip surgery as an infant, and right eye amblyopia. She reported occasional floaters and flashes in the right eye but denied any ocular complaints in the left eye. On examination, her visual acuity measured 3/200 OD and 20/30 OS, stable compared with her baseline. Her intraocular pressure was 18 mm Hg OD and 14 mm Hg OS. Right eye slitlamp biomicroscopy showed an irregular pupil, aphakia, and a pigmented mass behind the temporal iris (Figure 1A). Ophthalmoscopic examination results revealed a subluxed white cataractous lens in the inferior vitreous base (Figure 1B). An evaluation of the left eye yielded normal results aside for a moderate cataract. B-scan ultrasonography results showed the 2 masses described previously; both were moderately reflective, oval, and separate from the wall of the eye. Ultrasonography biomicroscopy (UBM) confirmed that the temporal mass and the inferiorly subluxed lens were separate from the iris and ciliary body and were similar to each other in size, shape, and density.A, Anterior fundus photograph of the right eye shows a dark mass behind the pupil temporally. B, Fundoscopy of the same eye shows a white cataractous lens subluxed into the inferior vitreous base.Pars plana lensectomy to remove the subluxed lensMagnetic resonance imaging of the brain and orbits
what would you do next?
What would you do next?
Plaque radiotherapy
Magnetic resonance imaging of the brain and orbits
Biopsy of the choroidal mass
Pars plana lensectomy to remove the subluxed lens
b
0
0
1
1
female
0
0
72
71-80
White
509
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2737084
A woman in her 30s presented with frequent falls, right-sided lower-extremity weakness, and intermittent confusion. She had a medical history significant for a renal transplant performed 4 years prior. She received induction with alemtuzumab and maintenance immunosuppression with tacrolimus and mycophenolate. She was not on any antiviral therapies prior to presentation. On admission, the patient developed left eye pain and slurred speech. Physical examination revealed expressive aphasia, partial left abducens nerve palsy, decreased motor strength in her right upper and lower extremities, and right-sided pronator drift. Laboratory evaluation showed a white blood cell count of 9.6 K/μL (reference range, 4.5-11.0 K/μL), and a lactic acid dehydrogenase level of 547 U/L (reference range, 160-450 U/L). Cerebrospinal fluid analysis revealed a white blood cell count of 51 cells/μL (reference range, 0-5 cells/μL) with 92% lymphocytes, a glucose level of 76 mg/dL (reference range, 40-70 mg/dL), and a protein level of 136 mg/dL (reference range, 12-60 mg/dL). A computed tomography (CT) scan of the brain showed multifocal vasogenic edema around the left lentiform nuclei, left temporal occipital white matter, and right parietal white matter, with mass effect causing near complete effacement of the third ventricle and subsequent rightward midline shift. Magnetic resonance imaging of the brain revealed multiple ring-enhancing lesions (Figure, A). A fluorine 18 fluorodeoxyglucose positron emission tomography/CT demonstrated increased focal fluorodeoxyglucose activity throughout the brain only. The patient underwent a biopsy of the right posterior temporal lobe (Figure, B and C).A, Ring-enhancing lesions are visible. B, A lesional biopsy specimen of the brain was obtained (hematoxylin-eosin, original magnification ×400). C, Immunohistochemical stain of CD20 (original magnification ×400). What Is Your Diagnosis?
Toxoplasmosis
Primary central nervous system posttransplant lymphoproliferative disorder
Glioblastoma
Metastatic disease from unknown primary neoplasm
B. Primary central nervous system posttransplant lymphoproliferative disorder
B
Primary central nervous system posttransplant lymphoproliferative disorder
Histologic results of the biopsy revealed abundant perivascular infiltrates of small lymphocytes with admixed large cells and occasional plasma cells (Figure, B). Immunohistochemical staining showed large cells positive for CD20 (Figure, C), BCL6, and CD30, and negative for CD10, consistent with a diagnosis of polymorphic primary central nervous system posttransplant lymphoproliferative disorder (PCNS-PTLD).Initially described in 1968,1 PTLD is a potentially fatal complication of both solid organ and hematopoietic stem cell transplant. The incidence of PTLD varies based on type of transplanted organ, immunosuppressive regimen, and patient characteristics. Posttransplant lymphoproliferative disorder manifesting after solid organ transplant (SOT) occurs in approximately 2% to 3% of cases, with highest incidence in intestinal and multiple organ transplant recipients (5%-20%), followed by lung and heart transplant (2%-10%), and then pancreatic, renal, and liver transplant (1%-5%).2,3The World Health Organization divides PTLD into 3 pathologic categories: early lesions, polymorphic, and monomorphic, with monomorphic PTLD fulfilling the criteria for one of the B-cell or natural killer/T-cell neoplasms recognized in immunocompetent individuals.4 About 50% of PTLD cases are driven by Epstein-Barr virus (EBV) infection in the setting of immunosuppression and subsequent T-cell inhibition.5 This patient’s pathology was positive for EBV, confirmed by in situ hybridization for EBV-encoded RNA; however, circulating EBV DNA was not detected in the serum.Primary central nervous system posttransplant lymphoproliferative disorder comprises 5% to 15% of all PTLD cases.6 Neuroradiographic findings of PCNS-PTLD are nonspecific and tend to have overlapping features with CNS infections and primary CNS lymphoma in immunocompromised individuals. Several case series7,8 report multicentric, contrast-enhancing, intra-axial lesions with extensive edema and marked expansion of the perivascular spaces. These lesions tend to be deeper in supratentorial structures and periventricular regions, and occur less often in the cerebellum and brainstem.6 Manifestations in the spinal cord and leptomeninges are rare.7,8 Given that isolated CNS involvement of PTLD is uncommon, clinical evaluation should include complete staging with positron emission tomography/CT to exclude systemic involvement. Diagnosis of PCNS-PTLD is established by immunohistopathologic analysis of a tissue biopsy specimen.Primary central nervous system posttransplant lymphoproliferative disorder appears to embody a distinct clinicopathologic entity within the PTLD spectrum. Although the highest incidence of PTLD occurs within the first year after transplant, the median time from solid organ transplant to PCNS-PTLD diagnosis is 3 to 5 years.7-9 Compared with non-CNS PTLD, PCNS-PTLD is more frequently associated with higher rates of EBV infection, prior renal transplant, and monomorphic as opposed to polymorphic subtype.8,9Given the rarity of PCNS-PTLD, no standardized treatment has been established. Treatment options for PCNS-PTLD are dictated by the underlying morphologic subtype and include reduction of immunosuppression for all lesions, with inclusion of rituximab therapy for polymorphic lesions.8,9 Chemotherapy should be used to treat the functionally able patient with monomorphic PCNS-PTLD. Agents with proven efficacy in the treatment of PCNS lymphoma, including high-dose methotrexate, are often used to treat PCNS-PTLD.10 Whole-brain radiotherapy remains an option for relapsed or refractory disease.Prognosis remains suboptimal for PCNS-PTLD, with a multicenter study of 84 patients demonstrating a 3-year progression-free survival rate of 32% and an overall survival rate of 43%.9 Response to first-line therapy, rather than choice of treatment, was the most important predictor of progression-free survival and Overall survival among patients with PCNS-PTLD.9The patient was diagnosed with polymorphic, EBV-positive PCNS-PTLD 4 years after renal transplant. After diagnosis, mycophenolate was discontinued and the tacrolimus trough goal was reduced. The patient also received 4 weekly doses of rituximab. She subsequently had improvement in neurologic function. This case highlights the importance of maintaining clinical suspicion for delayed and rare presentations of PTLD in transplant recipients. Multicenter analyses of treatment and outcome data are warranted given the uncertainties regarding optimal initial therapy in this patient population.
Oncology
A woman in her 30s presented with frequent falls, right-sided lower-extremity weakness, and intermittent confusion. She had a medical history significant for a renal transplant performed 4 years prior. She received induction with alemtuzumab and maintenance immunosuppression with tacrolimus and mycophenolate. She was not on any antiviral therapies prior to presentation. On admission, the patient developed left eye pain and slurred speech. Physical examination revealed expressive aphasia, partial left abducens nerve palsy, decreased motor strength in her right upper and lower extremities, and right-sided pronator drift. Laboratory evaluation showed a white blood cell count of 9.6 K/μL (reference range, 4.5-11.0 K/μL), and a lactic acid dehydrogenase level of 547 U/L (reference range, 160-450 U/L). Cerebrospinal fluid analysis revealed a white blood cell count of 51 cells/μL (reference range, 0-5 cells/μL) with 92% lymphocytes, a glucose level of 76 mg/dL (reference range, 40-70 mg/dL), and a protein level of 136 mg/dL (reference range, 12-60 mg/dL). A computed tomography (CT) scan of the brain showed multifocal vasogenic edema around the left lentiform nuclei, left temporal occipital white matter, and right parietal white matter, with mass effect causing near complete effacement of the third ventricle and subsequent rightward midline shift. Magnetic resonance imaging of the brain revealed multiple ring-enhancing lesions (Figure, A). A fluorine 18 fluorodeoxyglucose positron emission tomography/CT demonstrated increased focal fluorodeoxyglucose activity throughout the brain only. The patient underwent a biopsy of the right posterior temporal lobe (Figure, B and C).A, Ring-enhancing lesions are visible. B, A lesional biopsy specimen of the brain was obtained (hematoxylin-eosin, original magnification ×400). C, Immunohistochemical stain of CD20 (original magnification ×400).
what is your diagnosis?
What is your diagnosis?
Metastatic disease from unknown primary neoplasm
Glioblastoma
Primary central nervous system posttransplant lymphoproliferative disorder
Toxoplasmosis
c
1
1
1
1
female
0
0
35
31-40
White
510
original
https://jamanetwork.com/journals/jama/fullarticle/2737057
A 28-year-old man presented to the emergency department with diffuse oral lesions, fever, and nonproductive cough for 1 week. He was diagnosed with herpes simplex virus (HSV) and discharged home with acyclovir. Two days later, he returned with worsening oral lesions, painful phonation, poor oral intake, odynophagia, and new painful penile lesions. He denied any abdominal pain, diarrhea, rectal bleeding, weight loss, arthralgia, fatigue, vision changes, or skin rashes. He reported a similar episode of widespread oral lesions 18 months earlier that was less severe and self-resolving. He had no other medical conditions, took no medications, and was sexually monogamous with his wife, who was asymptomatic.The patient was hemodynamically stable and had a normal respiratory examination. He had a fever (39.4°C) on presentation, but his temperature abated overnight and he remained afebrile for the rest of the admission. His superior and inferior lips had extensive well-circumscribed erosions and ulcerations with erythematous edges, crusting, and a gray-white base (Figure, left). Lesions extended into the buccal mucosa and soft palate. His tongue was enlarged with fissures. A genital examination showed similar lesions on the penis, with scalloped borders (Figure, right). There were no skin lesions, lymphadenopathy, or hepatosplenomegaly.Left, Extensive well-circumscribed oral lesions limited to the mucosa in a 28-year-old man. Right, Similar lesions with scalloped borders on the distal shaft and glans of the penis in the same patient.Results of laboratory tests from the emergency department were normal, and a chest radiograph showed no evidence of pneumonia. Results of further testing for antinuclear antibodies, erythrocyte sedimentation rate, C-reactive protein level, HIV, HSV serology, syphilis, chlamydia, and gonorrhea were negative. Bacterial and viral cultures of the oral and penile lesions were also negative. Serum enzyme immunoassay for Mycoplasma pneumoniae IgM was positive at 1280 U/mL. The patient did not consent to a punch biopsy of the oral lesions. What Would You Do Next?
Perform upper endoscopy and colonoscopy
Test for anti–double-stranded DNA antibodies
Prescribe oral azithromycin
Prescribe oral suspension nystatin
M pneumoniae–induced rash and mucositis (MIRM)
C
Prescribe oral azithromycin
The key to the correct diagnosis is the elevated M pneumoniae IgM titers in a patient with a nonproductive cough and 2 sites of mucosal lesions. Although Crohn disease and systemic lupus erythematosus can present with oral lesions, the patient’s lack of associated gastrointestinal or musculoskeletal symptoms make these diagnoses unlikely. Since the patient is HIV-negative and immunocompetent, oral candidiasis is unlikely.M pneumoniae infections are often asymptomatic but can also present as upper respiratory tract infections, bronchitis, or pneumonia. Less commonly, patients with M pneumoniae infection can have mucocutaneous manifestations. MIRM is an extremely rare complication of M pneumoniae infection (exact incidence unknown) and often is mistaken for Stevens-Johnson syndrome or HSV-associated erythema multiforme. Typically, MIRM occurs in older children or adolescents and involves the oral, ocular, and genitourinary mucosal surfaces in 94%, 82%, and 63% of cases, respectively. Skin involvement is typically sparse and occurs in an acral distribution.1In 2015, Caravan et al proposed a diagnostic criterion for MIRM that included 10% or less body surface area detachment, 2 or more mucosal sites involved, few vesiculobullous skin lesions or atypical targets, and clinical or laboratory evidence of atypical pneumonia.1 The gold standard for diagnosis is M pneumoniae nucleic acid amplification testing (NAAT) because of its superior sensitivity and specificity in an acute infection.2 However, NAAT is costly and often time-inefficient to run (eg, if it needs to be sent to an outside laboratory).3 Serum enzyme immunoassay IgM testing is an alternative that is more widely available, faster, and less costly2,3; however, recent studies have demonstrated both significant heterogeneity in sensitivity and specificity and inconsistent diagnostic accuracy.3 This is in part because IgM antibodies begin to appear 7 to 9 days after initial infection, with a peak at 3 to 6 weeks, and therefore may be missed when testing during acute infection or reinfection in adults.3-6 A systematic review concluded that during the acute phase of infection, the use of IgM in combination with polymerase chain reaction allows for a more precise and reliable M pneumoniae diagnosis.3There are no evidence-based guidelines for the treatment of MIRM. In a systematic review of 202 cases of MIRM, 80% of patients received antibiotics, 35% received systemic corticosteroids, and 8% received intravenous immunoglobulin.1M pneumoniae is very susceptible to antibiotics that interfere with protein and DNA synthesis, which includes macrolides, tetracyclines, and quinolones.7,8 Nevertheless, the full clinical benefits of antimicrobial therapy are unclear, and further studies are required. The efficacy and clinical benefit of systemic corticosteroid and intravenous immunoglobulin therapy in the treatment of MIRM are not definitive.1Based on the same systematic review, MIRM had a good prognosis in 81% of patients; however, 4% required admission to a medical intensive care unit.1 Mortality was 3%; however, these instances were all from the 1940s (preantibiotic era). Pulmonary complications and lack of treatment with antibiotics were associated with poorer outcomes.1 Other complications included permanent ocular, oral, genital, or cutaneous lesions or B-cell lymphopenia.1,9NAAT was not performed in this instance because it was unavailable in-house; the diagnosis of MIRM was based on physical examination and positive IgM titers. The patient was treated with 5 days of oral azithromycin and had full resolution of cough and mucosal symptoms 1 week later, which supported the diagnosis. Follow-up IgM convalescent titers completed 7 months after treatment had a more than 4-fold decrease from the acute titers, further supporting the diagnosis.4
General
A 28-year-old man presented to the emergency department with diffuse oral lesions, fever, and nonproductive cough for 1 week. He was diagnosed with herpes simplex virus (HSV) and discharged home with acyclovir. Two days later, he returned with worsening oral lesions, painful phonation, poor oral intake, odynophagia, and new painful penile lesions. He denied any abdominal pain, diarrhea, rectal bleeding, weight loss, arthralgia, fatigue, vision changes, or skin rashes. He reported a similar episode of widespread oral lesions 18 months earlier that was less severe and self-resolving. He had no other medical conditions, took no medications, and was sexually monogamous with his wife, who was asymptomatic.The patient was hemodynamically stable and had a normal respiratory examination. He had a fever (39.4°C) on presentation, but his temperature abated overnight and he remained afebrile for the rest of the admission. His superior and inferior lips had extensive well-circumscribed erosions and ulcerations with erythematous edges, crusting, and a gray-white base (Figure, left). Lesions extended into the buccal mucosa and soft palate. His tongue was enlarged with fissures. A genital examination showed similar lesions on the penis, with scalloped borders (Figure, right). There were no skin lesions, lymphadenopathy, or hepatosplenomegaly.Left, Extensive well-circumscribed oral lesions limited to the mucosa in a 28-year-old man. Right, Similar lesions with scalloped borders on the distal shaft and glans of the penis in the same patient.Results of laboratory tests from the emergency department were normal, and a chest radiograph showed no evidence of pneumonia. Results of further testing for antinuclear antibodies, erythrocyte sedimentation rate, C-reactive protein level, HIV, HSV serology, syphilis, chlamydia, and gonorrhea were negative. Bacterial and viral cultures of the oral and penile lesions were also negative. Serum enzyme immunoassay for Mycoplasma pneumoniae IgM was positive at 1280 U/mL. The patient did not consent to a punch biopsy of the oral lesions.
what would you do next?
What would you do next?
Prescribe oral azithromycin
Perform upper endoscopy and colonoscopy
Prescribe oral suspension nystatin
Test for anti–double-stranded DNA antibodies
a
1
1
1
1
male
0
0
28
21-30
White
511
original
https://jamanetwork.com/journals/jama/fullarticle/2736971
A 39-year-old woman presented to the emergency department after she noticed that her left pupil was dilated when looking in the mirror earlier in the day. She had a medical history of hypertension and palmar and axillary hyperhidrosis. She denied double vision, ptosis, headache, and weakness. On examination, she appeared well and her blood pressure was 128/78 mm Hg; heart rate, 76/min; and respiratory rate, 13/min. Visual acuity was 20/20 in both eyes, there was no ptosis, and extraocular movements were full. Pupillary examination revealed anisocoria that was more pronounced in bright lighting conditions (Figure). The right pupil measured 6 mm in the dark and 3 mm in the light, whereas the left pupil measured 9 mm in the dark and 8 mm in the light. The left pupil was poorly reactive to light and to a near target. Slitlamp examination revealed normal-appearing anterior chambers without any signs of intraocular inflammation. Cranial nerve function was otherwise normal. The remainder of the neurologic examination revealed normal muscle tone, strength, reflexes, and gait.Patient’s pupil sizes shown in dim and bright lighting conditions.Order computed tomography angiography of the head to rule out aneurysmal compression of the left third cranial nerveOrder magnetic resonance imaging of the brain to rule out a brainstem strokePerform detailed review of systemic and topical medicationsPerform pharmacologic testing with apraclonidine to confirm Horner syndrome What Would You Do Next?
Order computed tomography angiography of the head to rule out aneurysmal compression of the left third cranial nerve
Order magnetic resonance imaging of the brain to rule out a brainstem stroke
Perform detailed review of systemic and topical medications
Perform pharmacologic testing with apraclonidine to confirm Horner syndrome
Pharmacologic mydriasis from topical glycopyrronium bromide (glycopyrrolate) for hyperhidrosis
C
Perform detailed review of systemic and topical medications
The key to the correct diagnosis in this case is the presence of an isolated dilated pupil in a well-appearing patient with an otherwise normal neurologic examination. In an alert and oriented patient with a palsy of the third cranial nerve, there is almost always associated ptosis or limitation of extraocular eye movements in addition to pupillary symptoms.1 Since these symptoms were not present in this case, further investigations for a third-nerve palsy, such as computed tomography angiography or magnetic resonance imaging, were not required. Horner syndrome is a rare condition that occurs when there is disruption of the sympathetic pathway and results in ipsilateral ptosis and miosis. A carotid dissection should be ruled out in acute cases, especially when there is associated neck pain or headache. Since there was no ptosis and the anisocoria was worse in the light, not the dark, Horner syndrome was not considered in the differential diagnosis. Given this information, a detailed review of the patient’s medications was performed and it was discovered that she was using glycopyrronium bromide (glycopyrrolate), an anticholinergic medication.Anisocoria refers to unequal pupil sizes and may be the sign of a serious underlying neurologic condition. It requires careful attention, particularly when there are associated neurologic symptoms such as ptosis or diplopia, which may indicate dysfunction of the third cranial nerve, which is vulnerable to compression from aneurysmal and other lesions.When evaluating a patient with anisocoria, it is important to first establish which pupil is abnormal—this can be achieved by assessing the anisocoria in dim and bright lighting conditions.2 If the anisocoria is greater in dim lighting conditions, the abnormal pupil is the smaller pupil since it is unable to fully dilate, implying a problem with the sympathetic pathway, which innervates the iris dilator muscle. If the anisocoria is greater in bright lighting conditions, as in this case, the abnormal pupil is the larger pupil. Another indicator that the left pupil was abnormal was the poor reaction to light. The differential diagnosis for a large pupil that is poorly reactive to light mainly includes a palsy of the third cranial nerve, pharmacologic mydriasis, and a tonic pupil.3 A tonic pupil denotes a pupil with parasympathetic denervation and results in a pupil that is poorly reactive to light but strongly reactive to a near target. This may be seen in the context of infection (eg, herpes zoster), orbital surgery, or systemic autonomic neuropathies but may also be idiopathic (so-called Adie tonic pupil).3A detailed review of the patient’s medications revealed that she was using topical glycopyrronium bromide (glycopyrrolate) for palmar and axillary hyperhidrosis. Glycopyrronium bromide is a synthetically derived anticholinergic medication with primarily antimuscarinic effects.4 It decreases sweat production and has been found to result in a reduction in perspiration after oral or topical administration.5 It has also been used as a preoperative medication to inhibit salivary gland and respiratory secretions.4 Other commonly used anticholinergic medications include scopolamine and atropine. Parasympathetic cholinergic input to the iris results in activation of the smooth muscle cells in the iris sphincter muscle and constriction of the pupil.6 Inhibiting this pathway via topical or oral medications will therefore result in mydriasis. This patient likely inadvertently touched her left eye or periocular tissues, resulting in absorption of the medication and dilation of her left pupil. This emphasizes the importance of asking patients in this setting if they have put any eye drops in their eyes or someone else’s eyes, or touched any topical medications or unfamiliar plants. To confirm the diagnosis of pharmacologic mydriasis, pilocarpine eye drops (1%), a muscarinic agonist, can be administered to both eyes. This will constrict the normal pupil but not the pharmacologically dilated pupil.The patient was informed that the dilated pupil was likely a result of the glycopyrronium bromide used for her hyperhidrosis. She temporarily discontinued the medication and her pupil returned to normal size after 5 days, confirming the diagnosis. At a follow-up appointment 2 weeks later, her pupils were both normal-sized and symmetric, and she continued to have no ptosis or extraocular dysmotility.
General
A 39-year-old woman presented to the emergency department after she noticed that her left pupil was dilated when looking in the mirror earlier in the day. She had a medical history of hypertension and palmar and axillary hyperhidrosis. She denied double vision, ptosis, headache, and weakness. On examination, she appeared well and her blood pressure was 128/78 mm Hg; heart rate, 76/min; and respiratory rate, 13/min. Visual acuity was 20/20 in both eyes, there was no ptosis, and extraocular movements were full. Pupillary examination revealed anisocoria that was more pronounced in bright lighting conditions (Figure). The right pupil measured 6 mm in the dark and 3 mm in the light, whereas the left pupil measured 9 mm in the dark and 8 mm in the light. The left pupil was poorly reactive to light and to a near target. Slitlamp examination revealed normal-appearing anterior chambers without any signs of intraocular inflammation. Cranial nerve function was otherwise normal. The remainder of the neurologic examination revealed normal muscle tone, strength, reflexes, and gait.Patient’s pupil sizes shown in dim and bright lighting conditions.Order computed tomography angiography of the head to rule out aneurysmal compression of the left third cranial nerveOrder magnetic resonance imaging of the brain to rule out a brainstem strokePerform detailed review of systemic and topical medicationsPerform pharmacologic testing with apraclonidine to confirm Horner syndrome
what would you do next?
What would you do next?
Perform pharmacologic testing with apraclonidine to confirm Horner syndrome
Order magnetic resonance imaging of the brain to rule out a brainstem stroke
Order computed tomography angiography of the head to rule out aneurysmal compression of the left third cranial nerve
Perform detailed review of systemic and topical medications
d
1
0
1
1
female
0
0
39
31-40
null
512
original
https://jamanetwork.com/journals/jama/fullarticle/2736434
A 65-year-old Vietnamese man with hypertension, type 2 diabetes mellitus, and chronic hepatitis B with cirrhosis presented with a 2-week history of shortness of breath at rest, orthopnea, and lower extremity edema. He reported a 4-month history of nonproductive cough, 5-kg weight loss, and fatigue. He immigrated to the United States as an adult more than 20 years before presentation. His temperature was 37°C, heart rate was 78/min, respiratory rate was 17/min, and blood pressure was 158/95 mm Hg. A chest radiographic image suggested cardiomegaly and a computed tomographic scan demonstrated a moderate to large pericardial effusion. A pericardial drain was placed and pericardial fluid was sent to the laboratory for evaluation. Initial pericardial fluid study results are presented in the Table. Empirical treatment for tuberculosis was initiated. Three days later, an adenosine deaminase (ADA) level of 118.1 U/L (normal range, 0.0-11.3 U/L) from pericardial fluid was reported from the laboratory.Perform additional pericardial effusion cytology and culture testingStop further nontubercular diagnostic testing and continue antitubercular therapyInitiate oral colchicine, nonsteroidal anti-inflammatory drugs, and steroids for systemic inflammation What Would You Do Next?
Perform additional pericardial effusion cytology and culture testing
Stop further nontubercular diagnostic testing and continue antitubercular therapy
Initiate oral colchicine, nonsteroidal anti-inflammatory drugs, and steroids for systemic inflammation
Perform an interferon-γ release assay (IGRA)
null
B
Stop further nontubercular diagnostic testing and continue antitubercular therapy
ADA is an enzyme in lymphocytes and myeloid cells that recycles toxic purine pathway metabolites, which are essential for DNA metabolism and cell viability.1,2 ADA levels are elevated in inflammatory effusions, including pleural, pericardial, and joint effusion, caused by bacterial infections, granulomatous inflammation (eg, tuberculosis, sarcoidosis), malignancy, and autoimmune diseases (eg, lupus, vasculitis).1,2 ADA is normally elevated in neutrophil-predominant effusions and is not a useful diagnostic test in the setting of neutrophil-predominant effusions.3 However, among lymphocyte-predominant effusions, levels of ADA are typically higher in those caused by tuberculosis (TB) than those caused by other conditions.1,2In lymphocyte-predominant effusions, the most common threshold for an ADA test result indicating TB is an ADA level greater than 40 U/L. An ADA level greater than 40 U/L has a sensitivity of 87% to 93% and specificity of 89% to 97% for TB.2,4 At a threshold of greater than 35 U/L, sensitivity is higher (93%-95%) but specificity is lower (74%-90%).3,5,6 Given a pretest probability of 70% for TB in a lymphocyte-predominant pericardial effusion in a patient from an endemic country,7 an ADA level greater than 40 U/L results in a posttest probability of 96%, while a level less than or equal to 40 U/L results in a posttest probability of 19%. The Centers for Medicare & Medicaid Services reimbursement rate for ADA testing is $8.10 (Current Procedural Terminology code 84311).In the current patient, the ADA test was ordered from pericardial fluid to confirm the presumptive diagnosis of pericardial TB, for which antitubercular therapy was initiated. RIPE (rifampin, isoniazid, pyrazinamide, ethambutol) therapy was started because of the high pretest probability of TB in a patient from a country with endemic TB with chronic cough, weight loss, and lymphocyte-predominant pericardial effusion. The patient’s ADA level of 118.1 U/L confirmed the diagnosis, enabling cessation of further evaluation and supporting continuation of antitubercular therapy.An alternative approach is to await a culture positive for TB before starting therapy. However, cultures positive for TB can take weeks to grow, which can allow infection to progress while awaiting confirmation. An ADA level greater than 40 U/L in a patient with a high pretest probability of disease results in a sufficiently high posttest probability to justify a full course of TB therapy. Furthermore, even with an ADA level less than or equal to 40 U/L in a patient with a high pretest probability, the posttest probability of approximately 20% is sufficiently high to warrant empirical therapy. However, the low ADA level would necessitate further diagnostic testing, including invasive pericardial biopsy and serologies, to rule out other causes (eg, sarcoid, malignancy, vasculitis) while treating the patient for TB. In that situation, empirical TB therapy could have been stopped if an alternative diagnosis was established.Elevated interferon-γ concentrations in effusions have high sensitivity (95.7%) and specificity (96.3%) for active TB, but their use is limited by cost and availability.1,4 Interferon-γ can also be measured from whole blood via IGRAs. However, IGRAs and the similar purified protein derivative skin test are only useful to diagnose latent TB. Laboratory test results indicating ADA values greater than 40 U/L cannot distinguish between latent and active TB (nonspecific), and IGRAs/purified protein derivatives are associated with false-negative results in up to 30% of patients with pulmonary TB and 50% of patients with disseminated TB,8,9 because active TB causes antigen-specific anergy of T cells.The patient’s sputum was negative for acid-fast bacilli smears and TB polymerase chain reaction. Pericardial fluid cytology demonstrated inflammatory cells, but was negative for malignant cells, acid-fast bacilli stain, and TB polymerase chain reaction. However, the pericardial fluid cultures grew TB at 3 weeks and the sputa cultures grew TB at 5 weeks. Seven months after the start of treatment, the patient was hospitalized and died of complications of pneumonia and septic shock unrelated to TB.ADA levels are typically elevated, and not diagnostically helpful, in neutrophil-predominant effusions.In lymphocyte-predominant effusions, ADA levels are elevated in effusions caused by tuberculosis, but typically not in effusions due to other diseases.In a patient with a low pretest probability of TB, an ADA level less than or equal to 40 U/L in a lymphocyte-predominant effusion essentially rules out TB.In a patient with a high pretest probability of pericardial TB, an ADA level greater than 40 U/L in a lymphocyte-predominant effusion is highly suggestive of TB, precluding the need for further diagnostic testing.
Diagnostic
A 65-year-old Vietnamese man with hypertension, type 2 diabetes mellitus, and chronic hepatitis B with cirrhosis presented with a 2-week history of shortness of breath at rest, orthopnea, and lower extremity edema. He reported a 4-month history of nonproductive cough, 5-kg weight loss, and fatigue. He immigrated to the United States as an adult more than 20 years before presentation. His temperature was 37°C, heart rate was 78/min, respiratory rate was 17/min, and blood pressure was 158/95 mm Hg. A chest radiographic image suggested cardiomegaly and a computed tomographic scan demonstrated a moderate to large pericardial effusion. A pericardial drain was placed and pericardial fluid was sent to the laboratory for evaluation. Initial pericardial fluid study results are presented in the Table. Empirical treatment for tuberculosis was initiated. Three days later, an adenosine deaminase (ADA) level of 118.1 U/L (normal range, 0.0-11.3 U/L) from pericardial fluid was reported from the laboratory.Perform additional pericardial effusion cytology and culture testingStop further nontubercular diagnostic testing and continue antitubercular therapyInitiate oral colchicine, nonsteroidal anti-inflammatory drugs, and steroids for systemic inflammation
what would you do next?
What would you do next?
Perform additional pericardial effusion cytology and culture testing
Stop further nontubercular diagnostic testing and continue antitubercular therapy
Perform an interferon-γ release assay (IGRA)
Initiate oral colchicine, nonsteroidal anti-inflammatory drugs, and steroids for systemic inflammation
b
0
1
1
0
male
0
0
65
61-70
Vietnamese
513
original
https://jamanetwork.com/journals/jama/fullarticle/2735973
A 51-year-old man with a history of hypertension, hypercholesterolemia, and nephrolithiasis presented to the emergency department with left flank and left upper quadrant abdominal pain. He stated that “I think I have a kidney stone blocking my kidney” but denied nausea, vomiting, fever, dysuria, hematuria, or weight loss. He had no history of chronic abdominal pain. Past surgical history was noteworthy for emergency exploratory laparotomy and splenectomy for blunt trauma 14 years prior. Physical examination revealed a comfortable-appearing patient in no acute distress. His temperature was 36.8°C; pulse, 77/min; and blood pressure, 125/86 mm Hg. Abdominal palpation elicited only mild epigastric tenderness without masses or rebound. There was no costovertebral angle tenderness.Laboratory data revealed normal levels of serum urea nitrogen and creatinine and normal liver function. A complete blood cell count showed leukocytosis (15 300/μL with 54% neutrophils, 38% lymphocytes, 8% monocytes); hemoglobin level, 15.5 g/dL; and hematocrit, 45.2%. Urinalysis revealed no red or white blood cells and was negative for leukocyte esterase and nitrate. A contrast-enhanced computed tomography (CT) scan of the abdomen and pelvis revealed a well-circumscribed mass with a small focus of central high density resulting in surrounding streak artifact (Figure 1).Axial image from an unenhanced computed tomogram of the patient’s abdomen.Perform an exploratory laparotomy and excision of the massPerform an interventional radiology–guided biopsy of the mass What Would You Do Next?
Perform esophagogastroduodenoscopy (EGD)
Perform diagnostic laparoscopy with peritoneal washings
Perform an exploratory laparotomy and excision of the mass
Perform an interventional radiology–guided biopsy of the mass
Gossypiboma (retained surgical sponge)
C
Perform an exploratory laparotomy and excision of the mass
The key to the correct diagnosis is the remote history of a trauma laparotomy and the CT scan scout image that shows the characteristic finding of a radiopaque marker associated with a retained surgical sponge (Figure 2). All surgical sponges are equipped with such a marker, which manifests radiographically as a convoluted hyperdensity. These abdominal lesions may be confused with a gastrointestinal stromal tumor or other gastrointestinal malignancies. Since the lesion appears radiographically to be outside the lumen of the gastrointestinal tract, EGD has no role. Without a confirmed diagnosis of malignancy, laparoscopy with peritoneal washings to identify occult metastases would be premature. An interventional radiology–guided biopsy is unnecessary, given the early recognition of the radiopaque marker associated with gossypiboma.A sponge or laparotomy pad is the item most commonly retained after surgical procedures and when left behind in a body cavity is known as a gossypiboma. The abdominal cavity is the most frequent location of retained surgical items (56%). Recent reviews list the incidence of retained surgical items as between 1 in 8800 and 1 in 18 760 operations.1A retained surgical item should always be considered in the differential diagnosis of a patient with a history of prior emergency abdominal surgery who presents with pain, infection, or a palpable mass. Typically, retained laparotomy pads are detected early after the procedure but may be discovered days, months, or even decades later, as in this unusual instance. The clinical manifestations of a gossypiboma depend on the time elapsed from the index operation to detection. Many patients are asymptomatic, although abdominal pain is the most common symptom. Other manifestations can include bleeding, fever, intestinal obstruction, perforation, or fistula formation into adjacent organs.2 Approximately 10% to 15% of cases of retained sponges are complicated by acute intra-abdominal sepsis and can lead to peritonitis and, rarely, death. Alternatively, the object may become encapsulated by a giant-cell foreign body reaction and form a “mass,” which can mimic malignancy.3The diagnosis is usually made with plain radiography if the characteristic radiopaque marker is identified. However, the marker can degrade over time, making the diagnosis more challenging. CT imaging can reveal a thick-walled mass with or without gas, a cystic lesion with an internal spongiform appearance, concentric layering, or mottled mural calcification.4Removal of gossypibomas requires repeat surgical intervention; therefore, prevention is key. The biggest risk factors for a retained surgical item are emergency operation, unplanned change in the operation, and a high body mass index.1,5 In 80% to 90% of instances in which a surgical sponge is retained, the sponge count is erroneously documented as “correct.”6 However, additional factors contributing to retained surgical items include distraction of operating room personnel, poor operative team communication, and lack of surgeon participation in the final sponge and instrument count.7 The Joint Commission recommends use of intraoperative radiographs whenever the surgical sponge count is incorrect or routinely in high-risk cases, such as lengthy operations extending through more than 1 nursing shift, a change in the planned operation, and operations involving 2 or more surgical teams.8 Even when intraoperative radiographs are performed, retained items can still be missed, eg, when the film does not include the area in question, in patients with high body mass index, if inadequate history is provided to the interpreting radiologist, or when a junior member of the team reads the film. Furthermore, radiopaque markers can be misidentified as clips, intestinal contrast, drains, calcifications, or wires. Newer technologies that augment manual system include computerized detection systems with bar code–embedded sponges or radiofrequency-tagged sponges that can be detected with a special handheld wand.9Given imaging findings suggesting that the mass was extraluminal, the patient underwent excision of the gossypiboma, which was mildly adherent to the greater curvature of the stomach and transverse colon but easily removed. The resection was initially uncomplicated, without compromise to surrounding viscera or vascular structures. The specimen appeared as a tan, heavily encapsulated mass that contained the retained surgical sponge. On the seventh postoperative day the patient developed peritonitis and was taken back to the operating room and discovered to have an ischemic transverse colon; the etiology for this was unclear. An extended right hemicolectomy and ileostomy were performed. Six months later his ileostomy was reversed and he has subsequently done well.
General
A 51-year-old man with a history of hypertension, hypercholesterolemia, and nephrolithiasis presented to the emergency department with left flank and left upper quadrant abdominal pain. He stated that “I think I have a kidney stone blocking my kidney” but denied nausea, vomiting, fever, dysuria, hematuria, or weight loss. He had no history of chronic abdominal pain. Past surgical history was noteworthy for emergency exploratory laparotomy and splenectomy for blunt trauma 14 years prior. Physical examination revealed a comfortable-appearing patient in no acute distress. His temperature was 36.8°C; pulse, 77/min; and blood pressure, 125/86 mm Hg. Abdominal palpation elicited only mild epigastric tenderness without masses or rebound. There was no costovertebral angle tenderness.Laboratory data revealed normal levels of serum urea nitrogen and creatinine and normal liver function. A complete blood cell count showed leukocytosis (15 300/μL with 54% neutrophils, 38% lymphocytes, 8% monocytes); hemoglobin level, 15.5 g/dL; and hematocrit, 45.2%. Urinalysis revealed no red or white blood cells and was negative for leukocyte esterase and nitrate. A contrast-enhanced computed tomography (CT) scan of the abdomen and pelvis revealed a well-circumscribed mass with a small focus of central high density resulting in surrounding streak artifact (Figure 1).Axial image from an unenhanced computed tomogram of the patient’s abdomen.Perform an exploratory laparotomy and excision of the massPerform an interventional radiology–guided biopsy of the mass
what would you do next?
What would you do next?
Perform esophagogastroduodenoscopy (EGD)
Perform an interventional radiology–guided biopsy of the mass
Perform an exploratory laparotomy and excision of the mass
Perform diagnostic laparoscopy with peritoneal washings
c
1
1
1
1
male
0
0
51
51-60
White
514
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2732678
A woman in her 50s with relapsed refractory acute myeloid leukemia and a remote history of breast cancer was admitted for treatment of presumed fungal pneumonia based on new pulmonary nodules on imaging and elevated serum β-D-glucan levels. She subsequently developed multiple asymptomatic pink papules on the trunk and extremities. The patient was afebrile and was receiving broad-spectrum antibiotic, antifungal, and antiviral treatment for a prior episode of neutropenic fever. Clinical examination revealed scattered pink papules with a white firm center and dark brown core scattered throughout the chest, abdomen (Figure, A and B), back, groin, and upper and lower extremities. Laboratory workup was significant for leukopenia with neutropenia, anemia, and thrombocytopenia. Creatinine and transaminase levels were within normal limits. Blood cultures were negative. A punch biopsy from the left flank was obtained for hematoxylin-eosin staining, and a punch biopsy from the left leg was sent for tissue and fungal culture. Histopathological analysis was subsequently performed (Figure, C and D).A, Multiple papules on the abdomen. B, Closer view of a single papule. C and D, Hematoxylin-eosin–stained lesional specimens. What Is Your Diagnosis?
Disseminated candidiasis with transepidermal elimination
Reactive perforating collagenosis
Disseminated cryptococcosis
Malassezia (formerly Pityrosporum) folliculitis
A. Disseminated candidiasis with transepidermal elimination
A
Disseminated candidiasis with transepidermal elimination
The analysis of the skin biopsy specimen revealed a large cluster of fungal organisms in the upper dermis extruding through a cuplike invagination of the epidermis into the stratum corneum (Figure, C and D). Periodic acid–Schiff (PAS) staining highlighted these fungal elements. Wound and fungal cultures subsequently grew Candida albicans. Histologic and laboratory findings supported a diagnosis of disseminated cutaneous candidiasis with transepidermal elimination, correlating clinically with the central core seen on examination. Ophthalmologic evaluation revealed chorioretinitis secondary to Candida infection, and otolaryngology assessment revealed thrush in the oropharynx. Blood cultures, however, remained negative. The patient’s antifungal regimen included amphotericin B and micafungin. The patient was transferred to the intensive care unit for septic shock 2 weeks after presentation and died shortly thereafter.Candida albicans is a dimorphic fungus that colonizes skin, genital, and oral mucosa in up to 70% of healthy individuals.1 The present patient displayed many of the known risk factors for invasive candidiasis, including prolonged hospital stays, neutropenia, and corticosteroid and broad-spectrum antibiotic treatment.2 Although her blood cultures were negative, she exhibited evidence of C albicans in the skin, oropharynx, and eyes. Blood cultures are negative in 30% to 50% of patients with disseminated candidiasis, and that number increases with use of systemic antifungal therapy.3,4While C albicans has traditionally been the predominant species in invasive Candida infections, there has been an increase in the incidence of non–C albicans Candida infections since the 1980s.2 A recent review of disseminated candidiasis in patients with neutropenia and skin lesions found that Candida tropicalis was the most commonly identified species, followed by Candida krusei and C albicans.5 It has been theorized that increased use of empirical fluconazole may contribute to infections with non-albicans species, specifically Candida glabrata and Candida krusei.6Candida albicans naturally exists in a spore form in the epidermis and invades as filamentous pseudohyphae in the dermis. The presence of C albicans at various levels of skin directs differentiated immune responses. Cutaneous host defenses against C albicans involve innate immunity via sensory cutaneous nerves, dermal dendritic cells, and melanocytes and adaptive immunity via helper T cell type 17 lineage.1 The present case reveals a clinicopathologic correlation between the punctumlike cores noted in the patient’s cutaneous papules and the transepidermal elimination of Candida spores. To our knowledge, this presentation of disseminated Candida in the skin has not yet been reported.Reactive perforating collagenosis may also present with papules with central keratotic plugs; however, biopsy would reveal collagen fibers rather than Candida organisms extruding through the dermal-epidermal junction. Disseminated cryptococcosis is a common opportunistic infection often associated with HIV infection and clinically presents as umbilicated papules; pathologic analysis reveals grouped pleomorphic yeasts and a gelatinous capsule, which stains red with mucicarmine. Malassezia folliculitis is a pruritic eruption caused by Malassezia furfur that presents as folliculocentric papules and pustules on the trunk, with histopathologic analysis revealing suppurative folliculitis with PAS stain highlighting fungal organisms in the follicle and surrounding dermis. The present patient had an increased risk for Malassezia folliculitis given immunosuppression and recent antibiotic courses, but the lesions were not folliculocentric.
Dermatology
A woman in her 50s with relapsed refractory acute myeloid leukemia and a remote history of breast cancer was admitted for treatment of presumed fungal pneumonia based on new pulmonary nodules on imaging and elevated serum β-D-glucan levels. She subsequently developed multiple asymptomatic pink papules on the trunk and extremities. The patient was afebrile and was receiving broad-spectrum antibiotic, antifungal, and antiviral treatment for a prior episode of neutropenic fever. Clinical examination revealed scattered pink papules with a white firm center and dark brown core scattered throughout the chest, abdomen (Figure, A and B), back, groin, and upper and lower extremities. Laboratory workup was significant for leukopenia with neutropenia, anemia, and thrombocytopenia. Creatinine and transaminase levels were within normal limits. Blood cultures were negative. A punch biopsy from the left flank was obtained for hematoxylin-eosin staining, and a punch biopsy from the left leg was sent for tissue and fungal culture. Histopathological analysis was subsequently performed (Figure, C and D).A, Multiple papules on the abdomen. B, Closer view of a single papule. C and D, Hematoxylin-eosin–stained lesional specimens.
what is your diagnosis?
What is your diagnosis?
Disseminated cryptococcosis
Malassezia (formerly Pityrosporum) folliculitis
Disseminated candidiasis with transepidermal elimination
Reactive perforating collagenosis
c
0
1
1
1
female
0
1
55
51-60
White
515
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2728242
A 30-year-old woman presented with a 4-month history of widespread thickened, verrucous, hyperpigmented plaques distributed symmetrically on her body, especially on the axillae (Figure 1A), anogenital region, inguinal skin, and both palms, with slight itching. The verrucous plaques also involved the conjunctiva, lips, and gingiva (Figure 1B). The patient had experienced a weight loss of 6 kg during the last 3 months. Within the last month, the patient’s thyroid became enlarged, and she developed anorexia and abdominal discomfort. Laboratory test results revealed remarkably elevated levels of carcinoembryonic antigen, carbohydrate antigen 19-9, carbohydrate antigen 125, tissue polypeptide antigen and carbohydrate antigen 242. Biopsy specimens of lesions on the axillae and lips both revealed a papillary structure with hyperkeratosis. An endoscopic examination and computed tomographic scan of the abdomen were performed.Axilla with velvety, hyperpigmented skin (A) and verrucous plaques on the lips and gingiva (B). What Is Your Diagnosis?
Benign acanthosis nigricans
Cutaneous T-cell lymphoma
Malignant acanthosis nigricans
Malignant Addison disease
C. Malignant acanthosis nigricans
C
Malignant acanthosis nigricans
The keys to the correct diagnosis are the verrucous, hyperpigmented plaques in atypical sites (mucous membranes, palms and anogenital region), the additional paraneoplastic findings (tripe palms), and recent obvious weight loss. Most cases of acanthosis nigricans (AN) are benign and are associated with obesity and insulin resistance in which blood glucose and insulin levels are important for diagnosis. The disease is less commonly associated with a malignant neoplasm. Malignant AN is predominantly associated with gastrointestinal tract tumors, especially carcinoma of the stomach.1The gastroscopy findings revealed esophageal papillomatous polyposis and a submucosal mass involving the gastric angle. Biopsy results revealed that the mass was a mix of adenocarcinoma and signet-ring cell carcinoma (Figure 2). A computed tomographic scan of the abdomen showed a thickening of the gastric wall and multiple organ metastases. The results of biopsies of the thyroid and pancreas suggested metastases of gastric cancer. The patient was then transferred for further chemotherapy without any dermatological treatment and died 4 months later due to cancer progression.Histologic specimen of the gastric mass showed various types of cancer. Poorly differentiated adenocarcinoma and signet-ring cell carcinoma had developed in the lamina propria of the gastric angle (hematoxylin-eosin stain, original magnification ×100).Acanthosis nigricans is a reactive skin pattern that presents as velvety to verrucous hyperpigmented plaques in body folds such as the axilla, neck, groin, and umbilicus. Malignant AN has a more striking clinical presentation involving unusual locations such as the oral cavity, conjunctiva, and palms, in accordance with the severity of the disease.2-4 It can occur with other cutaneous markers of internal malignant neoplasm, such as tripe palms and the Leser-Trélat sign.2,3 It is mostly associated with gastric adenocarcinoma, followed by pulmonary carcinoma, hepatic carcinoma, esophageal carcinoma, and ovarian cancer,2,3 arising either before or after detection of the tumor. The onset for most patients is between age 41 and 70 years.1 Patients with malignant AN are generally not obese but have experienced a recent weight loss. Histologically, malignant and benign AN cannot be distinguished from each other. Their major features are hyperkeratosis and epidermal papillomatosis with increased melanin in the basal layer of the epidermis. A thorough endoscopic and radiological screening for cancer is much more important than serological testing.Pathogenically, elevated levels of transforming growth factor α participate in tumor progression through autosecretion and paracrine secretion, stimulating keratinocyte growth via epidermal growth factor receptors. Insulinlike growth factor 1, fibroblast growth factor, and melanocyte-stimulating hormone α regulate melanocyte pigmentation and stimulate the growth of keratocytes, which also play roles in the pathogenesis of hyperplasia and hyperpigmentation.5The management of malignant AN should be focused on the underlying neoplasm. The efficacy of therapies intended to directly improve the skin lesions of AN, such as using topical retinoids or vitamin D analogs, is limited. Improvement or resolution of AN has been reported in patients successfully treated for the associated cancer.6 Nevertheless, the prognosis for malignant AN is poor since the cancer is frequently diagnosed at an advanced stage. Patients may be misdiagnosed with eczema or other skin disorders during the early stages of the disease. Timely recognition of malignant AN could lead to earlier detection of the carcinoma and may improve the prognosis of these patients.
Oncology
A 30-year-old woman presented with a 4-month history of widespread thickened, verrucous, hyperpigmented plaques distributed symmetrically on her body, especially on the axillae (Figure 1A), anogenital region, inguinal skin, and both palms, with slight itching. The verrucous plaques also involved the conjunctiva, lips, and gingiva (Figure 1B). The patient had experienced a weight loss of 6 kg during the last 3 months. Within the last month, the patient’s thyroid became enlarged, and she developed anorexia and abdominal discomfort. Laboratory test results revealed remarkably elevated levels of carcinoembryonic antigen, carbohydrate antigen 19-9, carbohydrate antigen 125, tissue polypeptide antigen and carbohydrate antigen 242. Biopsy specimens of lesions on the axillae and lips both revealed a papillary structure with hyperkeratosis. An endoscopic examination and computed tomographic scan of the abdomen were performed.Axilla with velvety, hyperpigmented skin (A) and verrucous plaques on the lips and gingiva (B).
what is your diagnosis?
What is your diagnosis?
Malignant Addison disease
Malignant acanthosis nigricans
Cutaneous T-cell lymphoma
Benign acanthosis nigricans
b
0
1
1
1
female
0
0
30
21-30
null
516
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2729678
A 70-year-old Indian woman presented with a generalized rash over the lower extremity associated with pedal edema extending up to the shins. The onset was gradual and started on the lower extremities and gradually extended to involve the lower trunk. The rash was associated with mild discomfort. The patient denied itching, weeping, or bleeding from the rash. She denied any constitutional symptoms (weight loss, fever, night sweats), joint pains, history of blood in urine, periorbital edema, and recent intake of medications or herbal medications. Social history was significant for 25 pack-years of smoking. Physical examination revealed multiple well-defined, coalescent, erythematous, palpable, purpuralike lesions that were predominantly present over the trunk and lower extremities (Figure, A and B). Laboratory test results at presentation included a hemoglobin level of 11.2 g/dL (to convert to g/L, multiply by 10), mean corpuscular volume of 84 fL, erythrocyte sedimentation rate of 72 mm/h, and a lactate dehydrogenase level of 412 IU/L. Hepatic and renal function levels were within normal limits. Complement levels were normal. Hepatitis B and C and HIV profiles were negative. A chest radiograph showed a 2-cm lesion in the upper lobe of the left lung. A computed tomographic scan of the chest showed a spiculated mass in the same region along with mediastinal lymphadenopathy. An ultrasound of the abdomen showed bilateral adrenal masses. A lesional skin biopsy specimen was obtained (Figure, C).Coalescent, erythematous, palpable lesions on the lower extremity (A) and trunk (B). C, Lesional skin biopsy sample. What Is Your Diagnosis?
Acrokeratosis paraneoplastica
Cutaneous leukocytoclastic vasculitis
Purpura fulminans
Henoch-Schönlein purpura
B. Cutaneous leukocytoclastic vasculitis
B
Cutaneous leukocytoclastic vasculitis
The histopathologic findings from a lesional skin biopsy specimen revealed neutrophilic infiltration of the small blood vessels in the skin along with necrosis and extravasation of the red blood cells (Figure, C). The findings from a biopsy specimen of a lung lesion revealed small cell carcinoma. The patient was diagnosed with extensive-stage small cell lung cancer and began systemic chemotherapy with cisplatin and etoposide. Unfortunately, the patient died soon after the first dose of chemotherapy due to neutropenic sepsis.Cutaneous manifestation of an internal malignant neoplasm may present as direct infiltration into the skin or may present indirectly as a paraneoplastic syndrome (PNS).1 The Curth postulates are the cornerstone for defining the association between internal malignant neoplasms and cutaneous disorders.1 Leukocytoclastic vasculitis is a histopathologic term denoting vasculitis of the small vessels with predominantly neutrophilic infiltrate.2 After degranulation, the neutrophils undergo apoptosis, a process known as leukocytoclasis, releasing nuclear debris, which is also called nuclear dust.2 According to the consensus statement developed at the Chapel Hill Conference on Nomenclature of Systemic Vasculitis in 1994, cutaneous leukocytoclastic vasculitis (CLV) is defined as an“[i]solated cutaneous leukocytoclastic angiitis without systemic vasculitis or glomerulonephritis.”2,3 As a PNS, CLV is the most commonly diagnosed vasculitis (45%) followed by polyarteritis nodosa (36.7%).4 Cutaneous leukocytoclastic vasculitis occurs most frequently with hematologic cancers followed by lung, gastrointestinal, or urinary tract tumors.5,6 Approximately 20% to 26% of patients with paraneoplastic CLV have lung cancer.4,6 Among lung cancers, adenocarcinoma and squamous cell are most commonly associated with CLV.4,6,7Cutaneous leukocytoclastic vasculitis presents as a palpable purpuric lesion that usually involves the dependent areas (mostly feet and lower extremities) and rarely involves the upper extremities and trunk.2 The lesions are usually chronic and persistent when associated with a malignant neoplasm.2 In contrast to the chronic pattern of CLV, Henoch-Schönlein purpura (HSP) is a recurring form of CLV that presents with symptom-free intervals.3,8 Deposits of IgA in the blood vessels are a defining feature of HSP. Henoch-Schönlein purpura usually affects children and has been reported as a PNS associated with solid organ malignant neoplasms, including small cell lung cancer.2,9 The diagnostic workup includes a skin biopsy to differentiate other cutaneous lesions from underlying vasculitis. Preferably, an immunofluorescence stain should be done in addition to a hematoxylin and eosin stain.2 However, the diagnostic yield of this test is considerably reduced after 48 hours owing to the destruction and removal of immunoglobulins deposited in the vessel wall.2 Judicious use of immunofluorescence for selected cases has been advocated recently, but the test is not readily available in settings with constrained resources, such as ours.10 In patients with chronic CLV, other causes not related to malignant neoplasms should be excluded, including hepatitis B and/or C virus, HIV, cryoglobulinemia, and systemic small-vessel vasculitis.The pathogenesis of CLV remains anecdotal. Leukocytoclastic vasculitis is a common response to circulating aggressors, be it microorganisms or immune complexes, and is likely influenced by the heterogeneity of the endothelium of postcapillary venules.2 The endothelial cells in these venules play a role in antigen presentation, and express Toll-like receptor family members, CD32 molecules, and histamine H1 receptors.2 Cutaneous leukocytoclastic vasculitis usually begins with deposition of immune complex in the vessel walls, leading to activation of the complement cascade (complement components C3a and C5a), which attracts neutrophils and eventually leads to degranulation and vessel wall destruction.2 The treatment of CLV associated with systemic malignant neoplasms is directed toward the primary cause, whereby improvement or worsening of the rash is seen in concordance with the treatment response of the underlying cancer.1,4 This case illustrates the importance of recognizing cutaneous manifestations of paraneoplastic syndromes, which can prompt an early diagnosis of an occult malignant neoplasm.
Oncology
A 70-year-old Indian woman presented with a generalized rash over the lower extremity associated with pedal edema extending up to the shins. The onset was gradual and started on the lower extremities and gradually extended to involve the lower trunk. The rash was associated with mild discomfort. The patient denied itching, weeping, or bleeding from the rash. She denied any constitutional symptoms (weight loss, fever, night sweats), joint pains, history of blood in urine, periorbital edema, and recent intake of medications or herbal medications. Social history was significant for 25 pack-years of smoking. Physical examination revealed multiple well-defined, coalescent, erythematous, palpable, purpuralike lesions that were predominantly present over the trunk and lower extremities (Figure, A and B). Laboratory test results at presentation included a hemoglobin level of 11.2 g/dL (to convert to g/L, multiply by 10), mean corpuscular volume of 84 fL, erythrocyte sedimentation rate of 72 mm/h, and a lactate dehydrogenase level of 412 IU/L. Hepatic and renal function levels were within normal limits. Complement levels were normal. Hepatitis B and C and HIV profiles were negative. A chest radiograph showed a 2-cm lesion in the upper lobe of the left lung. A computed tomographic scan of the chest showed a spiculated mass in the same region along with mediastinal lymphadenopathy. An ultrasound of the abdomen showed bilateral adrenal masses. A lesional skin biopsy specimen was obtained (Figure, C).Coalescent, erythematous, palpable lesions on the lower extremity (A) and trunk (B). C, Lesional skin biopsy sample.
what is your diagnosis?
What is your diagnosis?
Acrokeratosis paraneoplastica
Henoch-Schönlein purpura
Purpura fulminans
Cutaneous leukocytoclastic vasculitis
d
1
1
1
1
female
0
0
70
61-70
Indian
517
original
https://jamanetwork.com/journals/jamacardiology/fullarticle/2735318
A woman in her early 60s with a medical history of chest pain requiring a coronary angiogram (which demonstrated only mild luminal irregularities) approximately 1 year prior to presentation, hypertension, long-standing dyslipidemia, and multiparity presented to the emergency department with a sudden onset of dull, pressurelike chest pain radiating to the left arm. The pain started at rest, was 10 of 10 in severity, and was not alleviated by changing position. She also experienced concomitant nausea and diaphoresis. Her family history was notable for hypertension, and no other systemic diseases were reported. On physical examination, her blood pressure was 115/76 mm Hg, heart rate was regular at 72 beats per minute, respiratory rate was 20 breaths per minute, and oxygen saturation level was 98% on room air. The patient was afebrile. Physical examination also revealed mild bibasilar crackles and jugular venous distention, and no murmurs or lower extremity edema. Laboratory test results demonstrated an elevated troponin, and the 12-lead electrocardiogram showed marked ST-segment elevations in leads V2 to V5 (Figure, A). The ST-segment elevation myocardial infarction (STEMI) pager was activated, and the patient was transferred urgently to the catheterization laboratory. Coronary angiography of the patient’s left anterior descending coronary artery was performed, and the findings are depicted in the Figure, B.A, Twelve-lead electrocardiogram. B, Left anterior descending coronary artery angiogram.Treat medically with aspirin, β-blocker, clopidogrel, and a statinRevascularize with percutaneous coronary intervention and start medical therapy What Would You Do Next?
Treat medically with aspirin alone
Treat medically with aspirin, β-blocker, and clopidogrel
Treat medically with aspirin, β-blocker, clopidogrel, and a statin
Revascularize with percutaneous coronary intervention and start medical therapy
Spontaneous coronary artery dissection
C
Treat medically with aspirin, β-blocker, clopidogrel, and a statin
C. Treat medically with aspirin, β-blocker, clopidogrel, and a statinSpontaneous coronary artery dissection (SCAD) is a nontraumatic and noniatrogenic separation of the coronary arterial wall, typically leading to the formation of a false lumen, in the presence or absence of significant obstructive coronary artery disease.1 While the precise pathophysiological mechanism remains unclear, a tear in the intima of a coronary vessel or the development of an intramural hematoma due to bleeding from the rupture of the vasa vasorum have been suggested as possibilities.2,3 The disease process tends to affect women, particularly young women, with underlying fibromuscular dysplasia or other connective tissue disease (such as Marfan syndrome, Loeys-Dietz syndrome, and Ehlers-Danlos syndrome type 4).4 Hormonal therapy and postpartum or multiparous state are also known risk factors. Without early detection and treatment, SCAD can be life-threatening.3,5,6SCAD remains an underdiagnosed entity primarily owing to a low index of clinical suspicion and nonfamiliarity of clinicians with the angiographic variants of SCAD. Moreover, its presentation often mimics other more common acute coronary syndromes, including STEMI or non-STEMI, and therefore further confounds the diagnosis.7 Many patients with SCAD present with classic, dull, pressurelike chest pain at rest and have electrocardiographic changes (ie, ST-segment elevations or depressions).6Coronary angiography is the primary imaging modality used to diagnose SCAD.8 The following 3 specific angiographic findings outlined by Saw et al1 are often observed:Type 1: pathognomonic angiographic appearance of SCAD with contrast dye staining of the arterial wall with multiple radiolucent lumens;Type 2: diffuse (>20 mm), smooth stenosis of varying severity;Type 3: focal areas of stenosis that mirror atherosclerosis but are usually limited to 1 coronary vessel and are long (11-20 mm) and linear.Type 1: pathognomonic angiographic appearance of SCAD with contrast dye staining of the arterial wall with multiple radiolucent lumens;Type 2: diffuse (>20 mm), smooth stenosis of varying severity;Type 3: focal areas of stenosis that mirror atherosclerosis but are usually limited to 1 coronary vessel and are long (11-20 mm) and linear.The angiographic image of the patient’s left anterior descending coronary artery (Figure, B) depicts type 2 SCAD given the diffuse, smooth luminal narrowing observed (arrowheads). However, coronary angiography is imperfect for diagnosing SCAD because of limited visualization of the intra-arterial wall.8 Optical coherence tomography and intravascular ultrasonography have emerged as criterion standard diagnostic methods as they allow for direct visualization of vascular intima and media. However, these methods are not used frequently given the risk of propagating the dissection both mechanically, with the imaging catheter or guidewire, and hydraulically, with contrast injection required for optical coherence tomography.1In patients without persistent ischemia, a conservative treatment with aspirin, β-blocker, and clopidogrel is preferred.1 If a patient has concomitant long-standing dyslipidemia (as in this patient), the addition of a statin is suggested.1,9 In hemodynamically stable patients, percutaneous coronary intervention is relatively contraindicated given the risk of iatrogenic propagation of the dissection and exacerbation of an already ischemic myocardium.1The patient was treated conservatively with aspirin, β-blocker, clopidogrel, and a statin, and an initial improvement was observed in her symptoms. However, her symptoms became complicated thereafter by recurrent ischemia and infarction from the dissection, leading to left ventricular dysfunction (ejection fraction, 30%-35%), cardiogenic shock, and the eventual placement of a left ventricular assist device. While uncommon, left ventricular failure is a consequence of SCAD and results in extensive infarction.1 It usually occurs in patients with ongoing or recurrent ischemia, especially involving the left main coronary artery, and necessitates revascularization with either percutaneous coronary intervention or coronary artery bypass graft.1 The patient in this case died of a stroke considered to be a complication of her left ventricular assist device. However, her case raises the importance of physician awareness of SCAD and depicts that such patients must be regularly monitored for ongoing or recurrent ischemia despite being provided with optimal medical therapy.
Cardiology
A woman in her early 60s with a medical history of chest pain requiring a coronary angiogram (which demonstrated only mild luminal irregularities) approximately 1 year prior to presentation, hypertension, long-standing dyslipidemia, and multiparity presented to the emergency department with a sudden onset of dull, pressurelike chest pain radiating to the left arm. The pain started at rest, was 10 of 10 in severity, and was not alleviated by changing position. She also experienced concomitant nausea and diaphoresis. Her family history was notable for hypertension, and no other systemic diseases were reported. On physical examination, her blood pressure was 115/76 mm Hg, heart rate was regular at 72 beats per minute, respiratory rate was 20 breaths per minute, and oxygen saturation level was 98% on room air. The patient was afebrile. Physical examination also revealed mild bibasilar crackles and jugular venous distention, and no murmurs or lower extremity edema. Laboratory test results demonstrated an elevated troponin, and the 12-lead electrocardiogram showed marked ST-segment elevations in leads V2 to V5 (Figure, A). The ST-segment elevation myocardial infarction (STEMI) pager was activated, and the patient was transferred urgently to the catheterization laboratory. Coronary angiography of the patient’s left anterior descending coronary artery was performed, and the findings are depicted in the Figure, B.A, Twelve-lead electrocardiogram. B, Left anterior descending coronary artery angiogram.Treat medically with aspirin, β-blocker, clopidogrel, and a statinRevascularize with percutaneous coronary intervention and start medical therapy
what would you do next?
What would you do next?
Revascularize with percutaneous coronary intervention and start medical therapy
Treat medically with aspirin alone
Treat medically with aspirin, β-blocker, and clopidogrel
Treat medically with aspirin, β-blocker, clopidogrel, and a statin
d
1
1
1
1
female
0
0
62
61-70
null
518
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2731889
A 59-year-old Hispanic woman presented with a 6-month history of intermittent pain in the right posterior mandibular gingiva. Her local dentist had prescribed an antibiotic and performed a tooth extraction in the area 6 months prior with no improvement in symptoms. Ultimately, her pain became persistent and radiated to the right lower lip. She had a history of stage IV endometrial serous adenocarcinoma (ESA) 4 years prior that was treated with total hysterectomy, bilateral salpingo-oophorectomy, omentectomy, and peritoneal lavage. She had developed a recurrence for which she had just completed 3 cycles of treatment with carboplatin. She also had a history of anxiety, hypertension, and pterygium, and she was taking gabapentin, lorazepam, prochlorperazine, metoprolol succinate, bromfenac and ketorolac ophthalmic solution, and aprepitant. Intraoral examination revealed a 1.5 × 1.0-cm firm, erythematous gingival mass in the area of missing right mandibular second bicuspid and first molar with buccal and lingual expansion (Figure 1A). The right mandibular first bicuspid was grade 3 mobile. A periapical radiograph was obtained, and results showed a diffuse, poorly demarcated radiolucency measuring approximately 1.2 × 0.8 cm (Figure 1B). Findings from an incisional biopsy of the mass showed an infiltrative tumor (Figure 1C). Tumor cells were positive for CK7, PAX8, and WT-1, and negative for CK20 and TTF-1 (Figure 1C, inset).A, A 1.5 × 1-cm gingival mass is distal to the right mandibular first bicuspid. B, Diffuse, poorly demarcated radiolucency is seen in the area of the missing right mandibular second bicuspid and first molar (arrowheads). C, Multiple nests of round to ovoid epithelial cells form ducts (hematoxylin-eosin stain, original magnification ×100). Diffuse nuclear positivity for PAX8 (immunohistochemistry, original magnification ×200) (inset). What Is Your Diagnosis?
Pyogenic granuloma
Gingival/periodontal abscess
Oral metastasis
Squamous cell carcinoma
C. Oral metastasis
C
Oral metastasis
The histopathological and immunohistochemical findings were consistent with the patient’s preexisting ESA. Oral metastases in the jaws and soft tissues are uncommon and account for only 1% to 8% of all malignancies in the oral cavity. Bony metastases, particularly in the mandible, are twice as common as those in the oral soft tissues of the gingiva and tongue.1,2 The most common cancer that metastasizes in the oral cavity in female patients is carcinoma of the breast (25.4%-36.6%), while metastatic cancers of the female genital tract in the oral cavity are rare and have a frequency of 3% to 9%.2,3 The clinical presentation of metastatic disease in the oral cavity may be nonspecific. Patients may have pain, swelling, and/or bleeding. These signs may lead a clinician to first consider odontogenic infection or inflammatory gingival nodules such as pyogenic granuloma.3,4 Because odontogenic infections often present in a similar fashion and metastatic tumors are rare, lesions are often treated with antibiotics, which leads to diagnostic delay like with this case.3 It is likely the metastatic ESA in this case had originated in the bone (as it does in approximately two-thirds of cases) with extension into the soft tissues that presented as a gingival mass.Endometrial carcinoma is the most common gynecological cancer in the United States.5 There are 2 major categories of endometrial carcinoma: type 1 (endometrioid adenocarcinoma) and type 2 (ESA and clear cell carcinoma).6 Endometrial serous adenocarcinoma is an aggressive tumor and a histopathologically distinct subtype of type 2 endometrial carcinoma, which accounts for 5% to 10% of all endometrial cancers.7,8 It typically occurs in postmenopausal women with a median age at presentation of 68 years (range, 44-93 years).5 Unlike endometrioid adenocarcinoma, ESA is estrogen independent and arises from atrophic endometrium.9 Patients with ESA have a 5-fold increased risk of tumor recurrence compared with patients with endometrioid adenocarcinoma.6 In this case, despite previous total hysterectomy, bilateral salpingo-oophorectomy, omentectomy, and peritoneal lavage, the patient developed recurrence after 4 years. Results of a computed tomography scan revealed widespread recurrent disease despite the patient having been recently treated with carboplatin. Direct extension of the tumor into the fallopian tube was seen, and metastatic foci were identified in the mandible (Figure 2), lung, right common iliac, and mediastinal lymph nodes.Computed tomography scan of poorly demarcated radiolucency with soft tissue extension in the area of the right mandibular first bicuspid and missing second bicuspid and first molar (arrowheads).Distant metastasis of ESA is a common finding at the time of initial presentation.5 In a study of 129 cases, extrauterine metastasis was found in 72% of the cases.10 Endometrial serous adenocarcinoma is associated with poor prognosis and accounts for 40% of endometrial cancer–related deaths. The 5-year overall survival rate is 62.9% and 19.9% for stage I and IV disease, respectively.5,6 The patient was treated with a course of palliative radiotherapy of 3 Gy in 10 fractions. However, her disease worsened with progression of the mandibular mass and the lung nodules and, as such, she was started on pemetrexed therapy. At time of report, the patient was alive, 4 years and 10 months after the initial diagnosis of her tumor and 10 months after the diagnosis of her oral metastasis. Metastatic disease should always be considered in the broad differential diagnosis of a gingival nodule.
General
A 59-year-old Hispanic woman presented with a 6-month history of intermittent pain in the right posterior mandibular gingiva. Her local dentist had prescribed an antibiotic and performed a tooth extraction in the area 6 months prior with no improvement in symptoms. Ultimately, her pain became persistent and radiated to the right lower lip. She had a history of stage IV endometrial serous adenocarcinoma (ESA) 4 years prior that was treated with total hysterectomy, bilateral salpingo-oophorectomy, omentectomy, and peritoneal lavage. She had developed a recurrence for which she had just completed 3 cycles of treatment with carboplatin. She also had a history of anxiety, hypertension, and pterygium, and she was taking gabapentin, lorazepam, prochlorperazine, metoprolol succinate, bromfenac and ketorolac ophthalmic solution, and aprepitant. Intraoral examination revealed a 1.5 × 1.0-cm firm, erythematous gingival mass in the area of missing right mandibular second bicuspid and first molar with buccal and lingual expansion (Figure 1A). The right mandibular first bicuspid was grade 3 mobile. A periapical radiograph was obtained, and results showed a diffuse, poorly demarcated radiolucency measuring approximately 1.2 × 0.8 cm (Figure 1B). Findings from an incisional biopsy of the mass showed an infiltrative tumor (Figure 1C). Tumor cells were positive for CK7, PAX8, and WT-1, and negative for CK20 and TTF-1 (Figure 1C, inset).A, A 1.5 × 1-cm gingival mass is distal to the right mandibular first bicuspid. B, Diffuse, poorly demarcated radiolucency is seen in the area of the missing right mandibular second bicuspid and first molar (arrowheads). C, Multiple nests of round to ovoid epithelial cells form ducts (hematoxylin-eosin stain, original magnification ×100). Diffuse nuclear positivity for PAX8 (immunohistochemistry, original magnification ×200) (inset).
what is your diagnosis?
What is your diagnosis?
Oral metastasis
Gingival/periodontal abscess
Squamous cell carcinoma
Pyogenic granuloma
a
1
1
1
1
female
0
0
59
51-60
Hispanic
519
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2732495
A 27-year-old man with a history of pharyngeal gonorrhea presented with a 7-day history of severe sore throat and odynophagia, worse when swallowing solids than liquids. Owing to the odynophagia, he had presented to the emergency department for dehydration. At that time, he was febrile to 101°F and denied any dysphonia or dyspnea. The results of a rapid Streptococcus test, recent HIV screen, and a rapid plasma reagin test were all negative. He denied any genital lesions or symptoms at that time. He had been taking prophylactic emtricitabine-tenofovir daily and had received his last diphtheria booster 2 years prior to presentation. Treatment with amoxicillin clavulanate over several days had not prevented a worsening of his symptoms. On physical examination, there was no cervical lymphadenopathy, and there were no genital lesions. Flexible fiber-optic nasopharyngoscopy showed ulcerative lesions with exudate and erythema extending diffusely along the base of tongue, oropharynx, nasopharynx, hypopharynx, and epiglottis (Figure 1). What Is Your Diagnosis?
Diphtheria
Mononucleosis
Type 2 herpes simplex virus (HSV-2) pharyngitis
Pharyngeal gonorrhea
C. HSV-2 pharyngitis
C
Type 2 herpes simplex virus (HSV-2) pharyngitis
The patient underwent HIV, rapid plasma reagin, monospot, HSV-1, an HSV-2 blood tests. Oropharyngeal cultures were taken for bacteria, fungus, and gonorrhea/chlamydia. Results of a qualitative polymerase chain reaction assay performed on a tissue swab were positive for HSV-2 and negative for HSV-1. Screening detected HSV-1/HSV-2 IgM serum antibody, and IgG serum antibody findings were indeterminate, suggesting current or recent infection. All other laboratory and culture findings were negative; therefore, a diagnosis of HSV-2 pharyngitis was made. He subsequently began treatment with valacyclovir, 3 g/d. No exudates or lesions were noted on physical examination nor on flexible fiber-optic nasopharyngoscopy at his 2-week follow-up visit (Figure 2). At that time, he was no longer complaining of odynophagia.Type 2 HSV has seldom been associated with isolated oropharyngeal lesions, and only a handful of cases have been reported in the literature in immune-competent adults from oral genital transmission.1-3 Transmission of HSV is dependent on viral contact with mucosal surfaces or abraded skin in a susceptible seronegative individual. Although HSV-1 is highly prevalent in humans, oropharyngeal infections are rare.3,4 Type 2 HSV more commonly presents as a genital infection, and to our knowledge has only been reported to affect the oropharynx in neonates through vertical transmission, or by reactivation of latent virus in the immunocompromised state.1,5 On clinical presentation, patients with HSV pharyngitis often have ulcerative lesions with exudate. These lesions are thought to have originated with vesicular lesions and to have quickly progressed to the ulcerative state with exudate. In a study of 209 cases of clinically symptomatic primary HSV genital infections, Corey et al6 found that pharyngitis symptoms of ulcerative and exudative lesions of the posterior pharynx ranged from mild to severe. However, in the present case and in several previously reported cases, there has been no evidence of HSV-2 genital lesions, and HSV-2 remains an uncommonly recognized cause of oral lesions and isolated pharyngitis.1Diagnosis of HSV pharyngitis is made more challenging by the wide variation of findings on examination. Because white/gray exudate, with or without ulceration, is commonly seen in HSV pharyngitis, this may be difficult to distinguish from diphtheria. Diphtheria often presents with a similarly gradual onset of odynophagia, malaise, cervical lymphadenopathy, and a low-grade fever. In at least one-third of cases, a tightly adherent pseudomembrane can be observed, and may affect any portion of the respiratory tract extending from the nasal passages to the tracheobronchial tree.7 Patients with mononucleosis commonly present with tonsillar exudates, fever, and diffuse cervical lymphadenopathy.8 Oropharyngeal infections with Neisseria gonorrhoeae are most commonly asymptomatic, although sore throat and pharyngeal exudates with or without cervical lymphadenitis are present in some cases.9,10 Diphtheria, mononucleosis, HSV pharyngitis, and pharyngeal gonorrhea may all present in a similar manner, and only cultures or serologic testing can confirm one over the others. Thus, it is important to consider broad differential diagnoses in a patient for whom antibiotic therapy has failed.Given the rather broad range of differential diagnoses for pharyngeal exudates, a high index of suspicion is required for diagnosis of HSV pharyngitis. The patient in this case initially presented with odynophagia without any lesions, and he was treated for tonsillitis before returning for further workup and care. His history of pharyngeal gonorrhea and high-risk sexual behavior were concerning for infectious causes; however, HSV was not originally a primary consideration.This case illustrates the importance of considering HSV-2 as a diagnosis for patients presenting with new exudative or ulcerative pharyngeal lesions. Although it remains a rare cause of pharyngitis, we recommend including an HSV-1/HSV-2 assay as part of the workup in complicated exudative pharyngitis that does not respond to antibiotic treatment.
General
A 27-year-old man with a history of pharyngeal gonorrhea presented with a 7-day history of severe sore throat and odynophagia, worse when swallowing solids than liquids. Owing to the odynophagia, he had presented to the emergency department for dehydration. At that time, he was febrile to 101°F and denied any dysphonia or dyspnea. The results of a rapid Streptococcus test, recent HIV screen, and a rapid plasma reagin test were all negative. He denied any genital lesions or symptoms at that time. He had been taking prophylactic emtricitabine-tenofovir daily and had received his last diphtheria booster 2 years prior to presentation. Treatment with amoxicillin clavulanate over several days had not prevented a worsening of his symptoms. On physical examination, there was no cervical lymphadenopathy, and there were no genital lesions. Flexible fiber-optic nasopharyngoscopy showed ulcerative lesions with exudate and erythema extending diffusely along the base of tongue, oropharynx, nasopharynx, hypopharynx, and epiglottis (Figure 1).
what is your diagnosis?
What is your diagnosis?
Type 2 herpes simplex virus (HSV-2) pharyngitis
Diphtheria
Mononucleosis
Pharyngeal gonorrhea
a
0
1
1
1
male
0
0
27
21-30
null
520
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2733030
A woman in her 60s with a history of end-stage renal disease who had a kidney transplant 9 months previously presented with progressive cough, dyspnea, and hoarseness. Her history also included chronic gastroesophageal reflux disease and a 14-pack-year smoking history. She initially presented 3 months prior with upper respiratory symptoms, which continued to worsen despite oral antibiotic therapy. Her cough became productive with white, frothy phlegm that was accompanied by fever, wheezing, hoarseness, and fatigue. Her dyspnea worsened in the supine position.Flexible laryngotracheoscopy results revealed diffuse, white-to-yellow pseudomembranous changes involving the glottis and supraglottis (Figure, A), posterior tracheal wall, and left mainstem bronchus with skip lesions in the subglottis and distal 1 cm of the trachea (Figure, B). Glottic mobility was limited in both abduction and adduction with substantial airway obstruction. Computed tomographic findings revealed posterior tracheal wall thickening with the loss of the fat plane between the trachea and esophagus, and a moderate to severe obstruction within the distal left mainstem bronchus. Biopsy results are shown in Figure, C.Laryngoscopic images demonstrate pseudomembranous plaques lining supraglottis and glottis (A) and posterior wall of the trachea (B). Supraglottic biopsy demonstrates branching organisms superficial to squamous epithelium (original magnification ×400) (C). What Is Your Diagnosis?
Posttransplant lymphoproliferative disorder
Squamous cell carcinoma
Nocardiosis
Aspergillosis
D. Aspergillosis
D
Aspergillosis
Pathology results from supraglottic biopsy revealed branching, fungal microorganisms among superficial fragments of squamous epithelium. Culture and bronchoalveolar lavage results confirmed the diagnosis of Aspergillosis laryngotracheobronchitis.Aspergillus tracheobronchitis (ATB) is an unusual form of pulmonary aspergillosis that is seen in fewer than 10% of aspergillosis-related cases.1 Patients with neutropenia and/or who are immunocompromised are at particular risk for ATB.2 Patients usually present with dyspnea, cough, and wheezing. Diagnosis of this rare entity is often delayed because of its nonspecific clinical presentation and lack of radiographic findings at early stages.3 Radiologic findings may include thickening of airways, patchy infiltrates, or no findings at all.1 Definitive diagnosis requires visualization with laryngoscopy or bronchoscopy and both pathological and microbiological biopsies.4In 1995, Denning5 proposed a classification of 3 forms of ATB:Obstructive ATB: characterized by thick mucus plugs filled with aspergillus hyphae with little mucosal inflammation or invasionPseudomembranous ATB: characterized by extensive inflammation of the tracheobronchial tree with a membrane overlying the mucosa containing Aspergillus speciesUlcerative ATB: characterized by focal invasion of the tracheobronchial mucosa and/or cartilage by fungal hyphae (typically found around the suture line in patients who have had a lung transplant)Obstructive ATB: characterized by thick mucus plugs filled with aspergillus hyphae with little mucosal inflammation or invasionPseudomembranous ATB: characterized by extensive inflammation of the tracheobronchial tree with a membrane overlying the mucosa containing Aspergillus speciesUlcerative ATB: characterized by focal invasion of the tracheobronchial mucosa and/or cartilage by fungal hyphae (typically found around the suture line in patients who have had a lung transplant)The preferred agents for treatment and prevention of ATB are triazoles, with voriconazole recommended as the primary agent. Amphotericin B is an appropriate option for initial and salvage therapy. Echinocandins are effective in salvage therapy but are not recommended as monotherapy for primary treatment. Some evidence exists to suggest an additive or synergistic effect of treatment with polyenes and/or azoles with echinocandins; however, variable test designs and conflicting results have led to uncertainty as to how to interpret these findings. Early initiation of treatment is warranted while diagnostic evaluation is conducted. When feasible, eliminating or reducing the dose of immunosuppressive agents is advisable. Surgery for aspergillosis should be considered for local disease that is easily accessible to debridement.4In the present case, the patient developed acute, increased respiratory distress 9 days after admission secondary to profound plaque and debris nearly completely obstructing the glottis. After surgical debridement, a tracheostomy was placed for pulmonary hygiene. The patient was discharged 1 month after admission and treated with antifungal therapy for a total of 3 months. Results of repeat flexible laryngoscopy at 10 weeks from initial presentation revealed complete resolution of plaque and debris, and she was decannulated.This case demonstrates pseudomembranous and obstructive aspergillus tracheobronchitis in a solid organ transplant recipient. As the number of immunocompromised patients continues to rise, it is vital that the otolaryngology community remains attuned to opportunistic infections affecting the upper aerodigestive tract.
General
A woman in her 60s with a history of end-stage renal disease who had a kidney transplant 9 months previously presented with progressive cough, dyspnea, and hoarseness. Her history also included chronic gastroesophageal reflux disease and a 14-pack-year smoking history. She initially presented 3 months prior with upper respiratory symptoms, which continued to worsen despite oral antibiotic therapy. Her cough became productive with white, frothy phlegm that was accompanied by fever, wheezing, hoarseness, and fatigue. Her dyspnea worsened in the supine position.Flexible laryngotracheoscopy results revealed diffuse, white-to-yellow pseudomembranous changes involving the glottis and supraglottis (Figure, A), posterior tracheal wall, and left mainstem bronchus with skip lesions in the subglottis and distal 1 cm of the trachea (Figure, B). Glottic mobility was limited in both abduction and adduction with substantial airway obstruction. Computed tomographic findings revealed posterior tracheal wall thickening with the loss of the fat plane between the trachea and esophagus, and a moderate to severe obstruction within the distal left mainstem bronchus. Biopsy results are shown in Figure, C.Laryngoscopic images demonstrate pseudomembranous plaques lining supraglottis and glottis (A) and posterior wall of the trachea (B). Supraglottic biopsy demonstrates branching organisms superficial to squamous epithelium (original magnification ×400) (C).
what is your diagnosis?
What is your diagnosis?
Nocardiosis
Squamous cell carcinoma
Posttransplant lymphoproliferative disorder
Aspergillosis
d
0
1
1
1
female
0
0
65
61-70
White
521
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2733785
An 80-year-old man with acromegaly due to a hypoenhancing pituitary microadenoma opted for transsphenoidal resection. Preoperative magnetic resonance imaging examination of the sella revealed a left-sided pituitary microadenoma (Figure, A) and lobulated polypoid soft tissue masses in the superior nasal cavities arising from the olfactory clefts. These lesions demonstrated avid post–contrast enhancement with small areas of cystic change and heterogeneously hyperintense T2 signal. There was mild widening of the olfactory clefts, particularly on the left side (Figure, B). The intervening nasal septum was intact, and there was no intracranial extension. The patient did not have any noteworthy rhinological symptoms on review. He later underwent nasal endoscopy, and results showed tan-colored polypoid lesions emanating from the olfactory clefts of both nasal cavities (Figure, C). Biopsy findings revealed submucosal proliferation of seromucinous and respiratory epithelial glands (Figure, D).A, An incidental polypoid-enhancing mass in the nasal cavity demonstrating avid post–contrast enhancement (yellow arrowheads) was observed on the magnetic resonance imaging examination. The white arrowhead points to the pituitary adenoma. B, Widening and remodeling of the olfactory clefts (yellow arrowheads) was also observed on the magnetic resonance imaging examination. C, The incidental polypoid-enhancing mass in the nasal cavity (yellow arrowheads) as seen on nasal endoscopic analysis. D, Results of the nasal cavity mass biopsy (original magnification ×100) revealed submucosal proliferation of bland-appearing seromucinous and respiratory epithelial glands. What Is Your Diagnosis?
Low-grade sinonasal adenocarcinoma
Inflammatory nasal polyps
Inverted papilloma
Nasal cavity hamartoma
D. Nasal cavity hamartoma
D
Nasal cavity hamartoma
There are 2 types of sinonasal hamartomas identified on histopathologic analysis: respiratory epithelial adenomatoid hamartoma (REAH) and seromucinous hamartoma. The latter of these is included as a new entity in the fourth edition of WHO Classification of Head and Neck Tumours.1 These lesions are described as respiratory epithelial lesions in the current classification. It is difficult to differentiate the 2 types using histopathologic, endoscopic, or imaging analysis, and they are believed to represent a spectrum. Recently, a possible new subtype containing olfactory neuroepithelial cells was also described.2 It is still unclear whether these sinonasal lesions represent nonneoplastic lesions (hamartomas) or true benign neoplasms. Ozolek and colleagues3 demonstrated significant allelic loss in REAH, which raises the possibility of these being true neoplasms.On microscopic examination, the hamartomas are covered with ciliated respiratory epithelium, which shows invagination and submucosal glandular proliferation.4,5 The submucosal proliferating glands are predominantly multilayered ciliated respiratory epithelium in REAH or a single layer of cuboidal epithelium in seromucinous hamartoma, though a mixed picture is not uncommon, as seen in the present patient. A myxoid to fibrous stroma is often infiltrated with lymphocytes and plasma cells. Eosinophilic infiltration is absent in contrast with inflammatory polyps. Hyalinization of the basement membrane of the invaginated proliferating glands is common in REAH. In cases where there is coexistent osseous or cartilaginous metaplasia present, the term chondroosseous respiratory epithelial hamartoma may be applied. Chondroosseous respiratory epithelial hamartoma is a distinct entity from the nasal chondromesenchymal hamartoma, which occurs primarily in infants and may be characterized by locally aggressive involvement of the paranasal sinuses, nasal cavity, and orbits.1 Differentiating REAH from low-grade adenocarcinomas is very important and can be a challenge to the pathologist in a limited biopsy. Immunohistochemical stains also offer limited help.6Seromucinous hamartoma is rare and less common than REAH, and only a handful of cases exist in literature.5 Both can coexist with inflammatory sinonasal polyps. This frequently leads to their underrecognition. Typically, they are found in the middle-to-elderly age group. Nasal or sinus obstruction, rhinorrhea, dysosmia, or epistaxis can be the presenting symptoms, but they can be asymptomatic and picked up incidentally, as in the present case. They can occur anywhere in the nasal cavities or nasopharynx, but the posterior nasal septum and olfactory clefts are characteristic locations.5,7 On computed tomographic analysis, widening of the olfactory cleft and absence of aggressive features such as bony erosions or destruction are considered typical.8 REAH can be either unilateral or bilateral; as in this and previous cases, the bilaterality of a benign-appearing lesion may be a clue to the diagnosis.9 Magnetic resonance imaging provides better tissue characterization of the hamartomas, which are typically T2 hyperintense, polypoid lesions showing post–contrast enhancement. It is important to remember that the presence of post–contrast enhancement in particular can lead to a misdiagnosis of carcinoma.Sinonasal hamartomas are managed by endoscopic resection. These lesions rarely recur, and malignant transformation has not been reported.5 This necessitates a correct diagnosis for appropriate management. From an imaging point of view, the characteristic location, olfactory cleft widening, lack of aggressive features, intact cribriform plate, T2 brightness, and bilaterality are potential clues, but these lesions can still pose a challenge to the untrained eye.In the present case, additional and longer-term testing is needed to determine the endocrine status and outcome regarding the patient’s pituitary adenoma resection. The nasal cavity REAH was subtotally resected and stable at a 3-month postoperative magnetic resonance imaging examination.
General
An 80-year-old man with acromegaly due to a hypoenhancing pituitary microadenoma opted for transsphenoidal resection. Preoperative magnetic resonance imaging examination of the sella revealed a left-sided pituitary microadenoma (Figure, A) and lobulated polypoid soft tissue masses in the superior nasal cavities arising from the olfactory clefts. These lesions demonstrated avid post–contrast enhancement with small areas of cystic change and heterogeneously hyperintense T2 signal. There was mild widening of the olfactory clefts, particularly on the left side (Figure, B). The intervening nasal septum was intact, and there was no intracranial extension. The patient did not have any noteworthy rhinological symptoms on review. He later underwent nasal endoscopy, and results showed tan-colored polypoid lesions emanating from the olfactory clefts of both nasal cavities (Figure, C). Biopsy findings revealed submucosal proliferation of seromucinous and respiratory epithelial glands (Figure, D).A, An incidental polypoid-enhancing mass in the nasal cavity demonstrating avid post–contrast enhancement (yellow arrowheads) was observed on the magnetic resonance imaging examination. The white arrowhead points to the pituitary adenoma. B, Widening and remodeling of the olfactory clefts (yellow arrowheads) was also observed on the magnetic resonance imaging examination. C, The incidental polypoid-enhancing mass in the nasal cavity (yellow arrowheads) as seen on nasal endoscopic analysis. D, Results of the nasal cavity mass biopsy (original magnification ×100) revealed submucosal proliferation of bland-appearing seromucinous and respiratory epithelial glands.
what is your diagnosis?
What is your diagnosis?
Low-grade sinonasal adenocarcinoma
Nasal cavity hamartoma
Inverted papilloma
Inflammatory nasal polyps
b
1
1
1
1
male
0
0
80
71-80
White
522
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2732284
A 25-year-old woman was referred to the neuro-ophthalmology clinic because of a 6-month history of intermittent diplopia. The double vision occurred every day, multiple times throughout the day, and was provoked after looking to the right. There was no eye pain, headache, ptosis, difficulties swallowing, change in voice, or any other ocular or neurological symptoms.She had no history of strabismus, eye patching, or ocular surgery. Her medical history was notable for a growth hormone–producing pituitary macroadenoma for which she underwent surgical resection 2.5 years prior to presentation, followed by γ-knife radiosurgery (50 Gy) 6 months after the initial surgical procedure. She also had a history of congenital hip dysplasia. Medications included cabergoline, levothyroxine, ethinyl estradiol/drospirenone, and pasireotide.Her visual acuity was 20/20 OU. Her pupils were equal in size with no relative afferent pupillary defect. Automated perimetry was full in both eyes. The anterior and posterior segments were normal in both eyes. There was no proptosis or ptosis. On initial examination, the eye movements were full, with no ocular deviation in primary, right, or lateral gazes. However, on returning to a primary gaze from a sustained right-gaze position, a large angle exotropia was present, and on attempted left gaze, there was a limitation of adduction of the right eye (Figure). After 10 to 15 seconds, the exotropia resolved, and eye movements returned to normal.A, Eye positions in right gaze, and B, eye positions in primary gaze. After sustaining a right gaze position for 10 to 15 seconds (A) and then returning to primary gaze (B), there is a large-angle right exotropia. Note that the pupils are pharmacologically dilated. What Would You Do Next?
Initiate a trial of carbamazepine
Perform bilateral lateral rectus recessions
Order cranial magnetic resonance imaging
Obtain anti-acetylcholine receptor binding antibodies
Right sixth-nerve ocular neuromyotonia
A
Initiate a trial of carbamazepine
The differential diagnosis of intermittent double vision owing to an exodeviation is limited and includes third-nerve palsy, internuclear ophthalmoplegia, myasthenia gravis, orbital disease (such as myositis, tumor, or trauma), intermittent exotropia, and ocular neuromyotonia.1 In this patient, the findings on voluntary eye movements provided valuable information and narrowed the diagnosis to only 1 entity, a right sixth-nerve ocular neuromyotonia. Intermittent exotropia alone can occur from any of the entities mentioned, but only in a case of ocular neuromyotonia will there be an intermittent ocular misalignment after prolonged activity of the extraocular muscle innervated by the affected ocular motor cranial nerve. Symptomatic ocular neuromyotonia is treated medically, with the first-line treatment being carbamazepine (choice A), a voltage-gated sodium-channel blocker.2,3 Bilateral lateral rectus recessions (choice B) is not indicated for ocular neuromyotonia. Although most cases of acquired diplopia in a young person warrant neuroimaging, the history of γ-knife radiosurgery and eye movement findings consistent with ocular neuromyotonia obviated the need for a magnetic resonance imaging study (choice C). Checking anti-acetylcholine receptor binding antibodies (choice D) is not necessary, because myasthenia gravis does not cause an ocular deviation and limitation of eye movements after prolonged activity of a particular extraocular muscle.Ocular neuromyotonia was first described by E. Clark, MD, in 1966 in a patient with spasm of the superior oblique muscle, but it was K. Ricker and H. G. Mertens who coined the term in 1970.1,4 It is believed to be neurogenic in origin and specifically owing to ephaptic neural transmission (ie, lateral contact between nerve fibers allowing action potentials to travel between cell membranes instead of crossing synapses).1,2 Diplopic episodes are triggered by activation of the affected ocular motor cranial nerve, resulting in continued contraction of the innervated extraocular muscle with disruption of the Sherrington law of reciprocal innervation, leading to ocular misalignment and eye movement limitation opposite to the direction of the contracted extraocular muscle.Ocular neuromyotonia is a rare condition and has been most frequently associated with prior radiation therapy.4 However, γ-knife radiosurgery, which this patient received, has been previously documented in only 6 cases, to our knowledge.3,5 Giardina et al6 reviewed 66 cases published in the literature and found a female predominance (61.1%), with a mean age of 43.9 (range, 7-74) years. Fifty-seven percent of the cases involved the third cranial nerve, 39% the sixth cranial nerve, and 9.1% the fourth cranial nerve. The most common underlying pathologic mechanism was a suprasellar mass (60%), followed by an idiopathic origin (19.7%) and an association with thyroid dysfunction (4.5%). Of the suprasellar mass cases, 76.9% had therapeutic radiation exposure. Other less common conditions reported with ocular neuromyotonia included cranial trauma, thorium toxicity, Paget disease, nasopharyngeal carcinoma, central nervous system demyelination, botulinum neurotoxin injection, vitamin D deficiency, stroke, and cataract surgery.6,7The initial evaluation of any patient suspected of ocular neuromyotonia should begin with a detailed history specifically of prior radiation therapy. Misinterpreting the clinical findings can lead to unnecessary investigations, avoidable medical expenses, and inappropriate surgical intervention.The patient was prescribed 200 mg of carbamazepine twice a day, with the option to increase the dosage by 200 mg each week, going no higher than 1200 mg per day depending on the frequency of the double vision. Since starting the medication, she reported a decrease in the number of double-vision episodes.
Ophthalmology
A 25-year-old woman was referred to the neuro-ophthalmology clinic because of a 6-month history of intermittent diplopia. The double vision occurred every day, multiple times throughout the day, and was provoked after looking to the right. There was no eye pain, headache, ptosis, difficulties swallowing, change in voice, or any other ocular or neurological symptoms.She had no history of strabismus, eye patching, or ocular surgery. Her medical history was notable for a growth hormone–producing pituitary macroadenoma for which she underwent surgical resection 2.5 years prior to presentation, followed by γ-knife radiosurgery (50 Gy) 6 months after the initial surgical procedure. She also had a history of congenital hip dysplasia. Medications included cabergoline, levothyroxine, ethinyl estradiol/drospirenone, and pasireotide.Her visual acuity was 20/20 OU. Her pupils were equal in size with no relative afferent pupillary defect. Automated perimetry was full in both eyes. The anterior and posterior segments were normal in both eyes. There was no proptosis or ptosis. On initial examination, the eye movements were full, with no ocular deviation in primary, right, or lateral gazes. However, on returning to a primary gaze from a sustained right-gaze position, a large angle exotropia was present, and on attempted left gaze, there was a limitation of adduction of the right eye (Figure). After 10 to 15 seconds, the exotropia resolved, and eye movements returned to normal.A, Eye positions in right gaze, and B, eye positions in primary gaze. After sustaining a right gaze position for 10 to 15 seconds (A) and then returning to primary gaze (B), there is a large-angle right exotropia. Note that the pupils are pharmacologically dilated.
what would you do next?
What would you do next?
Obtain anti-acetylcholine receptor binding antibodies
Initiate a trial of carbamazepine
Perform bilateral lateral rectus recessions
Order cranial magnetic resonance imaging
b
0
1
1
1
female
0
0
25
21-30
null
523
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2732694
A 27-year-old man presented to the clinic for evaluation of a mass in his right eye. He had an unremarkable medical and ocular history and had not sustained any ocular trauma. He described a painless, reddish mass in his lateral conjunctiva, which had been present since birth. He was asymptomatic aside from rare episodes of pink tears in his right eye, and the lesion did not bother him cosmetically. He noted no blurry vision, diplopia, or pain with eye movements. His visual acuity was 20/20 OU, and there was no proptosis, ptosis, or afferent pupillary defect. Extraocular muscle movements were intact, and his intraocular pressure was 15 mm Hg OU. There was no change in lesion size observed with the Valsalva maneuver. A slitlamp examination revealed an 8-mm, round, fluctuant, vascular, multifocal mass involving the bulbar and palpebral conjunctiva near the lateral canthus (Figure). The results of the rest of the examinations, including cornea, anterior chamber, and dilated fundus examinations, were normal in both eyes.A vascular, multifocal mass (8 mm) involving the bulbar and palpebral conjunctiva near the lateral canthus, as shown by slitlamp examination.Obtain magnetic resonance imaging of the brain and orbits What Would You Do Next?
Obtain magnetic resonance imaging of the brain and orbits
Complete an excisional biopsy of the lesion
Complete needle drainage of the lesion
Initiate systemic propranolol
Superficial conjunctival lymphatic venous malformation
A
Obtain magnetic resonance imaging of the brain and orbits
The patient underwent magnetic resonance imaging of the brain and orbits (choice A), which demonstrated a nonenhancing lobular lesion in the right anterolateral periorbit without any posterior extension. The patient was clinically diagnosed with a superficial conjunctival lymphatic venous malformation, and he elected for observation.An excisional biopsy of the lesion (choice B) would not be the preferred next step because it is important to define the extent of the lesion prior to surgical intervention. While the patient lacked orbital symptoms and the lesion appeared superficial, lymphatic venous malformations are commonly multifocal and have deep components that must be assessed. Needle drainage of the lesion (choice C) would similarly not be the most appropriate next step without defining the lesion’s extent, and it is associated with a high likelihood of recurrence.1 Systemic propranolol (choice D) would not be the preferred choice because, while propranolol is commonly used to treat capillary hemangiomas in young children and infants, its use in older children and adults with vascular and lymphatic malformations is not well described and may not be as efficacious.2Lymphatic venous malformations (LVMs), or lymphangiomas, are benign, hamartomatous, vascular lesions that may present in the orbit or adnexa. They represent no-flow or low-flow system of cystlike channels lined by endothelial cells of lymphatic or venous origin. The distinction between lymphatic and venous malformations may not be clinically relevant because many lesions demonstrate a mixture of both cell types.3 The cystic spaces may be filled with blood, as in this case. They are typically present from birth and may remain stable or gradually enlarge, but orbital lesions may present with sudden proptosis owing to spontaneous hemorrhage.4 In classic cases, these lesions tend to swell when respiratory infections occur.Lymphatic venous malformations can be divided into superficial, deep, and combined, based on location. Superficial LVMs involving only the eyelid and/or conjunctiva, as in this patient, are the least common subtype.4 Superficial LVMs tend to be of cosmetic importance only and are commonly asymptomatic. Deep and combined LVMs demonstrate orbital involvement and represent most cases. Therefore, orbital imaging is important for evaluation and management. Magnetic resonance and computed tomographic imaging demonstrate multiloculated, heterogeneous masses with cystic spaces.5 Lymphatic venous malformations may be similar in appearance to other benign lesions, such as orbital varices, capillary hemangiomas, and cavernous hemangiomas. Variation of lesion size with the Valsalva maneuver and head positioning may be useful in distinguishing LVMs from orbital varices. Capillary hemangiomas may be present from birth but would be expected to regress over time. Diagnosis of LVM can be confirmed with histopathologic analysis, but a biopsy has the potential to cause excessive bleeding6 and would not likely change the patient’s management.Asymptomatic LVMs may be observed for growth or symptoms. Symptoms depend on lesion location and may include poor cosmesis, proptosis, pain, diplopia, and headache. In case of symptoms, the most commonly described treatment for orbital and adnexal LVMs is surgical resection. However, total resection often proves difficult because the lesions are prone to bleeding,5 and there is a high rate of recurrence with surgical excision.7 Furthermore, it is often not possible to fully resect the lesion.7 Therefore, if the patient is asymptomatic, it is generally recommended to choose observation.The patient has been observed without intervention. He has remained asymptomatic and without lesion growth at the time of latest follow-up.
Ophthalmology
A 27-year-old man presented to the clinic for evaluation of a mass in his right eye. He had an unremarkable medical and ocular history and had not sustained any ocular trauma. He described a painless, reddish mass in his lateral conjunctiva, which had been present since birth. He was asymptomatic aside from rare episodes of pink tears in his right eye, and the lesion did not bother him cosmetically. He noted no blurry vision, diplopia, or pain with eye movements. His visual acuity was 20/20 OU, and there was no proptosis, ptosis, or afferent pupillary defect. Extraocular muscle movements were intact, and his intraocular pressure was 15 mm Hg OU. There was no change in lesion size observed with the Valsalva maneuver. A slitlamp examination revealed an 8-mm, round, fluctuant, vascular, multifocal mass involving the bulbar and palpebral conjunctiva near the lateral canthus (Figure). The results of the rest of the examinations, including cornea, anterior chamber, and dilated fundus examinations, were normal in both eyes.A vascular, multifocal mass (8 mm) involving the bulbar and palpebral conjunctiva near the lateral canthus, as shown by slitlamp examination.Obtain magnetic resonance imaging of the brain and orbits
what would you do next?
What would you do next?
Obtain magnetic resonance imaging of the brain and orbits
Initiate systemic propranolol
Complete needle drainage of the lesion
Complete an excisional biopsy of the lesion
a
1
0
1
1
male
0
0
27
21-30
null
524
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2733447
A boy aged 13 years was referred for choroidal mass of the right eye. Visual acutiy was 20/60 OD and 20/20 OS. Intraocular pressures and anterior segment examination findings were normal. Fundus examination revealed orange, subretinal mass with associated subretinal fluid in the posterior pole. The lesion was followed closely and throughout a series of visits, the lesion and fluid continued to progress, the visual acuity worsened to 20/200, and the tumor mass extended closer to the fovea (Figure 1). The mass measured 15 mm in basal diameter with a thickness of 6.4 mm on B-scan ultrasonography.Large choroidal vascular mass of inferotemporal right choroid. Note large choroidal mass on B-scan ultrasonography. What Would You Do Next?
Photodynamic therapy
Iodine 125 plaque brachytherapy
Anti–vascular endothelial growth factor therapy injections
External beam radiotherapy
Circumscribed choroidal hemangioma
B
Iodine 125 plaque brachytherapy
Circumscribed choroidal hemangioma (CCH) is a benign, vascular tumor considered to be congenital in nature. It is classically described as an orange-red tumor, usually in the posterior pole.1 Circumscribed choroidal hemangioma is usually diagnosed when it is causing visual disturbance from exudative retinal detachment. Circumscribed choroidal hemangioma can be treated by a variety of methods including photodynamic therapy (PDT), laser photocoagulation, cryotherapy, external beam radiotherapy, stereotactic radiotherapy, proton beam radiotherapy, episcleral plaque radiotherapy, and transpupillary thermotherapy PDT.2The results of PDT for CCH are very favorable; however, the exact treatment regimen of PDT for CCH varies depending on lesion size and location. In 2 studies of PDT in CCH, tumor thickness ranged from 1.9 mm to 5.9 mm (smaller than in this patient) and thicker lesions are less responsive to PDT or require more sessions.2,3 Diffuse choroidal hemangiomas and those associated with Sturge-Weber syndrome can achieve similar thicknesses, but it is unusual for true circumscribed lesions to be this thick. Additionally, placement of an intravenous catheter, infusion of verteporfin, and multiple sessions of laser with a slitlamp delivery system would be challenging for a 13-year-old patient (choice A).Bevacizumab has been recently reported as an option for CCH treatment.4,5 Tumor thickness was not reported, but laser photocoagulation was used as adjunctive therapy for each patient. Sagong et al5 reported a case series of 3 patients receiving intravitreal bevacizumab injections as primary treatment for CCH. The tumor thickness of these patients ranged from 1.6 mm to 2.6 mm. Of note, each of these patients were also given adjunctive PDT. Bevacizumab injections were not an option in our case owing to the need for adjunctive therapy with either PDT or thermal laser, which as described above would have proved infeasible. Additionally, a tumor thickness of 6.5 mm has not been studied with bevacizumab, to our knowledge (choice C).Schilling et al6 published a case series of 36 eyes treated with external beam radiation for the treatment of CCH. The age of the patients ranged from 22 to 66 years. Of the tumors that received external beam radiotherapy without laser photocoagulation, none showed tumor regression on ultrasonography. In addition, Harbron et al7 published a series of 7257 children receiving external beam radiotherapy, of which 42 secondary malignant neoplasms were diagnosed. Although external beam radiotherapy may have been effective in this case, the risk of secondary malignant neoplasm outweighed benefit (choice D).Iodine 125 plaque brachytherapy is successfully used to treat many tumors of the retina and choroid. A case study of 8 patients with CCH who underwent episcleral plaque brachytherapy was reported by López-Caballero et al.8 The tumor thickness of the patients ranged from 2.8 mm to 6.5 mm, and all tumors were successfully treated with episcleral plaque brachytherapy targeting 29 Gy to the apex. With the ability to control our surroundings with anesthesia and need for a definitive, we elected plaque brachytherapy (choice B).This patient was treated with iodine 125 episcleral brachytherapy with apex dose of 35 Gy. At 6 months after plaque radiotherapy, his visual acuity improved to 20/100. His tumor apex decreased from 6.5 mm to 4.2 mm. His vision is stable, and he has no early signs of cataract or radiation retinopathy (Figure 2). We will continue to monitor as in our experience, radiation changes occur around 18 months after plaque radiotherapy.Regressed choroidal mass after iodine 125 plaque brachytherapy. Note reattachment of fovea and resolution of subretinal fluid.
Ophthalmology
A boy aged 13 years was referred for choroidal mass of the right eye. Visual acutiy was 20/60 OD and 20/20 OS. Intraocular pressures and anterior segment examination findings were normal. Fundus examination revealed orange, subretinal mass with associated subretinal fluid in the posterior pole. The lesion was followed closely and throughout a series of visits, the lesion and fluid continued to progress, the visual acuity worsened to 20/200, and the tumor mass extended closer to the fovea (Figure 1). The mass measured 15 mm in basal diameter with a thickness of 6.4 mm on B-scan ultrasonography.Large choroidal vascular mass of inferotemporal right choroid. Note large choroidal mass on B-scan ultrasonography.
what would you do next?
What would you do next?
Iodine 125 plaque brachytherapy
External beam radiotherapy
Photodynamic therapy
Anti–vascular endothelial growth factor therapy injections
a
0
1
1
1
male
0
0
13
11-20
null
525
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2734132
A 52-year-old man was referred to the neuro-ophthalmology clinic for evaluation of visual field loss. He had exotropia and amblyopia of the left eye. He was diagnosed as having glaucoma 6 months prior to presentation owing to optic nerve cupping and visual field deficits on automated perimetry and was taking latanoprost. His medical history was significant for pyruvate dehydrogenase deficiency, and his family history was notable for glaucoma in his grandmother. He was being treated by the neurology department for his pyruvate dehydrogenase deficiency with thiamine, vitamin C, and oxaloacetate. His systemic symptoms included hyperhidrosis and episodes of hand tremors, ataxia, and dysarthria that occur every 2 to 3 years.Best-corrected visual acuity was 20/40 −2 OD and 20/100 +2 OS, with a relative afferent pupillary defect in the left eye. Intraocular pressure (IOP) was 16 mm Hg OU, and pachymetry was 611 μM OD and 592 μM OS (normal mean [SD], 542.4 [33.5] μM). He identified 8/8 OD and 3/8 OS Ishihara color plates. He had an exotropia with full range of motion in both eyes. His nuclear sclerotic cataracts were not consistent with visual acuity in both eyes; his anterior segment examination was otherwise within normal limits. His dilated examination was normal except for cupping and pallor in both eyes with a supratemporal optic nerve pit in the right eye. The optic discs and automated perimetry are shown in the Figure. Optical coherence tomography (OCT) of the retinal nerve fiber layer showed a mean thickness of 64 μM OD and 47 μM OS (normal mean [SD], 97.3 [9.6] μM), and OCT of the macula showed perifoveal thinning with normal foveal thickness in both eyes.A and B, Fundus photographs show cupping with optic nerve pallor in both eyes and a supratemporal optic nerve pit in the right eye (arrowhead). C and D, Automated perimetry demonstrating temporal hemianopia respecting the vertical midline in the left eye with cecocentral scotoma in the right eye, consistent with a junctional scotoma.Repeated visual fields with IOP check in 1 month What Would You Do Next?
Observation
Magnetic resonance imaging of the brain/orbits
Referral to endocrinology
Repeated visual fields with IOP check in 1 month
Chiasmal syndrome secondary to pyruvate dehydrogenase deficiency
B
Magnetic resonance imaging of the brain/orbits
Junctional scotomas are visual field defects affecting the central or cecocentral visual field in 1 eye with a contralateral temporal deficit that respects the vertical midline. These scotomas localize to the junction of the optic nerve and chiasm and are not compatible with the diagnosis of glaucoma. The most frequent cause of junctional syndrome is pituitary adenoma,1 but it has also been reported in trauma, substance abuse, infection, inflammatory disease such as sarcoidosis or multiple sclerosis, meningioma, or aneurysm.2-4 Any patient with a junctional scotoma should undergo magnetic resonance imaging of the optic chiasm.Observation and repeating visual fields in 1 month (options A and D) are not appropriate because mass lesions, including pituitary apoplexy and aneurysm, frequently cause chiasmal syndrome and would require urgent treatment. Referral to endocrinology (option B) would not be appropriate at this time. Although the most common cause of chiasmal syndrome is a pituitary adenoma that would benefit from endocrinology consult, it would be inappropriate to make this referral without further evidence of a pituitary lesion.The pyruvate dehydrogenase complex is found in the mitochondria and is responsible for converting pyruvate to acetyl coenzyme A, thus producing the substrate for the tricarboxylic acid cycle and aerobic respiration. Deficits in pyruvate dehydrogenase lead to a decrease in aerobic respiration causing lactic acidosis and an increase in plasma concentrations of pyruvate. Owing to the high metabolic demand of the central nervous system tissue, neurologic manifestations of pyruvate dehydrogenase deficiency include peripheral neuropathy, ataxia, and seizures.5 Pyruvate dehydrogenase deficiency is a rare cause of optic neuropathy, and to our knowledge, there have only been 2 cases of optic neuropathy reported owing to pyruvate dehydrogenase deficiency.6,7 Similar to our case, both of these patients had ocular motility deficits with stable vision for many years before their acute visual loss owing to optic neuropathy. Owing to the rarity of patients surviving with pyruvate dehydrogenase deficiency to adulthood, there are no treatments that have been proven in prospective trials, but case reports have suggested that there are a subset of patients who respond to thiamine supplementation,8 activators of pyruvate dehydrogenase such as dichloroacetate,9 or a ketogenic diet.10Magnetic resonance imaging of the brain did not reveal abnormalities of the optic nerves or chiasm but did demonstrate atrophy of the cerebellum. Repeated serologic testing demonstrated increased levels of lactate and pyruvate. After 5 years of follow-up, his visual acuity remained stable at 20/30 OD and 20/70 OS. Ishihara color plates were 7/8 OD and 3/8 OS. His maximum IOP during this period was 17 mm Hg OU while receiving latanoprost. His fundus examination remained unchanged, and his mean deviation on visual fields remained stable in both eyes between his initial visit to his last follow-up (−10.00 dB to −9.34 dB OD and −16.23 dB to −16.69 dB OS).
Ophthalmology
A 52-year-old man was referred to the neuro-ophthalmology clinic for evaluation of visual field loss. He had exotropia and amblyopia of the left eye. He was diagnosed as having glaucoma 6 months prior to presentation owing to optic nerve cupping and visual field deficits on automated perimetry and was taking latanoprost. His medical history was significant for pyruvate dehydrogenase deficiency, and his family history was notable for glaucoma in his grandmother. He was being treated by the neurology department for his pyruvate dehydrogenase deficiency with thiamine, vitamin C, and oxaloacetate. His systemic symptoms included hyperhidrosis and episodes of hand tremors, ataxia, and dysarthria that occur every 2 to 3 years.Best-corrected visual acuity was 20/40 −2 OD and 20/100 +2 OS, with a relative afferent pupillary defect in the left eye. Intraocular pressure (IOP) was 16 mm Hg OU, and pachymetry was 611 μM OD and 592 μM OS (normal mean [SD], 542.4 [33.5] μM). He identified 8/8 OD and 3/8 OS Ishihara color plates. He had an exotropia with full range of motion in both eyes. His nuclear sclerotic cataracts were not consistent with visual acuity in both eyes; his anterior segment examination was otherwise within normal limits. His dilated examination was normal except for cupping and pallor in both eyes with a supratemporal optic nerve pit in the right eye. The optic discs and automated perimetry are shown in the Figure. Optical coherence tomography (OCT) of the retinal nerve fiber layer showed a mean thickness of 64 μM OD and 47 μM OS (normal mean [SD], 97.3 [9.6] μM), and OCT of the macula showed perifoveal thinning with normal foveal thickness in both eyes.A and B, Fundus photographs show cupping with optic nerve pallor in both eyes and a supratemporal optic nerve pit in the right eye (arrowhead). C and D, Automated perimetry demonstrating temporal hemianopia respecting the vertical midline in the left eye with cecocentral scotoma in the right eye, consistent with a junctional scotoma.Repeated visual fields with IOP check in 1 month
what would you do next?
What would you do next?
Observation
Magnetic resonance imaging of the brain/orbits
Repeated visual fields with IOP check in 1 month
Referral to endocrinology
b
0
0
1
1
male
0
0
52
51-60
null
526
original
https://jamanetwork.com/journals/jama/fullarticle/2734579
A 61-year-old man with a history of hypertension presented to the emergency department with a 1-day history of fever, dyspnea, and generalized weakness. His vital signs were temperature, 38.4°C (101.1°F); blood pressure, 94/40 mm Hg; heart rate, 116/min; and respiratory rate, 26/min. He was diaphoretic and had poor dentition. A soft S1 was present on cardiac examination, and crackles were auscultated in basilar lung fields bilaterally. Three sets of blood cultures from different sites were obtained. Cardiology, cardiac surgery, and infectious disease physicians were consulted. A bedside transthoracic echocardiogram was performed, the patient was transferred to the intensive care unit, and a transesophageal echocardiogram was obtained (Figure 1).Transesophageal echocardiogram obtained from the patient in diastole on the midesophageal long-axis view. What Would You Do Next?
Order diuresis with furosemide and serial echocardiograms
Perform emergency surgery for aortic valve repair
Place an intra-aortic balloon pump
Start broad-spectrum antibiotics and observe
Severe acute aortic regurgitation secondary to infective endocarditis
B
Perform emergency surgery for aortic valve repair
The key to the correct diagnosis is the presence of fever and hemodynamic instability with physical examination findings consistent with pulmonary edema and aortic regurgitation. Imaging demonstrated acute regurgitation, consistent with acute aortic valve insufficiency. Emergency intervention is indicated because of his symptoms of heart failure.Acute aortic regurgitation causes a sudden reflux of blood from the aorta into the left ventricle during diastole. Decreased cardiac output results in compensatory tachycardia and increased myocardial oxygen demand. The incompetent aortic valve reduces coronary arterial flow, impairing myocardial perfusion. Increased left atrial pressure causes flash pulmonary edema, leading to cardiopulmonary failure.1 Symptoms of acute aortic regurgitation include diaphoresis, fatigue, and dyspnea. Examination may reveal a widened pulse pressure from decreased diastolic pressure or a soft S1 murmur caused by increased left ventricular diastolic pressure and early mitral valve closure. A faint systolic murmur may be present if aortic regurgitation is severe. Bilateral crackles from pulmonary edema may be auscultated.1Acute native-valve aortic regurgitation is most commonly caused by infective endocarditis. As many as 54% of patients presenting with acute aortic regurgitation have infective endocarditis of the aortic valve.2 Risk factors include bicuspid aortic valve, previous infective endocarditis, and poor dentition.1,3 While 80% to 90% of acute native-valve aortic regurgitation is caused by staphylococci, streptococci, and enterococci, other organisms should be considered depending on risk factors for infective endocarditis, such as Candida in intravenous drug users. When infective endocarditis is suspected, 3 sets of blood cultures from different venipuncture sites should be collected and empirical antibiotics, such as vancomycin and cefepime, should be initiated.1,3 Early consultation with infectious disease specialists is important to ensure appropriate antibiotic selection. Using the modified Duke Criteria,1,4 a diagnosis of infective endocarditis can be established.4 The initial diagnostic test is a transthoracic echocardiogram because of its accessibility. A transesophageal echocardiogram is more sensitive than a transthoracic echocardiogram, with similar specificity in assessing hemodynamics and valvular damage in infective endocarditis (sensitivity, 70% vs 96%; specificity, 90% vs 90%),1 but the transesophageal echocardiogram is more invasive.Echocardiographic features associated with a complicated hospital course include ventricular dysfunction or perivalvular abscess. Severity of aortic regurgitation is based on pressure half-time (PHT; time for regurgitation to reach half of peak velocity), vena contracta (width of the regurgitant jet), and jet to left ventricular outflow tract (LVOT) ratio (ratio of regurgitant jet width to LVOT). Severe aortic regurgitation, defined as PHT of 300 m/s, jet:LVOT ratio greater than 0.65, or vena contracta wider than 6 mm, necessitates surgery, while regurgitation not meeting these criteria may be treated with antibiotics alone.5 Other indications for surgery include heart failure, endocarditis from organisms highly resistant to antibiotics, persistent fever and positive blood cultures despite appropriate antibiotics, mobile vegetations larger than 10 mm, septic emboli, and relapsing infection.5In severe aortic regurgitation, early intervention is associated with improved in-hospital and long-term mortality, and urgent surgical consultation is required.6 Initially, afterload reduction with nitroprusside and positive inotropy with dobutamine can improve hemodynamic function. β-Blockers should be avoided because of negative inotropic and chronotropic effects.1,3,7 Intra-aortic balloon pump is contraindicated in this setting because augmenting diastolic pressure worsens regurgitation.4Considerations for valve repair vs replacement include assessment of whether repair is likely to reestablish normal valve function and the increased thromboembolic risk associated with valve replacement. Compared with valve replacement, surgical repair is associated with better 5-year patient survival (88%-65%), comparable recurrence of infective endocarditis (6%-9%), but a lower 5-year rate of freedom from reoperation (65%-90%).7,8 There are no apparent advantages between replacement valve types in terms of reinfection (mechanical, bioprosthetic, homograft), but recent studies suggest better long-term patient survival with mechanical valves.9 Considerations include the need for lifelong anticoagulation with mechanical valves and the risk of structural deterioration of bioprosthetic valves.This patient’s transesophageal echocardiogram demonstrated central jet width of 67% of the LVOT, consistent with severe acute aortic regurgitation. Vegetations were identified between the left and right coronary cusps (Figure 2). The patient was started on intravenous dobutamine and taken to the operating room for emergency aortic valve replacement with a bioprosthetic valve. Intraoperative cultures isolated methicillin-sensitive Staphylococcus aureus. Postoperatively, the patient was treated for 6 weeks with intravenous nafcillin. Three weeks postoperatively he had normal ventricular function and no evidence of aortic regurgitation. He followed up with his dentist for treatment of his poor dentition.
General
A 61-year-old man with a history of hypertension presented to the emergency department with a 1-day history of fever, dyspnea, and generalized weakness. His vital signs were temperature, 38.4°C (101.1°F); blood pressure, 94/40 mm Hg; heart rate, 116/min; and respiratory rate, 26/min. He was diaphoretic and had poor dentition. A soft S1 was present on cardiac examination, and crackles were auscultated in basilar lung fields bilaterally. Three sets of blood cultures from different sites were obtained. Cardiology, cardiac surgery, and infectious disease physicians were consulted. A bedside transthoracic echocardiogram was performed, the patient was transferred to the intensive care unit, and a transesophageal echocardiogram was obtained (Figure 1).Transesophageal echocardiogram obtained from the patient in diastole on the midesophageal long-axis view.
what would you do next?
What would you do next?
Perform emergency surgery for aortic valve repair
Order diuresis with furosemide and serial echocardiograms
Place an intra-aortic balloon pump
Start broad-spectrum antibiotics and observe
a
1
0
0
1
male
0
0
61
61-70
null
527
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2729747
A man in his 50s presented with dysphagia, arthritis, joint contractures, carpal tunnel syndrome, and infiltrated and redundant skin with sheets of 1- to 2-mm-wide waxy, firm, skin-colored papules. The patient first noticed papules on his forehead 1 year prior to presentation that spread to his face, trunk, and extremities. He had mild dysphagia and severe joint pain in his hands and wrists, with carpal tunnel of his right wrist. Additionally, his daily activities were impaired owing to decreased mobility. During clinical evaluation, he denied cough, shortness of breath, chest pain, Raynaud phenomenon, fevers, night sweats, weight loss, seizures, or confusion.Physical examination revealed diffuse skin-colored to pink papules coalescing into plaques on the entire face, with exaggerated skin folds of the forehead (Figure, A), glabella, periocular regions, and superior cutaneous lip. This was accompanied by swelling and erythema of both ears. On his extremities and trunk, he had diffuse infiltration of the skin, with exaggerated skin folds (Figure, B), decreased finger flexion, and four-fifths grip strength. His nailfold capillaries were normal. Skin sensation was intact.A, Clinical image of the exaggerated skin folds of the forehead and glabella. B, Clinical image of the wrist and hand depicting the exaggerated skin folds and decreased finger flexion. C, Hematoxylin-eosin stain image from the punch biopsy featuring fibroblastic proliferation from the papillary to the reticular dermis (original magnification ×40). D, Hematoxylin-eosin and Alcian blue stain image featuring mucin deposition in the reticular dermis.Notable laboratory results included a normal level of thyroid-stimulating hormone, macrocytic anemia, normal creatinine and calcium levels, and a monoclonal spike on a serum protein electrophoresis test. Serum free light chains revealed normal κ free light chains, λ free light chains, and a normal κ/λ ratio. Immunofixation results revealed IgG λ monoclonal gammopathy. A punch biopsy was obtained for diagnostic clarification (Figure, C and D). What Is Your Diagnosis?
Cutaneous T-cell lymphoma (mycosis fungoides [MF])
Scleromyxedema
Lepromatous leprosy
Scleroderma
B. Scleromyxedema
B
Scleromyxedema
Histologic results of the biopsy showed a fibroblastic proliferation from the papillary to the reticular dermis, with admixed lymphocytes (Figure, C). There was a notable increase in dermal mucin, highlighted with Alcian blue (Figure, D). The biopsy results were consistent with scleromyxedema. Scleromyxedema is a rare, chronic, and progressive disease that can result in death.1 It usually occurs in middle-aged individuals in association with paraproteinemia.1 Immunoglobulin G λ light chain monoclonal gammopathy is associated in almost 80% of cases, with an average 1% per-year risk of progression to multiple myeloma.2,3Typical cutaneous findings in scleromyxedema include a generalized eruption of 1- to 3-mm-wide, firm, skin-colored to pink, flat-topped or dome-shaped papules coalescing into infiltrated plaques.1-3 The coalescing plaques and surrounding skin are often indurated and may have a shiny appearance. Additional distinguishing features include deep furrows on the trunk or extremities, producing the characteristic “shar-pei sign.” As the skin becomes infiltrated, patients may develop decreased oral aperture and decreased motility of smaller joints. The “donut sign” is a classic finding referring to a central depression surrounded by an elevated rim of thickened skin in the proximal interphalangeal joints. Finally, patients with scleromyxedema often develop deep facial furrows producing the characteristic leonine facies. Histopathological findings include an increased number of fibroblasts and stromal cells, thickened collagen bundles with variable amounts of mucin deposition in the mid and deep dermis, and sparing of the papillary dermis.3,4Systemic sclerosis (scleroderma) typically begins with edematous hands in combination with Raynaud phenomenon and nailfold capillary dilation and hemorrhage—findings not seen in scleromyxedema. The edema then progresses to sclerodactyly (tight, bound-down skin with tapering of the fingers), in contrast to the mobile and redundant skin seen in scleromyxedema. Patients with systemic sclerosis develop “pinched noses” and appear younger owing to loss of facial wrinkling from sclerosis. Patients with systemic sclerosis do not develop leonine facies. Histopathologic examination of systemic sclerosis reveals increased collagen deposition without an associated increase in fibroblasts or mucin, in contrast to the increase in fibroblasts and mucin typically seen in scleromyxedema.4-6Lepromatous leprosy and MF can also present with leonine facies and should be part of the differential diagnosis.7 As with scleromyxedema, lepromatous leprosy can also present with earlobe infiltration. Distinguishing clinical findings in lepromatous leprosy include saddle nose deformity, madarosis, rhinitis, oral and nasal nonhealing ulcers, and acquired ichthyosis. Lesions of lepromatous leprosy are often accompanied by anesthesia, hypoesthesia, and paresthesia. Eye findings include lagophthalmos, corneal anesthesia, episcleritis, and/or keratitis. Left untreated, patients with lepromatous leprosy will develop characteristic red or copper-colored papules or nodules called lepromas. The histopathological findings of lepromatous leprosy include a normal epidermis, a band of normal-appearing dermis (Grenz zone), and a nodular infiltrate of foamy histiocytes (Virchow cells), plasma cells, and lymphocytes with abundant bacilli and globi (clumps of bacilli). Typically, MF presents with well-defined, erythematous, pruritic patches in UV-protected areas.8 Leonine facies is a rare presentation of MF that has been associated with folliculotropic involvement, stage IV disease, and blood involvement.9 Histopathological findings of MF include superficial perivascular and perifollicular infiltrate with folliculotropism.8 There is a variable amount of mucinous degeneration of the follicular epithelium that is more evident with special stains, including Alcian blue or colloidal iron. There is no increase in fibroblasts.The diagnosis of scleromyxedema requires the combination of characteristic clinical and histopathological findings without thyroid disease.1-3 Our patient presented with typical cutaneous and extracutaneous findings of scleromyxedema with corroborative biopsy results. Once the patient completed 6 months of intravenous immunoglobulin therapy in combination with systemic glucocorticoids, his systemic symptoms resolved, and his skin findings dramatically improved. He remained in remission 1.5-years later without further therapy.
Dermatology
A man in his 50s presented with dysphagia, arthritis, joint contractures, carpal tunnel syndrome, and infiltrated and redundant skin with sheets of 1- to 2-mm-wide waxy, firm, skin-colored papules. The patient first noticed papules on his forehead 1 year prior to presentation that spread to his face, trunk, and extremities. He had mild dysphagia and severe joint pain in his hands and wrists, with carpal tunnel of his right wrist. Additionally, his daily activities were impaired owing to decreased mobility. During clinical evaluation, he denied cough, shortness of breath, chest pain, Raynaud phenomenon, fevers, night sweats, weight loss, seizures, or confusion.Physical examination revealed diffuse skin-colored to pink papules coalescing into plaques on the entire face, with exaggerated skin folds of the forehead (Figure, A), glabella, periocular regions, and superior cutaneous lip. This was accompanied by swelling and erythema of both ears. On his extremities and trunk, he had diffuse infiltration of the skin, with exaggerated skin folds (Figure, B), decreased finger flexion, and four-fifths grip strength. His nailfold capillaries were normal. Skin sensation was intact.A, Clinical image of the exaggerated skin folds of the forehead and glabella. B, Clinical image of the wrist and hand depicting the exaggerated skin folds and decreased finger flexion. C, Hematoxylin-eosin stain image from the punch biopsy featuring fibroblastic proliferation from the papillary to the reticular dermis (original magnification ×40). D, Hematoxylin-eosin and Alcian blue stain image featuring mucin deposition in the reticular dermis.Notable laboratory results included a normal level of thyroid-stimulating hormone, macrocytic anemia, normal creatinine and calcium levels, and a monoclonal spike on a serum protein electrophoresis test. Serum free light chains revealed normal κ free light chains, λ free light chains, and a normal κ/λ ratio. Immunofixation results revealed IgG λ monoclonal gammopathy. A punch biopsy was obtained for diagnostic clarification (Figure, C and D).
what is your diagnosis?
What is your diagnosis?
Scleroderma
Scleromyxedema
Lepromatous leprosy
Cutaneous T-cell lymphoma (mycosis fungoides [MF])
b
0
1
1
1
male
0
0
55
51-60
null
528
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2730770
A man in his 60s presented with a 10-year history of a slowly progressive, asymptomatic cutaneous eruption on his left leg (Figure 1A). He had a 16-year history of diabetes mellitus that was being treated with metformin. Hematological and biochemical test results were unremarkable. There was no history of another endocrine disorder or malignant neoplasm. There was no family history of similar cutaneous findings. Physical examination revealed reddish brown hyperkeratotic papules 1 to 10 mm wide on the front and back of the left leg from knee to ankle. The rest of the physical examination findings were unremarkable. Removal of the scales caused slight bleeding. A lesional skin biopsy was performed (Figure 1B and C).A, Clinical image of the reddish brown hyperkeratotic papules on the front of the left leg. B and C, Hematoxylin-eosin–stained lesional skin biopsy specimens revealing compact hyperkeratosis, epidermal atrophy, and bandlike lymphocyte infiltrate in the superficial dermis. What Is Your Diagnosis?
Stucco keratoses
Hyperkeratosis lenticularis perstans
Disseminated superficial actinic porokeratosis
Kyrle disease
B. Hyperkeratosis lenticularis perstans
B
Hyperkeratosis lenticularis perstans
Histopathologic examination revealed focal epidermal atrophy, orthokeratotic hyperkeratosis, and a bandlike infiltrate of lymphocytes in the superficial dermis compatible with unilateral hyperkeratosis lenticularis perstans (HLP) (Figure 1B and C). He was treated with acitretin and topical tacalcitol with substantial improvement of his cutaneous lesions (Figure 2).The clinical image after treatment with systemic retinoids and vitamin D derivates demonstrates improvement of hyperkeratotic papules.Described by Flegel1 in 1958, HLP is a disease of uncertain origin, though it has been shown to have an autosomal dominant inheritance pattern with late onset.2 This rare disorder is characterized by numerous symmetric hyperkeratotic papules. Lesions are most common on the distal portion of the legs and dorsal surface of the feet. Arms, palms, and soles can be affected. Other parts of the body, including the oral mucosa, are rarely affected, and the trunk is usually spared.3 The patient has localized unilateral HLP, which is a rarely seen form of the disease4,5 The lesions are usually hyperkeratotic papules 1 to 5 mm in diameter, with an initial erythematous coloration that with time acquires brownish coloration. When the papules are dislodged, they give rise to a depression with punctate bleeding. It runs a chronic course, and lesions persist indefinitely.Histologically, there is compact hyperkeratosis with focal parakeratosis on an atrophic epidermis, with a thinned or absent stratum granulosum. In the papillary dermis, there is a band of lymphocytic infiltrate with dilatation and proliferation of superficial vessels.6 Given the rarity of cases, no standard modality exists for treatment. Reported treatments include topical tretinoin, fluorouracil cream, and vitamin D derivates.7,8 Systemic retinoids have been used with good results.9,10Differential diagnosis of HLP includes causes of acral keratosis, particularly stucco keratoses, disseminated superficial actinic porokeratosis, and Kyrle disease. Stucco keratoses are usually located on the distal portion of the extremities and clinically manifest as small, scaly white or greyish hyperkeratotic papules, but they do not bleed when scraped off, and histopathologic evaluation reveals hyperkeratosis with epidermal hyperplasia instead of atrophy of the epidermis. Disseminated superficial actinic porokeratosis is characterized by brownish hyperkeratotic papules predominantly found on the arms and legs, and symptoms appear later in adult life. The papules have a hyperkeratotic ridge at the periphery, and histologic results show the presence of cornoid lamellae. Acquired reactive perforating dermatosis (Kyrle disease) presents with keratotic papules up to 1 cm in diameter that are seen primarily on the legs and arms. It is most often seen in conjunction with diabetes or renal failure. Kyrle disease has more prominent keratotic plugs, and there is transepidermal elimination of connective tissue.This patient had an uncommon presentation of HLP given the rarity of unilateral presentation. It is important for clinicians to recognize HLP because it may mimic other more common dermatoses such as porokeratosis or stucco keratoses.
Dermatology
A man in his 60s presented with a 10-year history of a slowly progressive, asymptomatic cutaneous eruption on his left leg (Figure 1A). He had a 16-year history of diabetes mellitus that was being treated with metformin. Hematological and biochemical test results were unremarkable. There was no history of another endocrine disorder or malignant neoplasm. There was no family history of similar cutaneous findings. Physical examination revealed reddish brown hyperkeratotic papules 1 to 10 mm wide on the front and back of the left leg from knee to ankle. The rest of the physical examination findings were unremarkable. Removal of the scales caused slight bleeding. A lesional skin biopsy was performed (Figure 1B and C).A, Clinical image of the reddish brown hyperkeratotic papules on the front of the left leg. B and C, Hematoxylin-eosin–stained lesional skin biopsy specimens revealing compact hyperkeratosis, epidermal atrophy, and bandlike lymphocyte infiltrate in the superficial dermis.
what is your diagnosis?
What is your diagnosis?
Disseminated superficial actinic porokeratosis
Stucco keratoses
Hyperkeratosis lenticularis perstans
Kyrle disease
c
0
1
1
1
male
0
0
10
0-10
null
529
original
https://jamanetwork.com/journals/jamaneurology/fullarticle/2729008
A man in his 60s was evaluated in the epilepsy monitoring unit for various spells he had been having up to 5 times a day over the previous 8 months. He described transient episodes of expressive speech difficulties, paroxysmal dizziness, and involuntary nonsuppressible jerks of the right arm, which occurred without warning and were associated with mild disorientation. Video electroencephalography (EEG) captured the movements (Figure, A). They consisted of involuntary, synchronous contractions of the right face, arm, and leg that were sustained for a few seconds before muscle relaxation occurred (Figure, B and C; Video). The movements were not elicited by action or exertion. There was no epileptic abnormality that correlated with the movements on EEG, and head magnetic resonance imaging was unremarkable.A, Electroencephalogram (EEG) during the event (bipolar montage, 60-Hz filter, 7-uV/mm sensitivity). B, Patient position prior to the event. C, Patient position while experiencing an involuntary, sustained contraction of the right lower face, arm, and leg lasting almost 2 seconds before spontaneous resolution. The event took place during the time span marked with red vertical lines on the EEG and did not show a clear change from baseline rhythm. Muscle artifacts are seen prior to and during the event. What Is Your Diagnosis?
Myoclonus
Paroxysmal dyskinesia
Faciobrachial dystonic seizures
Tic disorder
C. Faciobrachial dystonic seizures
C
Faciobrachial dystonic seizures
Paroxysmal dyskinesia (choice B) is unlikely, because the phenomena are not triggered by movement or exertion, which would suggest kinesigenic or exercise-induced dyskinesia, respectively. Paroxysmal nonkinesigenic dyskinesia may occur, but the attacks would last minutes to hours rather than seconds. Furthermore, his age at onset is late for this group of genetic disorders. A tic disorder (choice D) is also unlikely, because there is no premonitory urge that is relieved by performing the tic, and the movement is not even briefly suppressible. The movement is jerky, but contractions are sustained for a few seconds, which is too prolonged for myoclonus (choice A) and more reminiscent of dystonic posturing.The phenomenology of the patient’s abnormal movement was most closely in keeping with faciobrachial dystonic seizures (FBDS), and serum testing was positive for anti–leucine-rich glioma-inactivated 1 (LGI-1) antibodies by a cell-based assay. These tonic muscle contractions classically affect the lower face and ipsilateral arm, but involvement of the leg, alternating episodes, or bilateral episodes may occur.1 They are typically brief (<3 seconds) and can occur up to hundreds of times a day.2 These movements are nearly pathognomonic for anti–LGI-1 autoimmunity and can be seen in about half of patients with this trait.3 Although EEG is frequently obscured by muscle artifact, preceding rhythmic δ activity and EEG attenuation after FBDS may be seen.4 Associated ictal features may occur with FBDS, including fear, oral automatism, and/or loss of awareness.4The LGI-1 protein is secreted by neurons in the central nervous system and forms a transsynaptic complex involved in synaptic transmission of neuronal excitability.5 The association between LGI-1 protein dysfunction and altered neuronal excitability is seen in autosomal-dominant lateral temporal–lobe epilepsy, also known as autosomal-dominant partial epilepsy with auditory features, in which an LGI-1 gene mutation is identified in one-third to half of affected individuals.6 This disorder is usually characterized by a family history of focal seizures, often involving auditory auras (humming, buzzing, ringing, or [less frequently] complex sounds or distortions) or ictal aphasia in the absence of structural abnormalities or other substantial neurological deficits.6Anti–LGI-1 IgG4 antibodies are thought to be causative of LGI-1 encephalitis, which is the second most common cause of autoimmune encephalitis after anti–N-methyl-d-aspartate receptor encephalitis and has an incidence of about 1 case per million population.5 Anti–LGI-1 autoimmunity often begins with subtle focal seizures or FBDS, which precede the onset of cognitive decline, personality changes, psychiatric symptoms, and tonic-clonic seizures typical of autoimmune limbic encephalitis.3 Other clinical manifestations include sleep disturbances, paroxysmal dizzy spells, parkinsonism, and myoclonus.7 Hyponatremia may be found in up to half of affected patients and can be a clue to the diagnosis. A malignant condition is identified in about 10% of patients and may occur in the thymus, lung, breasts, prostate, ovaries, kidneys, or alimentary tract.5 On magnetic resonance imaging, T2 hyperintensity of the medial temporal lobes is commonly found in patients with anti–LGI-1 limbic encephalitis, and T1 hyperintensity of the basal ganglia may be seen in patients with FBDS.7Recognition of FBDS is crucial to arrange for appropriate testing of anti–LGI-1 antibodies. Serum should be tested, because it may have a higher sensitivity than cerebrospinal fluid.3,7 Expedited immunotherapy in patients with FBDS is more effective than antiseizure medications to control the movements and is associated with reduced long-term disability.8 Early treatment may also prevent the evolution from FBDS to limbic encephalitis in patients with preserved cognition, further emphasizing the need to accurately diagnose these movements promptly.1 Even with immunotherapy, however, relapses can occur in one-third of patients,3 and close clinical monitoring is required so that treatment can be intensified if necessary.The patient was treated with 1-g doses of intravenous methylprednisolone for 5 days, with an oral prednisone taper that consisted of 50 mg daily for a month, followed by further decreases of 10 mg monthly. After 1 day of treatment, he had full resolution of FBDS and no further spells of any kind. On 4-month follow-up, he continued to be symptom-free.
Neurology
A man in his 60s was evaluated in the epilepsy monitoring unit for various spells he had been having up to 5 times a day over the previous 8 months. He described transient episodes of expressive speech difficulties, paroxysmal dizziness, and involuntary nonsuppressible jerks of the right arm, which occurred without warning and were associated with mild disorientation. Video electroencephalography (EEG) captured the movements (Figure, A). They consisted of involuntary, synchronous contractions of the right face, arm, and leg that were sustained for a few seconds before muscle relaxation occurred (Figure, B and C; Video). The movements were not elicited by action or exertion. There was no epileptic abnormality that correlated with the movements on EEG, and head magnetic resonance imaging was unremarkable.A, Electroencephalogram (EEG) during the event (bipolar montage, 60-Hz filter, 7-uV/mm sensitivity). B, Patient position prior to the event. C, Patient position while experiencing an involuntary, sustained contraction of the right lower face, arm, and leg lasting almost 2 seconds before spontaneous resolution. The event took place during the time span marked with red vertical lines on the EEG and did not show a clear change from baseline rhythm. Muscle artifacts are seen prior to and during the event.
what is your diagnosis?
What is your diagnosis?
Faciobrachial dystonic seizures
Paroxysmal dyskinesia
Myoclonus
Tic disorder
a
1
1
1
1
male
0
0
65
61-70
null
530
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2728251
A woman in her early 70s with a history of hypertension, Cushing disease status post–pituitary radiotherapy (1973), left sphenoid wing meningioma involving the left optic nerve and cavernous sinus status postsurgical debulking (2009), radiotherapy with concurrent temozolomide followed by intravenous bevacizumab, and a single prior episode of deep venous thrombosis presented with 2 months of eye redness and decreased vision in her right eye. She previously received a diagnosis of viral conjunctivitis at an outside institution and was treated with topical prednisolone acetate without improvement before presenting to our institution. On examination, visual acuity was 20/25 OD and hand motions OS. A relative afferent pupillary defect was present on the left in the setting of left optic atrophy from the patient’s locally invasive meningioma. Confrontational visual fields were otherwise full in the right eye. Intraocular pressures were within normal limits in both eyes. Hertel exophthalmometry was notable for 3 mm of proptosis on the right. Slitlamp examination demonstrated 3 to 4+ meibomian gland dysfunction in both eyes, but with prominent conjunctival injection in the right eye only. Dilated funduscopy revealed scattered intraretinal hemorrhages (IRHs) and cystoid macular edema (CME) in the right eye, which was also seen on optical coherence tomography (Figure 1). Vital signs, complete blood cell count, and the results of thyroid function tests were within reference ranges. Magnetic resonance imaging and magnetic resonance angiography of the brain and orbits with and without contrast showed a stable size of the meningioma and were otherwise normal.A, Color fundus photograph of the right eye showing scattered intraretinal hemorrhages and a blunted foveal light reflex. B, Optical coherence tomography of the right eye showing cystoid macular edema manifesting as foveal-involving cystoid intraretinal spaces and a small pocket of subfoveal subretinal fluid.Perform an intravitreal injection of an antivascular endothelial growth factor agent What Would You Do Next?
Perform an intravitreal injection of an antivascular endothelial growth factor agent
Start systemic corticosteroids
Obtain laboratory workup for uveitis
Order cerebral angiography
Venous stasis retinopathy in the right eye associated with a right carotid-cavernous sinus fistula
D
Order cerebral angiography
The differential diagnosis for CME with IRHs includes retinal vein occlusion, microvascular disease (eg, diabetic retinopathy, radiation retinopathy), hyperviscosity syndrome (eg, plasma cell dyscrasias), and orbital diseases resulting in impaired venous outflow. This patient initially presented elsewhere with eye redness and decreased vision in the right eye, but by the time she presented to our institution, there was greater evidence of inadequate venous outflow (ie, unilateral proptosis, corkscrew vessels extending to the limbus, blood in Schlemm canal, IRHs, and CME). These findings can classically result from venous congestion (eg, thyroid eye disease, retrobulbar mass), arteriovenous fistulization (eg, carotid cavernous sinus fistula [CCF], Sturge-Weber syndrome), or an idiopathic disease (eg, Radius-Maumenee syndrome).1 The patient’s neuroimaging was unrevealing, but given the strong clinical suspicion for CCF, a cerebral angiogram was obtained, which revealed an indirect CCF supplied by dural branches of the right cavernous internal carotid artery. Cerebral angiography is considered to be the diagnostic standard and can detect a CCF missed by less-invasive neuroimaging methods (eg, magnetic resonance angiography). In this case, use of an antivascular endothelial growth factor agent would be inappropriate because doing so would delay the diagnosis and timely treatment of the underlying CCF. Systemic therapy with corticosteroids and a laboratory workup for uveitis are also unwarranted because there was no evidence of concurrent intraocular or orbital inflammation.A CCF is an abnormal vascular communication between the internal and/or external carotid artery and cavernous sinus and may present with any combination of unilateral proptosis, engorged episcleral vessels, chemosis, diplopia, decreased vision, and even stroke.2 It may present with a bruit or pulsatile proptosis, neither of which was present in the patient described herein. The Barrow classification system categorizes CCFs as direct (type A) and indirect (types B-D).3 Direct CCFs are high-flow fistulas that typically occur following head trauma, progress rapidly, and require urgent treatment. Indirect CCFs, however, are low-flow fistulas classically seen in elderly women that tend to present more insidiously, with conjunctival injection and chemosis being the most common presenting signs.4Carotid-cavernous sinus fistulas can also present with decreased vision owing to CME and IRHs. Venous stasis retinopathy is more frequently associated with indirect CCFs and may not manifest until the venous system posterior to the cavernous sinus becomes obstructed and arterial blood flow is redirected anteriorly to the orbit, resulting in chemosis, proptosis, IRHs, CME, retinal ischemia, optic neuropathy, and/or secondary glaucoma from elevated episcleral venous pressure.5,6 When IRHs are observed, fluorescein angiography may help to determine the state of retinal perfusion and, thus, visual prognosis.Whereas direct CCFs should be treated urgently, indirect CCFs may spontaneously close and can be observed.2-4 However, indirect CCFs that fail to self-thrombose or are vision threatening (eg, severe exposure keratopathy, venous stasis retinopathy, optic neuropathy, secondary glaucoma) should be promptly treated. Endovascular embolization is considered to be the treatment of choice, with good long-term outcomes and low complication rates.7 Return of baseline visual acuity after successful CCF closure has been previously reported.8,9The patient’s CCF was embolized by neurosurgery. One week later, her vision returned to baseline, with marked improvement of her CME (Figure 2).Optical coherence tomography of the right eye showing marked improvement in cystoid macular edema within 1 week of endovascular embolization.
Ophthalmology
A woman in her early 70s with a history of hypertension, Cushing disease status post–pituitary radiotherapy (1973), left sphenoid wing meningioma involving the left optic nerve and cavernous sinus status postsurgical debulking (2009), radiotherapy with concurrent temozolomide followed by intravenous bevacizumab, and a single prior episode of deep venous thrombosis presented with 2 months of eye redness and decreased vision in her right eye. She previously received a diagnosis of viral conjunctivitis at an outside institution and was treated with topical prednisolone acetate without improvement before presenting to our institution. On examination, visual acuity was 20/25 OD and hand motions OS. A relative afferent pupillary defect was present on the left in the setting of left optic atrophy from the patient’s locally invasive meningioma. Confrontational visual fields were otherwise full in the right eye. Intraocular pressures were within normal limits in both eyes. Hertel exophthalmometry was notable for 3 mm of proptosis on the right. Slitlamp examination demonstrated 3 to 4+ meibomian gland dysfunction in both eyes, but with prominent conjunctival injection in the right eye only. Dilated funduscopy revealed scattered intraretinal hemorrhages (IRHs) and cystoid macular edema (CME) in the right eye, which was also seen on optical coherence tomography (Figure 1). Vital signs, complete blood cell count, and the results of thyroid function tests were within reference ranges. Magnetic resonance imaging and magnetic resonance angiography of the brain and orbits with and without contrast showed a stable size of the meningioma and were otherwise normal.A, Color fundus photograph of the right eye showing scattered intraretinal hemorrhages and a blunted foveal light reflex. B, Optical coherence tomography of the right eye showing cystoid macular edema manifesting as foveal-involving cystoid intraretinal spaces and a small pocket of subfoveal subretinal fluid.Perform an intravitreal injection of an antivascular endothelial growth factor agent
what would you do next?
What would you do next?
Order cerebral angiography
Obtain laboratory workup for uveitis
Start systemic corticosteroids
Perform an intravitreal injection of an antivascular endothelial growth factor agent
a
1
1
1
1
female
0
0
72
71-80
null
531
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2728252
A 75-year-old woman was referred to the ophthalmology clinic for evaluation of double vision and a droopy left upper eyelid. Over the previous 5 months, the patient had experienced gradual onset of binocular, horizontal diplopia and ptosis of the left upper eyelid. There were no neurological deficits and no pain. Her symptoms were constant, without fluctuation during the day. There was no history of trauma or prior surgery of the eye. Her medical history was notable for hypertension and type 2 diabetes. Family history was positive for hypertension in her mother, prostate cancer in her father, and ductal carcinoma in situ in her daughter. She was a current daily smoker at presentation. An ophthalmologic examination revealed a visual acuity of 20/25 OD and 20/30 OS. Pupillary, intraocular pressure, and dilated fundus examinations were unremarkable. Abduction was severely restricted in the left eye, but it did not elicit pain, and extraocular muscle movements were otherwise full. There was substantial resistance to retropulsion of the left eye. Bilateral blepharoptosis, with more severity on the left side, was noted. The margin-to-reflex distance 1 was 2 mm on the right and 0 mm on the left. A 3.5-mm left enophthalmos was noted compared with the right side, and there was hollowing of the left superior sulcus. Testing of cranial nerves V and VII were unremarkable.Computed tomographic scan of the orbits with contrast What Would You Do Next?
Computed tomographic scan of the orbits with contrast
Myasthenia gravis testing
Referral for strabismus surgery
Thyroid function testing
Infiltrating lobular breast carcinoma
A
Computed tomographic scan of the orbits with contrast
While the differential diagnosis of strabismus is broad, the findings of enophthalmos and resistance to retropulsion of the globe, in the context of the patient’s age, sex, and family history of cancer, should raise concern for a malignant orbital causative mechanism. Myasthenia gravis testing (choice B) would not be preferred because the patient lacks the characteristic fluctuating symptoms. Thyroid function testing (choice D) would not be preferred because thyroid dysfunction would be expected to have presented earlier in life. Strabismus surgery (choice C) would not address the underlying cause of strabismus. Furthermore, pursuing laboratory testing or strabismus surgery may delay the diagnosis of a malignant and potentially fatal condition.Orbital computed tomography and magnetic resonance imaging with contrast (choice A) were performed and demonstrated a diffuse infiltrative lesion within the left orbit, including the extraocular muscles (Figure). The patient underwent a left orbitotomy with exploration and biopsy of the mass, which produced pathological findings consistent with infiltrating lobular breast carcinoma. Although a breast examination revealed fullness of the left breast near the areola, a diagnostic mammogram, breast ultrasonography, and magnetic resonance imaging of the breast were unremarkable. Despite negative imaging, she underwent a core-needle biopsy of the left breast in the area of fullness, which confirmed the diagnosis of an infiltrating, well-differentiated breast carcinoma with lobular features.Fat-suppressed, T1-weighted, post-contrast magnetic resonance image of the orbits demonstrating a diffuse infiltration of the left intraconal and extraconal orbital contents, not easily distinguished from and isointense to the extraocular muscles (arrowheads). There is gross enophthalmos of the left globe compared with the right.Breast carcinoma is the most common primary tumor to metastasize to the orbits.1 Orbital breast carcinoma metastases characteristically involve infiltration of the extraocular muscles and orbital fat. While this tumor classically involves cicatrization that results in enophthalmos, as in this case, some patients may present with exophthalmos.2 Typically, orbital metastasis will occur late in the course of breast cancer, with a mean latency between the diagnosis of primary and metastatic orbital disease of 5 years,3 with a maximum latency of as many as 25 years.4 Therefore, it is quite rare for an orbital metastasis to be the presenting finding. In such cases, breast examination or imaging will typically be revealing of the primary tumor.4-6 The presented case was unique in the absence of radiographic evidence of a primary malignant condition.In most cases of orbital breast carcinoma metastasis, personal medical history will be revealing of the primary tumor, and biopsy of the orbital lesion may be deferred.7 However, absence of established diagnosis of a primary malignant condition does not rule out the possibility of metastasis, and imaging is essential in such cases. Imaging may provide diagnostic clues,8 and a biopsy will provide definitive diagnosis and guide further testing or treatment.Treatment of orbital breast carcinoma typically involves systemic chemotherapy. External-beam radiotherapy is a generally safe and effective adjunctive treatment option,2,9 but it must be weighed against the risks of radiation to the orbit. Surgical excision is generally contraindicated, because this would not cure the underlying disease and can cause considerable morbidity, depending on the extent of involvement. However, it may be considered in case of severe symptoms refractory to other palliative efforts.2This case demonstrates the possibility that an ophthalmologist may be the first to diagnose a metastatic malignant condition such as breast carcinoma. The ophthalmologist should be aware of this possibility, and metastasis should remain on the differential in the appropriate clinical context, even in the absence of a personal cancer history in the patient. Imaging with thin-cut orbital sections should be considered in patients with strabismus but also those with enophthalmos and resistance to retropulsion.The patient is currently undergoing chemotherapy. Radiotherapy to the orbit is deferred unless she develops worsening visual symptoms or progression.
Ophthalmology
A 75-year-old woman was referred to the ophthalmology clinic for evaluation of double vision and a droopy left upper eyelid. Over the previous 5 months, the patient had experienced gradual onset of binocular, horizontal diplopia and ptosis of the left upper eyelid. There were no neurological deficits and no pain. Her symptoms were constant, without fluctuation during the day. There was no history of trauma or prior surgery of the eye. Her medical history was notable for hypertension and type 2 diabetes. Family history was positive for hypertension in her mother, prostate cancer in her father, and ductal carcinoma in situ in her daughter. She was a current daily smoker at presentation. An ophthalmologic examination revealed a visual acuity of 20/25 OD and 20/30 OS. Pupillary, intraocular pressure, and dilated fundus examinations were unremarkable. Abduction was severely restricted in the left eye, but it did not elicit pain, and extraocular muscle movements were otherwise full. There was substantial resistance to retropulsion of the left eye. Bilateral blepharoptosis, with more severity on the left side, was noted. The margin-to-reflex distance 1 was 2 mm on the right and 0 mm on the left. A 3.5-mm left enophthalmos was noted compared with the right side, and there was hollowing of the left superior sulcus. Testing of cranial nerves V and VII were unremarkable.Computed tomographic scan of the orbits with contrast
what would you do next?
What would you do next?
Referral for strabismus surgery
Myasthenia gravis testing
Computed tomographic scan of the orbits with contrast
Thyroid function testing
c
0
1
1
0
female
0
0
75
71-80
null
532
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2729895
A 74-year-old man was referred to the ocular oncology clinic for a right eye corneal mass. He had a history of herpetic keratitis and cataract surgery 2 years prior, both in the right eye, complicated by postoperative bullous keratopathy without treatment. The patient noted a corneal scar after the cataract surgery that gradually thickened over the ensuing 6 months. In our office, his examination in the right eye was notable for hand motion visual acuity and a pearly white, raised, 10 × 10-mm, gelatinous, corneal lesion with both intrinsic and feeder vessels (Figure 1A). There was no view of the anterior segment. Ultrasound biomicroscopy showed a hyperechoic opacity on the surface of the cornea, with maximal thickness of 1.5 mm. A cleft was noted between the lesion and the cornea (Figure 1B). There was no extension posteriorly into the anterior chamber, iris, or ciliary body.A, Slitlamp external photograph of right-eye corneal lesion showing prominent elevation and both intrinsic and feeder vessels. B, Ultrasound biomicroscopy showing hyperechoic anterior corneal lesion with hypoechoic cleft labeled centrally. The lesion did not involve any other adjacent structures. What Would You Do Next?
Local therapy with 5-fluorouracil
Incisional biopsy
Excisional biopsy
Observe
Corneal keloid
C
Excisional biopsy
Given the cleft seen on biomicroscopy (Figure 1B), a #57 ultrasharp blade was used to gently but completely excise the lesion. Pathology results were positive for corneal keloid (hyperplastic corneal pannus),1 a rare corneal lesion usually occurring during the first 3 decades of life.2 No link has been found with cutaneous keloids, nor is this lesion more common among African American or Asian individuals. While bilateral cases are typically associated with systemic disorders such as Lowe syndrome and Rubenstein-Taybi syndrome, isolated unilateral cases are usually secondary to corneal trauma, surgery, keratouveitis, or infection.3-8Ultrasound biomicroscopy is helpful in defining the extent of these hyperechoic lesions and involvement of adjacent ocular structures, which is uncommon (Figure 1B). The differential diagnosis of corneal keloid includes (in order of relevance in this case) squamous cell carcinoma, limbal dermoid, corneal myxoma, Salzmann nodular degeneration, fibrous histiocytoma, anterior staphyloma, metabolic disease, and others. While diagnosis can be made based on clinical presentation, the gold standard is histopathologic examination. In contrast to true cutaneous keloids, the corneal variant is marked by less prominent hyalinized collagen bundles, disruption of the Bowman membrane, and increased relative cellularity and vascularity (Figure 2).9 This histologic distinction supports abandoning the term keloid for hyperplastic pannus.1 Pathogenesis is still debatable, but evidence suggests that corneal stromal keratocytes undergo transformation into fibroblasts and myofibroblasts.10 Treatment for corneal keloids (excision) is generally reserved for those causing significant visual dysfunction.The excisional biopsy showed irregular and vascularized connective tissue with a few bands of hyalinized collagen at the arrowheads. As previously stated, this case requires excisional biopsy (choice C) and histopathologic examination. The 5-fluorouracil choice (choice A) would not be a reasonable next step in this unusual case in the absence of histopathologic confirmation of ocular surface squamous neoplasia. Incisional biopsy (choice B) would not address the patient’s severely limited vision, and incomplete excision is associated with keloid recurrence.7 Observation (choice D) alone does not address the patient’s poor vision and would not be appropriate given the possibility of neoplasia.At postoperative week 2, the patient still had counting fingers visual acuity that persisted at week 12. His examination was notable for a 360° pannus and central corneal haze. At postoperative week 16, he had hand motion visual acuity at 3 ft, an irregular corneal surface without epithelial defect, superficial neovascularization across the corneal surface, and diffuse stromal haze and edema. He is subjectively doing well but will be followed up for the development of recurrence.
Ophthalmology
A 74-year-old man was referred to the ocular oncology clinic for a right eye corneal mass. He had a history of herpetic keratitis and cataract surgery 2 years prior, both in the right eye, complicated by postoperative bullous keratopathy without treatment. The patient noted a corneal scar after the cataract surgery that gradually thickened over the ensuing 6 months. In our office, his examination in the right eye was notable for hand motion visual acuity and a pearly white, raised, 10 × 10-mm, gelatinous, corneal lesion with both intrinsic and feeder vessels (Figure 1A). There was no view of the anterior segment. Ultrasound biomicroscopy showed a hyperechoic opacity on the surface of the cornea, with maximal thickness of 1.5 mm. A cleft was noted between the lesion and the cornea (Figure 1B). There was no extension posteriorly into the anterior chamber, iris, or ciliary body.A, Slitlamp external photograph of right-eye corneal lesion showing prominent elevation and both intrinsic and feeder vessels. B, Ultrasound biomicroscopy showing hyperechoic anterior corneal lesion with hypoechoic cleft labeled centrally. The lesion did not involve any other adjacent structures.
what would you do next?
What would you do next?
Excisional biopsy
Incisional biopsy
Local therapy with 5-fluorouracil
Observe
a
1
0
1
1
male
0
0
74
71-80
White
533
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2723577
A 31-year-old gravida 1, para 1 woman with a history of Wolff-Parkinson-White disease and recently diagnosed stage II Hodgkin lymphoma presented with a pruritic rash. She was diagnosed with Hodgkin lymphoma at 35 weeks’ gestation, and 1 week later she underwent cesarean delivery. At 3 weeks’ postpartum, she began chemotherapy (cycle 1, day 1) with adriamycin, bleomycin, vinblastine, and dacarbazine (ABVD). On day 15 of cycle 1, she presented with linear, hyperpigmented streaks and welts on her arms, back, and legs. Intense, diffuse pruritus had developed within 24 hours of the patient’s first day of treatment, and her symptoms were poorly responsive to antihistamines and topical steroids. Pruritus was followed by the appearance of dark scratch marks as demonstrated in the Figure. Laboratory test results revealed a white blood cell count of 1.6 ×103 cells/mm3, with an absolute neutrophil count of 500 cells/mm3 and a total bilirubin of 0.3 mg/dL (to convert to μmol/L, multiply by 17.104), which was unchanged from the baseline total bilirubin.Clinical photograph of the patient’s back shows scratch marks that appeared after diffuse pruritus.Prior to the diagnosis of Hodgkin lymphoma, she was receiving routine prenatal care, and the only complication was pruritus without a rash that developed during the first trimester. She was evaluated for right supraclavicular adenopathy that was unresponsive to antibiotics during the third trimester. Findings from a lymph node biopsy were consistent with classic Hodgkin lymphoma, nodular sclerosis subtype. Staging imaging revealed bilateral supraclavicular jugular chain, paratracheal and mediastinal lymphadenopathy, and the erythrocyte sedimentation rate was 67 mm/h. She was diagnosed with stage II, unfavorable-risk Hodgkin lymphoma. What Is Your Diagnosis?
Bleomycin toxic effects
Paraneoplastic skin disease
Polymorphic eruption of pregnancy
Vinblastine toxic effects
A. Bleomycin toxic effects
A
Bleomycin toxic effects
This patient developed bleomycin flagellate dermatitis, a cutaneous reaction to bleomycin characterized by the appearance of pruritic, whiplike streaks of hyperpigmentation, predominantly affecting the trunk and extremities. Bleomycin is a sulfur-containing, antineoplastic agent that blocks the G2 phase of the cell cycle through inhibition of thymidine integration into replicating DNA, degradation of single- and double-stranded DNA, and cleavage of cellular RNA.1 Bleomycin distributes widely throughout the body and is inactivated in every organ except the skin and lungs owing to the absence of the hydrolase enzyme responsible for the drug’s metabolism.2 Absence of this enzyme permits drug accumulation, which can lead to pulmonary fibrosis and various adverse cutaneous manifestations associated with bleomycin.1Flagellate dermatitis is primarily a clinical diagnosis because there are no pathognomonic histologic features. Early histologic findings are consistent with a fixed-drug reaction, and in later stages, reflect a postinflammatory reaction.3 Time of onset varies, occurring within hours or up to 9 weeks following exposure, and the reported incidence of bleomycin flagellate dermatitis is between 8% and 22%.4 Symptoms are generally well controlled with antihistamines, corticosteroids, and withdrawal of bleomycin. Early descriptions of bleomycin flagellate dermatitis documented an apparent dose-dependent occurrence in 8% to 66% of patients; however, the flagellate exanthem has since been reported with bleomycin doses as low as 5 mg with no predictable patient demographic.2-4 Nonetheless, Ziemer et al4 observed in a meta-analysis that women receiving bleomycin specifically for lymphoproliferative disorders might be most susceptible to developing acute-onset flagellate dermatitis at lower doses of exposure.This patient initially discontinued bleomycin for 1 cycle, but she tolerated successful reintroduction of bleomycin with an adjuvant steroid regimen. She remained in remission more than 2 years after chemotherapy completion and had no evidence of bleomycin pulmonary toxic effects. Until recently, there have been no definitive guidelines regarding whether to withdraw bleomycin or rechallenge following an eruption of flagellate dermatitis. The Response Adapted Therapy for Advanced Hodgkin Lymphoma (RATHL) trial randomized patients with negative findings on positron-emission tomography after 2 cycles of ABVD to continue ABVD or deescalate treatment to adriamycin, vinblastine, and dacarbazine (AVD).5 The absolute difference in the 3-year progression-free survival rate between the 2 groups was 1.6% (95% CI, −3.2% to 5.3%) and there was a lower incidence of pulmonary toxic effects when bleomycin was omitted.5 Another important phase 3 clinical trial (ECHELON-1)6 compared brentuximab vedotin plus AVD (A+AVD) with standard ABVD in previously untreated patients with stage III or IV Hodgkin lymphoma. Brentuximab vedotin plus AVD was superior, with a 4.9% lower combined risk of disease progression, death, or incomplete disease response at 2 years, and pulmonary toxic effects were documented in less than 1% of patients receiving A+AVD compared with 3% in those receiving ABVD.6 Although these studies do not specifically address flagellate dermatitis, the data suggest an impending transition in the standard of care favoring withdrawal of bleomycin treatment in patients with complications associated with the toxic effects of the drug, especially if they demonstrate favorable initial clinical response to treatment.Cutaneous toxic effects are a common reaction among chemotherapies in general, and bleomycin is associated with a variety of cutaneous manifestations, including inflammatory nodules, plaques, blistering, nail changes, and alopecia.3 Flagellate dermatitis, however, is considered a classic presentation for the toxic effects of bleomycin.4 Paraneoplastic skin involvement related to Hodgkin disease is well documented but typically encompasses nonspecific occurrence of severe pruritus with or without prurigo, acquired ichthyosis, alopecia or other eruptions that do not reflect tumor invasion. Rarely, cutaneous dissemination of malignant cells can cause more skin changes, which classically develop as painless papules, nodules, plaques, and ulcerations, that are histologically comparable to their affected lymph node counterparts and generally portend a poor prognosis.7,8 Polymorphic eruption of pregnancy could be considered as a differential diagnosis because it occurs in the immediate postpartum period; however, variable pruritic papules and plaques are the defining features of polymorphic eruption of pregnancy.9
Oncology
A 31-year-old gravida 1, para 1 woman with a history of Wolff-Parkinson-White disease and recently diagnosed stage II Hodgkin lymphoma presented with a pruritic rash. She was diagnosed with Hodgkin lymphoma at 35 weeks’ gestation, and 1 week later she underwent cesarean delivery. At 3 weeks’ postpartum, she began chemotherapy (cycle 1, day 1) with adriamycin, bleomycin, vinblastine, and dacarbazine (ABVD). On day 15 of cycle 1, she presented with linear, hyperpigmented streaks and welts on her arms, back, and legs. Intense, diffuse pruritus had developed within 24 hours of the patient’s first day of treatment, and her symptoms were poorly responsive to antihistamines and topical steroids. Pruritus was followed by the appearance of dark scratch marks as demonstrated in the Figure. Laboratory test results revealed a white blood cell count of 1.6 ×103 cells/mm3, with an absolute neutrophil count of 500 cells/mm3 and a total bilirubin of 0.3 mg/dL (to convert to μmol/L, multiply by 17.104), which was unchanged from the baseline total bilirubin.Clinical photograph of the patient’s back shows scratch marks that appeared after diffuse pruritus.Prior to the diagnosis of Hodgkin lymphoma, she was receiving routine prenatal care, and the only complication was pruritus without a rash that developed during the first trimester. She was evaluated for right supraclavicular adenopathy that was unresponsive to antibiotics during the third trimester. Findings from a lymph node biopsy were consistent with classic Hodgkin lymphoma, nodular sclerosis subtype. Staging imaging revealed bilateral supraclavicular jugular chain, paratracheal and mediastinal lymphadenopathy, and the erythrocyte sedimentation rate was 67 mm/h. She was diagnosed with stage II, unfavorable-risk Hodgkin lymphoma.
what is your diagnosis?
What is your diagnosis?
Polymorphic eruption of pregnancy
Vinblastine toxic effects
Bleomycin toxic effects
Paraneoplastic skin disease
c
0
1
1
1
female
1
0
31
31-40
White
534
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2725399
A man in his 30s presented with painless swelling of the left upper limb and nodules on the left side of the chest and abdomen. The lesions had started a month previously as red nodules on the left axilla, ipsilateral chest, and back. Twenty days later, he developed edema in the left upper limb. While being evaluated for the cutaneous lesions, the patient was diagnosed with AIDS and began antiretroviral therapy.On examination, 6 erythematous, firm, subcutaneous nodules were present on the left side of the chest, abdomen, and left upper limb, varying in size from 2 × 2 cm to 4 × 4 cm (Figure, A). There was diffuse erythema and edema on the left upper limb, leading to difficulty in limb movement (Figure, A). Multiple dilated superficial vessels were present on the posterior aspect of the left arm and ipsilateral chest wall. Left axillary lymph nodes were grossly enlarged, firm, nontender, and nonmatted, with the largest lymph node measuring 6 × 4 cm. The patient’s CD4 cell count was 141/μL (to convert to ×109/L, multiply by 0.001), and his HIV load was 504 473 copies/mL. A skin biopsy sample was obtained from the plaque and sent for histopathologic examination (Figure, B and C).A, Multiple erythematous indurated plaques and nodules confined to the left side of the chest and abdomen with axillary lymphadenopathy and upper limb edema. B, Skin biopsy sample showing infiltration with lymphoid cells and few histiocytes in the subcutaneous tissue (hematoxylin-eosin, original magnification ×400). C, On immunohistochemical analysis, these cells showed nuclear positivity for c-Myc (c-Myc, original magnification ×400). What Is Your Diagnosis?
Kaposi sarcoma
Diffuse large B-cell lymphoma
Burkitt lymphoma
Leukemia cutis
C. Burkitt lymphoma
C
Burkitt lymphoma
Skin biopsy findings revealed large atypical lymphocytes with coarse chromatin, prominent nucleoli, and a scant amount of deeply basophilic cytoplasm in the lower dermis and subcutaneous tissue. Brisk mitosis and many atypical mitotic figures were observed. The classic starry-sky pattern, an effect of benign histiocytes engulfing apoptotic tumor cells, could be seen focally in the dermis. On immunohistochemical analysis, these atypical lymphoid cells were strongly positive for CD10 and CD20 and showed nuclear positivity for c-Myc (>40%) (Figure, C), with a Ki-67 index of more than 95%. These cells were negative for CD3, CD30, terminal deoxynucleotidyl transferase (TdT), and Bcl2. The results of immunohistochemical detection of latent membrane protein 2 expression and Epstein-Barr virus (EBV) encoded RNA in situ hybridization procedures were positive. Cytogenetic analysis revealed translocation (8;14), consistent with Burkitt lymphoma (BL).There was a rapid increase in the size and number of lesions in the patient within 3 weeks. Computed tomography of the chest and abdomen showed discrete to conglomerate, multiple enlarged lymph nodes in the left axilla. The left subclavian artery was compressed by the rapidly growing axillary lymphadenopathy, leading to upper limb edema. Complete blood cell count and renal and liver function test results were normal at baseline. The patient was scheduled for chemotherapy; however, he was subsequently unavailable for follow-up.Burkitt lymphoma is a rapidly proliferating, aggressive B-cell neoplasm with a doubling time as short as 25 hours.1,2 It is classified into endemic, sporadic, and immunodeficiency-associated types. The endemic type is mostly associated with EBV and frequently affects jaw and other facial bones of African children aged 4 to 7 years.3 The sporadic and immunodeficiency-associated subtypes are more common in adults and frequently present with extranodal (particularly gastrointestinal tract) involvement.2 Sporadic types often do not show the rearrangement of the c-Myc and immunoglobulin genes, and EBV positivity is seen in less than 15% to 30% patients. Immunodeficiency type is observed in patients with HIV infection, allograft recipients, and patients with primary immunodeficiency. It is more frequent in patients with CD4 cell counts of 200/μL or higher, and EBV positivity is observed in 40% of patients or more.4 Burkitt lymphoma is composed of monomorphic, intermediate-sized, mature B cells, which are positive for CD19, CD20, CD22, CD79a, CD10, and Bcl6 but negative for CD5 and Bcl2, with abundant mitotic figures and a typical starry-sky appearance on histologic analysis.2 The genetic hallmark of BL is overexpression of c-Myc, which drives its pathogenesis.5Cutaneous presentation of BL is rare and mostly attributed to hematogenous spread, local invasion, or iatrogenic seeding of malignant cells through surgical procedures.6 This case showed evidence of regional metastases on the same side of the chest, back, and abdomen, not directly communicating with the main tumor. These, in our opinion, represent extensions of tumor via lymphatics as evidenced by predominant subcutaneous tissue involvement in the patient.1,7The main differential diagnoses of BL include other high-grade B-cell lymphomas, including diffuse large B-cell lymphoma (DLBCL). Immunohistochemical features that favor BL over DLBCL are CD10+, Bcl6+, Bcl2−, and TdT−, with a Ki-67 index close to 100%.8 Dysregulation of c-Myc can occur as a primary event in BL or as a secondary event in 5% to 15% of patients with DLBCL and 30% to 50% of patients with aggressive B-cell lymphomas unclassified with features intermediate between DLBCL and BL. Secondary c-Myc changes occur often against a background of complex karyotype and often confer aggressive clinical behavior, including poor survival rate and increased risk of relapse in the central nervous system. The adverse prognosis arises because of concurrent BCL2 (OMIM 151430) or BCL6 (OMIM 109565) gene rearrangements, which are subsequently termed double- or triple-hit lymphomas.8Leukemia cutis can be easily ruled out based on characteristic histologic and immunohistochemical findings.9 AIDS-associated Kaposi sarcoma appears more erythematous-violaceous and has characteristic histopathologic features with vascular proliferation, slitlike spaces, and solid cords and fascicles of spindle cells interspersed between vascular channels.10In conclusion, cutaneous BL is rare, and the diagnosis is mainly confirmed by histopathologic analysis; however, the rapid increase in the size of lesions in this patient was an important clinical clue.
Oncology
A man in his 30s presented with painless swelling of the left upper limb and nodules on the left side of the chest and abdomen. The lesions had started a month previously as red nodules on the left axilla, ipsilateral chest, and back. Twenty days later, he developed edema in the left upper limb. While being evaluated for the cutaneous lesions, the patient was diagnosed with AIDS and began antiretroviral therapy.On examination, 6 erythematous, firm, subcutaneous nodules were present on the left side of the chest, abdomen, and left upper limb, varying in size from 2 × 2 cm to 4 × 4 cm (Figure, A). There was diffuse erythema and edema on the left upper limb, leading to difficulty in limb movement (Figure, A). Multiple dilated superficial vessels were present on the posterior aspect of the left arm and ipsilateral chest wall. Left axillary lymph nodes were grossly enlarged, firm, nontender, and nonmatted, with the largest lymph node measuring 6 × 4 cm. The patient’s CD4 cell count was 141/μL (to convert to ×109/L, multiply by 0.001), and his HIV load was 504 473 copies/mL. A skin biopsy sample was obtained from the plaque and sent for histopathologic examination (Figure, B and C).A, Multiple erythematous indurated plaques and nodules confined to the left side of the chest and abdomen with axillary lymphadenopathy and upper limb edema. B, Skin biopsy sample showing infiltration with lymphoid cells and few histiocytes in the subcutaneous tissue (hematoxylin-eosin, original magnification ×400). C, On immunohistochemical analysis, these cells showed nuclear positivity for c-Myc (c-Myc, original magnification ×400).
what is your diagnosis?
What is your diagnosis?
Kaposi sarcoma
Leukemia cutis
Burkitt lymphoma
Diffuse large B-cell lymphoma
c
0
0
1
1
male
0
0
35
31-40
null
535
original
https://jamanetwork.com/journals/jamacardiology/fullarticle/2727334
An adult with a history of alcohol abuse was admitted to the cardiac unit with progressive shortness of breath, lower extremity edema, and tachycardia, which had developed during several days. He had no history of drug use or cardiac events. On physical examination, he had warm extremities. His body temperature was 36.8°C. His blood pressure was 100/60 mm Hg, heart rate was 130 beats/min, respiratory rate was 24 breaths/min, and pulse oximetry was 98% on ambient air. There were bibasilar rales on lung auscultation. The heart sounds were normal, and bilateral ankle edema was present. Laboratory tests revealed hyponatremia (sodium, 124 mEq/L [to convert to millimoles per liter, multiply by 1]), a compensated metabolic acidosis with a bicarbonate concentration of 14 mEq/L (to convert to millimoles per liter, multiply by 1), and a lactate concentration of 82.0 mg/dL (to convert to millimoles per liter, multiply by 0.111). The D-dimer level was 0.0003 μg/mL (to convert to nanomoles per liter, multiply by 5.476), and the pro–brain-type natriuretic peptide level was 6062 pg/mL (to convert to nanograms per liter, multiply by 1). The electrocardiogram on presentation is shown in the Figure. Echocardiography (Video 1 and Video 2) revealed a left ventricular ejection fraction of 50%. What Would You Do Next?
Start extracorporeal membrane oxygenation therapy
Administer intravenous β-blocker therapy
Administer intravenous bicarbonate therapy
Administer intravenous thiamine therapy
Shoshin (wet) beriberi
D
Administer intravenous thiamine therapy
At first glance, this case might be interpreted as an ordinary case of severe heart failure due to alcoholic cardiomyopathy. However, echocardiography revealed only a mild reduced left ventricular function, which does not fully explain all the clinical features. In particular, the metabolic acidosis with high lactate levels should trigger an explanation for an alternative diagnosis.Thiamine deficiency (vitamin B1), also known as beriberi, is a rare condition in developed countries. However, individuals prone to malnourishment, such as individuals with alcoholism, patients receiving total parenteral nutrition without adequate vitamin supplements, or individuals on a diet or after weight loss surgery, are more likely to develop thiamine deficiency.1 Thiamine plays a fundamental role in cellular metabolism, has a half-life of 18 days, and is quickly depleted. Acidosis and the inability to use the Krebs cycle are the major pathophysiologic mechanisms of the clinical manifestations of thiamine deficiency.Beriberi may present in 2 forms: a dry form with neurologic features and a wet form with the clinical features of high-output heart failure (shortness of breath, tachycardia, and edema).2,3 In rare cases, a fulminant variant termed Shoshin beriberi (translated from Japanese sho, meaning acute damage, and shin, meaning heart) may occur, which is characterized with features of cardiovascular collapse and multiorgan failure. Inappropriate management may prove fatal, and the only definitive treatment of beriberi is thiamine supplementation. Intravenous administration of thiamine markedly improves the hemodynamic parameters.4 In critically ill patients suspected of having a malnourished diet who have unexplained heart failure, lactic acidosis, or multiorgan failure, thiamine administration should be considered without delay.After excluding pulmonary embolism with computed tomography and other causes of unexplained lactate acidosis, intravenous administration of 200 mg of thiamine led to swift recovery of the patient, with normalization of hemodynamic parameters and metabolic acidosis within hours. Laboratory tests later confirmed vitamin B1 deficiency. After a few days, the patient was discharged with a prescription for oral thiamine and advised to monitor his alcohol and dietary intake.
Cardiology
An adult with a history of alcohol abuse was admitted to the cardiac unit with progressive shortness of breath, lower extremity edema, and tachycardia, which had developed during several days. He had no history of drug use or cardiac events. On physical examination, he had warm extremities. His body temperature was 36.8°C. His blood pressure was 100/60 mm Hg, heart rate was 130 beats/min, respiratory rate was 24 breaths/min, and pulse oximetry was 98% on ambient air. There were bibasilar rales on lung auscultation. The heart sounds were normal, and bilateral ankle edema was present. Laboratory tests revealed hyponatremia (sodium, 124 mEq/L [to convert to millimoles per liter, multiply by 1]), a compensated metabolic acidosis with a bicarbonate concentration of 14 mEq/L (to convert to millimoles per liter, multiply by 1), and a lactate concentration of 82.0 mg/dL (to convert to millimoles per liter, multiply by 0.111). The D-dimer level was 0.0003 μg/mL (to convert to nanomoles per liter, multiply by 5.476), and the pro–brain-type natriuretic peptide level was 6062 pg/mL (to convert to nanograms per liter, multiply by 1). The electrocardiogram on presentation is shown in the Figure. Echocardiography (Video 1 and Video 2) revealed a left ventricular ejection fraction of 50%.
what would you do next?
What would you do next?
Administer intravenous thiamine therapy
Administer intravenous bicarbonate therapy
Administer intravenous β-blocker therapy
Start extracorporeal membrane oxygenation therapy
a
0
1
1
1
male
0
0
40
31-40
null
536
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2730376
A woman in her 30s presented with a 1-year medical history of a painless, slowly enlarging, midline neck mass without associated dysphagia, odynophagia, weight loss, or history of infection. The mass was soft, mobile, and compressible in midline level IA, measuring 7 cm and posterior-superiorly displacing the tongue. The mass was visible as a clear blue lesion in the floor of the mouth and was nontender. Computed tomographic (CT) scan of the neck with IV contrast was performed (Figure). The patient was taken to the operating room for a successful combined intraoral and transcervical approach to excision.Computed tomographic images. A, Coronal view of midline neck mass. B, Sagittal view of midline neck mass. What Is Your Diagnosis?
Simple ranula
Epidermoid cyst
Vascular malformation
Odontogenic abscess
B. Epidermoid cyst
B
Epidermoid cyst
Epidermoid cysts (ECs) are classically small, benign, cystic masses with 32% of lesions occurring in the head and neck.1 They usually arise because of failure of primitive epithelial cells to separate from underlying deep tissue during branchial arch formation.2 True ECs are fluid-filled lesions lined by simple squamous epithelium and a layer of keratin. They have been referred to by other terms such as epidermal cysts, epidermal inclusion cysts, sebaceous cysts, and seborrheic cysts. However, the terms “seborrheic cysts” and “sebaceous cysts” are misnomers, not synonymous with epidermoid cysts. Epidermoid cysts are normally close to the skin and can be located anywhere on the face, scalp, and neck. However, in the deep tissue planes, ECs in the head and neck make up only 1.6% to 6.9% of cases in the entire body.3The typical presentation of an EC is a soft, slow-growing, nontender mass without fixation to underlying tissues. Epidermoid cysts frequently do not cause symptoms and go unnoticed until they reach a large size or become infected.4 Obstructive symptoms like dysphagia, odynophagia, dysphonia, and dyspnea are more typical for lesions above the mylohyoid because this results in superior displacement of the tongue. In contrast, masses below the mylohyoid cause protrusion into the chin in the front of the neck, giving rise to the characteristic double chin appearance.5,6 In this case, the cyst was quite large, measuring over 8 cm in greatest dimension, but it produced no obstructive symptoms.Both benign and malignant neoplasms may present as cystic lesions in the floor of mouth.7 However, malignant disease is generally limited to necrotic squamous cell carcinoma lymphadenopathy. These lesions generally are thick-walled and found in the submandibular space, which is inconsistent with the sublingual lesion described herein. Venous malformations may also present as cystic masses in the neck with the presence of phleboliths as the characteristic imaging finding.8 Arteriovenous malformations may be visualized as high-flow lesions with multiple tortuous channels.8 The absence of characteristic imaging findings excludes vascular malformation as a viable consideration in this case.On CT scan, ECs are described as unilocular masses, and ranulas appear as well-circumscribed, lobulated cystic lesions with central homogeneous low CT attenuation.8 However, not all lesions present with these characteristic radiologic signs. Indeed, Coit et al7 note that ECs cannot be absolutely distinguished from ranulas by radiographic means alone. In this case, initial imaging and physical examination findings were more consistent with a ranula than an EC. The mass had a bluish coloration on examination, which is a common feature of ranulas. Findings of the CT scan showed a cystic, homogenous mass without hypoattenuating fat nodules, calcifications, or septations contained in the floor of the mouth and sublingual space, increasing our suspicion of a ranula. We initially planned for intraoral marsupialization with sublingual gland excision. However, working diagnosis was intraoperatively changed to dermoid cyst vs EC owing to thick cyst wall. The decision was made to completely excise the mass leaving the sublingual gland. Final pathologic diagnosis confirmed EC.Although inflamed ECs without infection may be treated with intralesional steroids alone, complete surgical excision or enucleation is regarded as the definitive treatment of choice.4 Recurrence after total excision is rare.9 Giant ECs greater than 5 cm are uncommon, and few cases in the head and neck have been reported.10 Herein we present a case of a giant deep tissue EC mimicking a simple ranula, serving as a reminder to include EC in the differential for cystic masses of the floor of mouth or midline of the neck.
General
A woman in her 30s presented with a 1-year medical history of a painless, slowly enlarging, midline neck mass without associated dysphagia, odynophagia, weight loss, or history of infection. The mass was soft, mobile, and compressible in midline level IA, measuring 7 cm and posterior-superiorly displacing the tongue. The mass was visible as a clear blue lesion in the floor of the mouth and was nontender. Computed tomographic (CT) scan of the neck with IV contrast was performed (Figure). The patient was taken to the operating room for a successful combined intraoral and transcervical approach to excision.Computed tomographic images. A, Coronal view of midline neck mass. B, Sagittal view of midline neck mass.
what is your diagnosis?
What is your diagnosis?
Vascular malformation
Epidermoid cyst
Odontogenic abscess
Simple ranula
b
1
1
0
1
female
0
0
1
0-10
null
537
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2731469
A 36-year-old otherwise healthy white woman with a history of chronic sinusitis presented with progressive left facial pain and swelling of 5 months’ duration. Associated symptoms included epistaxis and left-sided epiphora. Prior to presentation, she had been treated as an outpatient with multiple rounds of oral antibiotics and steroids for presumed sinusitis without improvement. Social history included routine cocaine use 10 years ago. Physical examination demonstrated substantial erythema and edema of the left infraorbital region and left nasal sidewall and purulent nasal secretions (Figure 1A). Endoscopic nasal examination revealed substantial swelling of the left nasal vestibule completely obstructing the left naris, swelling of the left nasolacrimal duct orifice, nearly total septal perforation with areas of necrotic bone, and no identifiable intranasal landmarks except for a remnant of the left middle turbinate (Figure 1B). There was no cervical lymphadenopathy. Laboratory evaluation revealed a white blood cell count of 12 300/μL with normal neutrophil count; erythrocyte sedimentation rate of 83 mm/h; and a C-reactive protein level of 134 mg/L. (To convert white blood cells to ×109/L, multiply by 0.001; C-reactive protein to nanomoles per liter, multiply by 9.524.) A maxillofacial computed tomographic (CT) scan demonstrated cartilaginous and anterior osseous nasal septal perforation and left nasal and preseptal soft-tissue thickening with associated periosteal reaction of the nasal process of the maxilla.A, Periorbital and nasal cellulitis. B, Nasal endoscopic examination demonstrating significant soft-tissue necrosis. What Is Your Diagnosis?
Granulomatosis with polyangiitis
Cocaine-induced midline destructive lesion
Extranodal natural killer/T-cell lymphoma, nasal type
Chronic invasive fungal sinusitis
B. Cocaine-induced midline destructive lesion
B
Cocaine-induced midline destructive lesion
The patient began treatment with broad-spectrum intravenous antibiotics, which was deescalated to oral trimethoprim-sulfamethoxazole and rifampin treatment for 6 weeks after nasal swab culture grew methicillin-sensitive Staphylococcus aureus. A repeated nasal culture at follow-up grew Pseudomonas aeruginosa, so she was then treated with a 6-week regimen of levofloxacin and rifampin. After completion of the antibiotics regimen, her condition showed only mild improvement, so she underwent biopsy and surgical debridement of necrotic tissue, which exposed an underlying nasocutaneous fistula over the left nasal sidewall (Figure 2). Biopsy specimens demonstrated necroinflammatory debris and acute and chronic osteomyelitis, which together with her history were suggestive of cocaine-induced midline destructive lesion (CIMDL). No pathognomonic lesions suggestive of granulomatosis with polyangiitis (GPA) were seen.Axial noncontrast computed tomographic maxillofacial scan after antibiotic treatment demonstrates destruction of the left frontal process of the maxilla and cartilaginous and osseous nasal septum.Cocaine acts as a local anesthetic, vasoconstrictive, and euphoric agent. It blocks sodium channels and thereby halts electrical propagation of neurons to cause local anesthesia.1 It also inhibits the reuptake of norepinephrine and dopamine at the presynaptic nerve endings, leading to an accumulation of these neurotransmitters at nerve synapses.2 In the peripheral nervous system, norepinephrine primarily accumulates, causing an increase in sympathetic tone.3 In the central nervous system, dopamine accumulation is responsible for cocaine’s euphoric and addictive properties.3,4 It is reported that close to 30 million people in the United States have used cocaine and that 6 million of them are compulsive users; however, CIMDL is only seen in 4.8% of users.5,6Nasal septal perforation, nasal cavity wall erosion, pharyngeal wall ulceration, and hard palate perforation are all manifestations of CIMDL. The damage to the nasal architecture is multifactorial, primarily the result of intense vasoconstriction with repeated cocaine use, leading to local tissue necrosis. However, the chemical irritation from adulterants, such as lactose and inositol, and mechanical irritation from self-instrumentation and high-velocity cocaine crystal inhalation have also been implicated in mucosal and perichondrial erosion.7The clinical presentation of CIMDL also mimics certain systemic diseases, most notably GPA. Clinical differentiation can be made based on more severe destruction of nasal architecture, such as loss of inferior and middle turbinates, and lack of systemic manifestations in CIMDL.8 Histopathological findings are very similar in CIMDL and GPA unless discriminatory findings of GPA are seen, such as necrotizing granulomas and multinucleated giant cells.8 Finally, positive findings for antineutrophil cytoplasmic antibodies (ANCAs) are common in both patient populations, but reaction against human neutrophil elastase (HNE) in CIMDL is its most distinguishing feature.9 The presence of HNE-ANCA is what is believed to predispose select cocaine users to CIMDL because it enhances the local inflammatory response to injury—highlighting a possible autoimmune component to the disease.9 It is also reported that there is a time and cocaine-dose dependence to developing the disease, suggesting a chronicity, rather than fulminant stage, to the disease entity.6It is important to remember that patients tend to understate cocaine use, as did this patient, who only reported recent use after her pain progressed. Treatment includes complete abstinence from cocaine, debridement of necrotic tissue, and treatment of any superinfection. The most common surgical procedures are closure of septal and palatal perforation, with most surgeons requiring 6 to 12 months of cocaine abstinence prior to intervention.9 Of note, immunosuppression is not indicated; therefore, correct identification of the underlying disease process is critical.6 The present case demonstrates an uncommonly severe presentation of CIMDL and illustrates the importance of correct and timely diagnosis to guide subsequent treatment.
General
A 36-year-old otherwise healthy white woman with a history of chronic sinusitis presented with progressive left facial pain and swelling of 5 months’ duration. Associated symptoms included epistaxis and left-sided epiphora. Prior to presentation, she had been treated as an outpatient with multiple rounds of oral antibiotics and steroids for presumed sinusitis without improvement. Social history included routine cocaine use 10 years ago. Physical examination demonstrated substantial erythema and edema of the left infraorbital region and left nasal sidewall and purulent nasal secretions (Figure 1A). Endoscopic nasal examination revealed substantial swelling of the left nasal vestibule completely obstructing the left naris, swelling of the left nasolacrimal duct orifice, nearly total septal perforation with areas of necrotic bone, and no identifiable intranasal landmarks except for a remnant of the left middle turbinate (Figure 1B). There was no cervical lymphadenopathy. Laboratory evaluation revealed a white blood cell count of 12 300/μL with normal neutrophil count; erythrocyte sedimentation rate of 83 mm/h; and a C-reactive protein level of 134 mg/L. (To convert white blood cells to ×109/L, multiply by 0.001; C-reactive protein to nanomoles per liter, multiply by 9.524.) A maxillofacial computed tomographic (CT) scan demonstrated cartilaginous and anterior osseous nasal septal perforation and left nasal and preseptal soft-tissue thickening with associated periosteal reaction of the nasal process of the maxilla.A, Periorbital and nasal cellulitis. B, Nasal endoscopic examination demonstrating significant soft-tissue necrosis.
what is your diagnosis?
What is your diagnosis?
Granulomatosis with polyangiitis
Cocaine-induced midline destructive lesion
Extranodal natural killer/T-cell lymphoma, nasal type
Chronic invasive fungal sinusitis
b
1
1
0
1
female
0
0
36
31-40
White
538
original
https://jamanetwork.com/journals/jama/fullarticle/2731736
A 24-year-old woman who worked on a farm in Connecticut developed fever, chills, vomiting, and a truncal maculopapular rash 3 weeks before presentation. One week after symptom onset, she remained febrile (maximum temperature, 39.6°C [103.2°F]) and the rash spread to her palms and soles, with some progression to pustules (Figure, left panel). She developed right knee pain, followed by pain in other joints. Two weeks after symptom onset, she presented to a local emergency department, reporting inability to stand due to severe joint pain. Results of tests for sexually transmitted infections and respiratory viruses were negative. She was presumptively diagnosed with a viral illness and discharged home with supportive care. Approximately 3 weeks after symptom onset, she presented to the emergency department again with persistent fever, worsening arthralgia, back pain, and progressive purpuric and pustular rash. Her vital signs were unremarkable. White blood cell count was 12 100/μL (reference range, 4000/μL-10 000/μL), with 84% neutrophils. Urinalysis results and levels of serum electrolytes and liver enzymes were within normal limits. Gram stain of blood culture is shown in the Figure (right panel).Obtain joint fluid for bacterial analysis and cultures What Would You Do Next?
Inquire about rodent and other animal exposures
Obtain joint fluid for bacterial analysis and cultures
Test for Borrelia burgdorferi
Test vesicular fluid for enterovirus
Rat bite fever
A
Inquire about rodent and other animal exposures
The key to the correct diagnosis was symptoms of persistent fever, migratory polyarthralgias, and purpuric rash extending to the palms and soles. When questioned, the patient recalled a rat bite 3 days before symptom onset. Based on this history, pustule fluid and blood were submitted for culture. Blood cultures became positive, and Gram stain revealed filamentous gram-negative bacilli in “nests,” with occasional bulbous swellings. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry failed to identify the organism, but 16S rDNA sequencing identified Streptobacillus moniliformis, the causative agent of rat bite fever. No growth occurred from pustule fluid cultures.Because of the nonspecific presentation of rat bite fever, patients with a high index of clinical suspicion should be carefully questioned for rodent exposure.1 Even without a known exposure, rat bite fever should be considered in patients with an unexplained febrile illness and rash, arthritis, or risk factors such as occupational or recreational exposure to rodents.2 The duration of the patient’s symptoms and absence of oral symptoms made the diagnosis of hand, foot, and mouth disease due to enterovirus unlikely. Although testing for tick-transmitted diseases or joint fluid analysis may eventually be warranted, the next best step is obtaining a thorough history regarding exposure to rodents. This approach improves pretest probability and avoids premature conclusions by developing a focused differential diagnosis based on history and examination.3Rat bite fever is caused by S moniliformis in the United States or Spirillum minus in Asia. S moniliformis is a fastidious, facultatively anaerobic, filamentous gram-negative bacteria that colonizes the oral and nasopharyngeal tracts of rats, mice, squirrels, gerbils, and other rodents. Rat bite fever is not a notifiable disease in the United States, and, while incidence appears low, its precise incidence is unknown.1 However, infections are increasing, especially in children, because of the popularity of rodents as pets.4 Transmission also occurs by ingesting food contaminated by urine of colonized rats, causing a syndrome known as “Haverhill fever.”5Rat bite fever is characterized by fever, chills, headache, and vomiting 3 to 10 days after exposure. Without treatment, fever typically follows a relapsing-remitting pattern. Most patients develop a maculopapular, petechial, or purpuric rash, especially on the extensor surfaces of the extremities and extending to the hands and feet. Lesions can evolve into pustules and hemorrhagic vesicles. Most patients develop migratory polyarthralgias involving both small and large joints.1Rodent bite wounds should be thoroughly cleansed, and antibiotic prophylaxis and tetanus immunization may be warranted if not up to date. Rabies prophylaxis is almost never indicated for small rodent bites.6 Symptoms of rat bite fever can present early, but diagnosis is usually delayed or missed because of poor memory of rodent exposure. Additionally, the nonspecific presentation may mimic viral infections, sexually transmitted infections, drug reactions, and vasculitis.1,7S moniliformis is fastidious, and its growth is inhibited by an anticoagulant used in blood cultures. In this patient, growth occurred in the aerobic blood culture bottle but only on anaerobic solid media. No special incubation or plating strategies were used for the blood cultures, but supplemented culture methods have historically been recommended to identify S moniliformis.1 There should be communication with the microbiology laboratory to optimize the diagnostic approach for patients with suspected rat bite fever. Identification of cultured S moniliformis is challenging, as it may not be present in commercial identification databases. Definitive identification may require specialized biochemical analysis,1 sequencing, or both.Infectious diseases consultation can assist with risk assessment, laboratory communication to optimize testing, and choice of empirical antimicrobial therapy. Penicillin is the treatment of choice when diagnosis is confirmed; cephalosporins and tetracyclines may also be effective. Rat bite fever can be associated with serious complications including endocarditis, hepatitis, nephritis, septic arthritis, osteomyelitis, meningitis, and sepsis. When diagnosed, rat bite fever can be successfully treated; however, mortality can reach 13% without treatment.7-10,The patient was initially treated with penicillin and doxycycline empirically. After the diagnosis of rat bite fever, treatment was changed to intravenous penicillin G monotherapy. Transthoracic echocardiography did not reveal valvular vegetations. Persistent back pain prompted magnetic resonance imaging of the spine, which revealed changes consistent with lumbar osteomyelitis. Treatment was extended to 6 weeks, and the patient’s symptoms resolved without need for repeat imaging.
General
A 24-year-old woman who worked on a farm in Connecticut developed fever, chills, vomiting, and a truncal maculopapular rash 3 weeks before presentation. One week after symptom onset, she remained febrile (maximum temperature, 39.6°C [103.2°F]) and the rash spread to her palms and soles, with some progression to pustules (Figure, left panel). She developed right knee pain, followed by pain in other joints. Two weeks after symptom onset, she presented to a local emergency department, reporting inability to stand due to severe joint pain. Results of tests for sexually transmitted infections and respiratory viruses were negative. She was presumptively diagnosed with a viral illness and discharged home with supportive care. Approximately 3 weeks after symptom onset, she presented to the emergency department again with persistent fever, worsening arthralgia, back pain, and progressive purpuric and pustular rash. Her vital signs were unremarkable. White blood cell count was 12 100/μL (reference range, 4000/μL-10 000/μL), with 84% neutrophils. Urinalysis results and levels of serum electrolytes and liver enzymes were within normal limits. Gram stain of blood culture is shown in the Figure (right panel).Obtain joint fluid for bacterial analysis and cultures
what would you do next?
What would you do next?
Obtain joint fluid for bacterial analysis and cultures
Test for Borrelia burgdorferi
Inquire about rodent and other animal exposures
Test vesicular fluid for enterovirus
c
0
1
1
1
female
0
0
24
21-30
White
539
original
https://jamanetwork.com/journals/jama/fullarticle/2730536
A 71-year-old otherwise healthy woman presented with an enlarging, immobile, painless mass in her right gluteal area. She had no other associated symptoms. On examination, she had a large, palpable, nontender right gluteal mass. She had no neurovascular deficits. Results of laboratory studies were within normal limits. Computed tomography (CT) revealed an 11-cm heterogeneous lipomatous tumor involving the right gluteal musculature (Figure).Axial (left) and coronal (right) computed tomography images of patient’s right gluteal mass. What Would You Do Next?
Incisional biopsy of the lesion
Core needle biopsy of the lesion
Simple excision of the lesion
Repeat imaging in 6 months
Well-differentiated/dedifferentiated liposarcoma
B
Core needle biopsy of the lesion
Core needle biopsy should be performed to obtain tissue diagnosis. Incisional biopsy should be avoided and can complicate surgical and oncologic management.1 Surgical management should be deferred until a tissue diagnosis is made and preoperative workup is complete. Because this tumor is enlarging, larger than 5 cm, deep (subfascial), and heterogeneous, it should be considered malignant until proven otherwise. Repeat imaging is unnecessary and likely would be inadequate.Liposarcomas are rare, often aggressive, malignancies that require surgical resection and multidisciplinary management. There are approximately 2400 new cases of liposarcoma diagnosed in the United States per year.2,3 Subtypes of liposarcoma include well-differentiated, dedifferentiated, myxoid, and pleomorphic. A well-differentiated liposarcoma of the extremity may also be referred to as an atypical lipomatous tumor.As with all soft tissue sarcomas, liposarcomas are most frequently found on the extremities or in the retroperitoneum. Lipomatous tumors are relatively asymptomatic. A lipomatous tumor arising in the extremity typically presents as a painless mass and in the retroperitoneum occasionally presents with vague abdominal symptoms.Cross-sectional imaging, either CT or magnetic resonance imaging (MRI), should be obtained for all patients. Extremity tumors that are large (>5 cm), subfascial, or enlarging and any retroperitoneal mass should be considered malignant until proven otherwise. In most cases, core needle biopsy should be obtained for tissue diagnosis. Incisional biopsy should be avoided and can complicate surgical and oncologic management.1MDM2 amplification, detected by fluorescence in situ hybridization, can provide additional information. This is particularly useful in distinguishing a well-differentiated liposarcoma (MDM2 amplification) from a benign lipoma (no MDM2 amplification), as this is often a difficult histologic distinction, radiologic distinction, or both.4 Dedifferentiated (MDM2 amplification), myxoid (no MDM2 amplification), and pleomorphic (no MDM2 amplification) liposarcomas have more characteristic appearances on histology and imaging, so MDM2 amplification may be less useful in these cases.All patients diagnosed with liposarcoma should undergo CT of the chest to complete preoperative staging, as sarcomas spread hematogenously, most frequently to the lung. A chest radiograph may be an appropriate substitute for CT of the chest for patients diagnosed with a well-differentiated liposarcoma/atypical lipomatous tumor in an extremity. Patients diagnosed with myxoid liposarcoma should additionally undergo CT of the abdomen/pelvis and be considered for MRI of the spine, as this liposarcoma subtype can metastasize to other anatomical locations.Surgery remains the mainstay of treatment for liposarcoma, and oncologic margin–negative resection is associated with improved disease-free survival. Amputation is rarely, if ever, required for primary extremity disease. Identifying malignant tumors preoperatively is critical for surgical planning and avoiding unnecessarily aggressive operations to obtain local control. A patient who undergoes a simple excision for a presumed lipoma that is found to be a liposarcoma should undergo repeat imaging and often a wide reresection of the surgical site, as there is a 50% to 80% rate of gross residual disease in this setting.5Radiation therapy has been shown to improve local control for patients with primary high-grade extremity soft tissue sarcomas, including liposarcomas. However, the exceptions of the use of radiation are small, low-grade liposarcomas or well-differentiated liposarcomas/atypical lipomatous tumors, given their low risk of recurrence and the potential long-term complications from radiation.6 Although the benefit is less clear for retroperitoneal disease, radiation may be used in highly selected cases for the treatment of aggressive histologic subtypes by centers with expertise in treating sarcoma in this location. For both the extremity and retroperitoneum, preoperative (neoadjuvant) radiation is preferred because of its favorable late toxicity profile, shorter course, and potential operative benefits of downsizing the tumor.7 The benefit of systemic therapy (chemotherapy, molecularly targeted therapy, or immunotherapy) for high-risk liposarcomas is less clear and is only considered for specific histologic subtypes or for patients with metastatic or locally advanced disease.8Risk of local recurrence (5%-15%) and distant metastasis (5%-70%) of extremity liposarcomas ranges widely and is dependent on the histologic subtype, grade, and size. In comparison to extremity liposarcomas, the risk of local recurrence for retroperitoneal liposarcomas is significantly higher (40%-50%). Widely validated prognostic models or nomograms more accurately determine oncologic prognosis than existing TNM staging systems. Websites, such as the Memorial Sloan Kettering Cancer Center nomogram, and applications, such as Sarculator (which uses validated nomogram data), give patient-specific prognosis data and are used by both sarcoma oncologists and patients.9A patient diagnosed with liposarcoma should be referred to a sarcoma specialty center whenever possible, as multidisciplinary treatment and surveillance by physicians specializing in sarcoma has been shown to improve patient outcomes.10Core needle biopsy revealed a spindle and pleomorphic sarcoma, most consistent with a well-differentiated/dedifferentiated liposarcoma with MDM2 amplification present. CT of the chest showed no systemic disease. After a multidisciplinary discussion, the patient underwent neoadjuvant radiation followed by resection. She has undergone surveillance imaging every 6 months since her surgery and remains disease-free 2 years postoperatively.
General
A 71-year-old otherwise healthy woman presented with an enlarging, immobile, painless mass in her right gluteal area. She had no other associated symptoms. On examination, she had a large, palpable, nontender right gluteal mass. She had no neurovascular deficits. Results of laboratory studies were within normal limits. Computed tomography (CT) revealed an 11-cm heterogeneous lipomatous tumor involving the right gluteal musculature (Figure).Axial (left) and coronal (right) computed tomography images of patient’s right gluteal mass.
what would you do next?
What would you do next?
Repeat imaging in 6 months
Core needle biopsy of the lesion
Simple excision of the lesion
Incisional biopsy of the lesion
b
1
0
0
1
female
0
0
71
71-80
null
540
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2728813
A man in his 40s was admitted to the hospital with a 10-day history of edema and presented with vesicles and bullae on his right cheek, neck, back, both eyelids, and the dorsal surface of his hands (Figure 1A). Prior to presentation, he had been treated for herpes zoster with famciclovir for 1 week, during which the lesions kept developing and were accompanied with irregular fever (maximum temperature, 39°C). Physical examination revealed edema on both eyelids; multiple papules, blisters, and crusted erosions with thin exudation on his right cheek and neck; and erythematous plaques and tense blisters and bullae on his back and on the dorsal surface of both hands (Figure 1B). A full laboratory workup was performed for autoantibodies for systemic lupus erythematosus (SLE), pemphigus, and bullous pemphigoid; skin and bone-marrow biopsies and direct immunofluorescence were performed; and immunohistochemichal analysis, a swab of exudation, and bacterial, fungal, and atypical mycobacterial cultures from blood and tissue were also examined.A, Edema on both eyelids; multiple papules, blisters, and crusted erosions with thin exudation on right cheek and neck. B, Erythematous plaques, tense blisters, and bullae on both hands (right hand depicted). What Is Your Diagnosis?
Famciclovir-resistant disseminated herpes zoster
Bullous Sweet syndrome
Bullous Wells syndrome
Bullous pemphigoid
C. Bullous Wells syndrome
C
Bullous Wells syndrome
The complete blood cell count showed an elevated eosinophil count (1.36 × 109/L). No additional anomalies were found besides a mild increase in eosinophil levels in the bone marrow biopsy. Enterococcus faecalis was isolated from the exudation swab. Blood and tissue cultures and autoantibody profiles of SLE, pemphigus (Dsg-1 and Dsg-3), and bullous pemphigoid (BP-180 and BP-230) were all negative. Histopathologic examination indicated irregular hyperplasia and spongiosis of the epidermis with hyperkeratosis and localized necrosis (Figure 2A). Additionally, extensive edema of dermal papillary and mixed infiltrate of marked diffuse eosinophils and lymphocytes were found in the dermis, accompanied by scattered neutrophils (Figure 2B). Direct immunoflourescence results for IgA, IgG, IgM, and C3 were negative. Initially, the patient was given levofloxacin for 5 days owing to a concern of bacterial infection but did not respond. Following the histopathologic results, the diagnosis of bullous Wells syndrome was made. Intravenous methylprednisolone at 1.5mg/kg/d was given for 1 week, followed by rapid relief of fever and lesions. The lesions completely cleared, and the patient showed no sign of relapse at 12-month follow-up.Hematoxylin-eosin stained sections. A, Irregular hyperplasia and spongiosis of the epidermis, extensive edema of dermal papillary and massive inflammatory cell infiltration. B, Extensive edema of dermal papillary and mixed infiltrate of marked diffuse eosinophils and lymphocytes in the dermis, accompanied by scattered neutrophils.Wells syndrome, also known as eosinophilic cellulitis, was first described in 1971.1 The main clinical variants include plaque type, annular granulomalike, urticarialike, papulovesicular, bullous, papulonodular, and fixed drug eruptionlike, among which the bullous type is quite rare and more likely to affect adults.2The low prevalence of Wells syndrome, along with its variability of manifestation, often delays its diagnosis until a patient does not respond to an initial antimicrobial regimen.3 Although the onset of Wells syndrome is acute, the systemic symptoms are generally mild. The irregular fever presented in this case is not a common complication. Besides bacterial cellulitis, differential diagnoses of Wells syndrome include, but are not limited to, necrotizing fasciitis, parasitoses, urticaria, Churg-Strauss syndrome, granuloma annulare, and hypereosinophilic syndrome.4 In patients presenting blisters or bullae, the diagnoses of herpes virus infection, bullous pemphigoid, acute contact dermatitis, bullous SLE, and bullous Sweet syndrome should also be considered. Notably, bullous Sweet syndrome can present symptoms very similar to the present case. However, the histologic traits of Sweet syndrome feature dense neutrophils and nuclear dust in the mid-dermis, with occasional eosinophils or lymphocytes.A variety of triggers are reported to be associated with Wells syndrome, including drugs, infections, insect bites, cancer, and vaccinations.5 However, in this case, no obvious triggers could be found. The history, physical examination, and regular laboratory workup excluded systemic diseases. Furthermore, the blood tests and bone-marrow biopsy provided no evidence to indicate either hematological or myeloproliferative diseases. Therefore, this case was diagnosed as idiopathic Wells syndrome.A gold standard for Wells syndrome diagnostic criteria is currently nonexistent. Heelan et al6 have proposed a set of diagnostic criteria for Wells syndrome, which include 4 major characteristics (2 of which need to be present) and 4 minor ones (at least 1 of which needs to be present). However, larger patient cohorts are needed for its validation. In a recent review, Räßler et al5 suggested that the correlation of clinical features, the course of skin lesions, and histopathological examination of a skin biopsy are necessary for a definitive diagnosis of Wells syndrome.Owing to the benign course and generally good prognosis, local therapy is the main strategy for treating Wells syndrome, whereas systemic treatment is used in cases with widespread lesions or systemic involvement. Topical and systemic glucocorticosteroids are the most common treatments for Wells syndrome and to our knowledge are the only clearly beneficial therapies reported so far.5 Other treatment options include cyclosporine, dapsone, oral or topical tacrolimus, antihistamines, interferon-α, interferon-γ, tumor necrosis factor inhibitors, sulphone or sulfasalazine, psoralen and UV-A therapy, colchicines, antimalarial drugs, azathioprine, minocycline, and griseofulvin.5
Dermatology
A man in his 40s was admitted to the hospital with a 10-day history of edema and presented with vesicles and bullae on his right cheek, neck, back, both eyelids, and the dorsal surface of his hands (Figure 1A). Prior to presentation, he had been treated for herpes zoster with famciclovir for 1 week, during which the lesions kept developing and were accompanied with irregular fever (maximum temperature, 39°C). Physical examination revealed edema on both eyelids; multiple papules, blisters, and crusted erosions with thin exudation on his right cheek and neck; and erythematous plaques and tense blisters and bullae on his back and on the dorsal surface of both hands (Figure 1B). A full laboratory workup was performed for autoantibodies for systemic lupus erythematosus (SLE), pemphigus, and bullous pemphigoid; skin and bone-marrow biopsies and direct immunofluorescence were performed; and immunohistochemichal analysis, a swab of exudation, and bacterial, fungal, and atypical mycobacterial cultures from blood and tissue were also examined.A, Edema on both eyelids; multiple papules, blisters, and crusted erosions with thin exudation on right cheek and neck. B, Erythematous plaques, tense blisters, and bullae on both hands (right hand depicted).
what is your diagnosis?
What is your diagnosis?
Bullous Wells syndrome
Bullous pemphigoid
Famciclovir-resistant disseminated herpes zoster
Bullous Sweet syndrome
a
0
0
1
1
male
0
0
45
41-50
null
541
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2729070
A woman in her 60s presented to the clinic with a 3-month history of pain, swelling, and erythema on her face. Eleven years earlier, she had been diagnosed with left breast carcinoma and had undergone a mastectomy with subsequent radiotherapy and chemotherapy. Seven years later, she had been found to have left supraclavicular lymph nodes metastasis and had undergone a surgical excision. On presentation for the current problem, the patient experienced pain on palpation, but denied fever, chills, or other symptoms. A physical examination revealed violaceous swelling with telangiectasia and necrosis on the forehead, eyelids, nose, and cheek, predominantly involving the left side of the face (Figure 1). A biopsy of the lesion was performed for histopathological analysis (Figure 2).A, A full image and B, a three-quarters image of the face, showing telangiectatic, swelling erythema mainly involving the left side of the forehead, eyelids, nose, and cheek. What Is Your Diagnosis?
Cutaneous angiosarcoma
Radiation dermatitis
Wolf isotopic response
Telangiectatic metastatic breast carcinoma
D. Telangiectatic metastatic breast carcinoma
D
Telangiectatic metastatic breast carcinoma
Histopathological examination revealed hyperkeratosis, epidermal atrophy, and dilated vessels in the dermis. One vessel had nests of highly atypical cells (Figure 2A). On immunohistochemical examination, epithelial cells in the dilated vessels were positive for CD31, and the atypical cells were strongly positive for GATA3 (Figure 2B). A diagnosis of telangiectatic metastatic breast carcinoma (TMBC) was made.A, Highly atypical cells in a dilated vessel in the dermis (hematoxylin-eosin stain; original magnification, ×10). B, Atypical cells in dilated vessels (streptavidin-peroxidase stain; original magnification, ×20).Breast carcinoma is the most prevalent cancer and the second most common cause of cancer death in women.1 Metastatic breast carcinoma most frequently involves the lymph nodes, lung, liver, and bone marrow; cutaneous metastases are relatively uncommon, with an incidence of 2.42%.2 Clinical manifestations can include nodules (47%-80%), alopecia neoplastica (2%-12%), telangiectatic carcinoma (8%-11%), carcinoma erysipeloides (3%-6%), and melanoma-like metastases (6%).1,3 Common sites of cutaneous metastases are the chest and abdomen; the face is rarely affected.4,5 To our knowledge, 2 cases of metastatic breast carcinoma involving the face have been reported, simulating cutaneous angiosarcoma: one with facial telangiectasia and erythema4 and the other with a large red-purplish plaque and stony, hard nodules.5Cases of TMBC have variable clinical appearances and usually present as fleshy to erythematous nodules on the chest; they rarely involve the face. Cutaneous angiosarcoma usually begins as bruise-like patches on the skin, as well as erythematous or violaceous macules, patches, papules, and nodules on the face and scalp of elderly people.6 Lesions may be solitary or multiple; facial swelling may occur. The clinical difference might be the lesion spreading pattern. Cutaneous angiosarcoma usually spreads centrifugally and involves both sides of the face, whereas metastases from a left breast carcinoma mainly affect the left side of the face, as in this case. Histopathologically, cutaneous angiosarcoma is lined by atypical endothelial cells that exhibit apparent nuclear pleomorphism and high mitotic activity, as opposed to those in TMBC, which typically displays tumor cells within dilated vessels in the superficial dermis that can be seen in cutaneous metastases of other carcinomas. Panels of immunohistochemical markers can help determine primary sites of metastasis, including estrogen, progesterone, cytokeratin 7, GATA binding protein 3, and gross cystic disease fluid protein–15.Other differential diagnoses include radiation dermatitis, Wolf isotopic response, and herpes zoster. Radiation dermatitis usually exhibits poikilodermatous changes on the skin surface, including erythema, telangiectasia, hyperpigmentation, and hypopigmentation. Since the patient underwent radiotherapy 11 years earlier, this lesion may be confused with chronic radiation dermatitis, which also presents as painful erythema with telangiectasia and necrosis and displays vascular dilation histopathologically.7 However, other histopathological features of chronic radiation dermatitis did not exist, including pigmentary alteration in the epidermis, dermal fibrosis, irregular shaped fibroblasts, and loss of adnexal structures. It is also important to suspect herpes zoster when a painful, edematous erythema with unilateral dermatomal distribution occurs in an elderly woman who had received immunosuppressive therapy. Histopathologically, herpes zoster is characterized by intraepidermal vesicles, ballooning degeneration of enlarged keratinocytes, and eosinophilic intranuclear inclusion bodies in multinucleated giant cells.8 Also, Wolf isotopic response is the subsequent occurrence of a new skin condition at the site of another unassociated, previously healed disease. The most common primary disease is herpes zoster, which has been reported to be followed by a variety of conditions.9 Thus, the cutaneous angiosarcoma–like lesion in zosteriform distribution raised the possibility of Wolf isotopic response. However, the patient did not recall a varicella-zoster infection or any skin condition on her face. The presence of atypical cells in dilated vessels excluded these diagnoses.The 5-year survival rate is reported10 to be 38% in patients with cutaneous metastases vs 99% in those with localized breast cancer. Recognition is crucial for early diagnosis and management, and suspicion for metastatic spread should be raised in patients with breast cancer who have telangiectatic facial swelling. Unfortunately, the patient was in poor health and could not undergo or afford further treatment. She was still alive at 5-month follow-up.
Dermatology
A woman in her 60s presented to the clinic with a 3-month history of pain, swelling, and erythema on her face. Eleven years earlier, she had been diagnosed with left breast carcinoma and had undergone a mastectomy with subsequent radiotherapy and chemotherapy. Seven years later, she had been found to have left supraclavicular lymph nodes metastasis and had undergone a surgical excision. On presentation for the current problem, the patient experienced pain on palpation, but denied fever, chills, or other symptoms. A physical examination revealed violaceous swelling with telangiectasia and necrosis on the forehead, eyelids, nose, and cheek, predominantly involving the left side of the face (Figure 1). A biopsy of the lesion was performed for histopathological analysis (Figure 2).A, A full image and B, a three-quarters image of the face, showing telangiectatic, swelling erythema mainly involving the left side of the forehead, eyelids, nose, and cheek.
what is your diagnosis?
What is your diagnosis?
Cutaneous angiosarcoma
Telangiectatic metastatic breast carcinoma
Radiation dermatitis
Wolf isotopic response
b
0
0
1
1
female
0
0
65
61-70
null
542
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2729591
A man in his 50s presented with an abdominal skin eruption of 2 days’ duration. His medical history included hepatitis C, cirrhosis, and an orthotopic liver transplant (OLT) 3 years prior that was subsequently treated with tacrolimus, 5 mg, and mycophenolate, 750 mg, twice daily. Approximately 2½ years after the transplant, the patient developed gastric outlet obstruction secondary to an infiltrative gastric wall mass. The gastric wall mass was found to be associated with plasmablastic posttransplantation lymphoproliferative disorder (pPTLD). He was subsequently instructed to stop use of mycophenolate and decrease use of tacrolimus to 0.25 mg daily, then treated with 1 cycle of CHOP chemotherapy (cyclophosphamide, doxorubicin hydrochloride, vincristine sulfate, and prednisone) and lenalidomide, resulting in symptomatic and radiologic improvement. Five weeks after treatment with CHOP chemotherapy, the patient presented with an acute-onset abdominal skin eruption without fever or pain. A computed tomographic scan of the abdomen and pelvis showed possible abdominal wall cellulitis, so treatment with piperacillin-tazobactam and vancomycin was empirically started. Physical examination revealed confluent indurated violaceous papules and plaques coalescing into an annular pattern encircling the healed OLT incisional scar with central sparing (Figure 1A). A 4-mm punch biopsy specimen was obtained for histopathological evaluation (Figure 1B and C) and bacterial and fungal cultures.A, Confluent, indurated, violaceous papules and plaques coalescing into an annular pattern encircling the well-healed orthotopic liver transplant incisional scar. B, Hematoxylin-eosin staining of the punch biopsy shows an atypical, hyperchromatic infiltrate present throughout the superficial and deep dermis. C, High magnification reveals crushed but markedly atypical-appearing lymphoid cells with nuclear hyperchromasia and scant amounts of cytoplasm. Multiple mitotic and apoptotic figures are easily identified. What Is Your Diagnosis?
Reactive granulomatous disorder
Posttransplantation lymphoproliferative disorder
Deep fungal infection
Cutaneous T-cell lymphoma
B. Posttransplantation lymphoproliferative disorder
B
Posttransplantation lymphoproliferative disorder
Histopathologic analysis showed diffuse, atypical lymphoid proliferation infiltrating throughout the superficial and deep dermis with epidermal sparing (Figure 1B). High-powered examination revealed enlarged, markedly atypical-appearing crushed hyperchromatic cells with irregular nuclei and scant amounts of pale eosinophilic and amphophilic cytoplasm (Figure 1C). Numerous apoptotic and mitotic figures were identified. Plasmacytic features were seen. The histologic differential included cutaneous involvement by the known pPTLD vs high-grade lymphoma or leukemia cutis (acute B-cell or T-cell lymphoblastic lymphoma or leukemia), diffuse large B-cell lymphoma, mantle cell lymphoma, acute myeloid leukemia, and small round blue cell tumors (Merkel cell carcinoma and metastatic small cell carcinoma). In situ hybridization and kappa light chain immunohistochemical staining were negative for CD3, CD20, BCL2, BCL6, and Epstein-Barr virus (EBV). CD138 (Figure 2) and lambda chain staining were strongly and diffusely positive. The histology and presence of plasmacytic features in the setting of similar immunohistochemical findings to the patient’s gastric pPTLD made the diagnosis most likely multiorgan pPTLD.Immunohistochemical stain shows that the lesional cells are strongly and diffusely positive for the plasma cell marker CD138.Owing to liver rejection concern, treatment with tacrolimus was initially continued. The patient was treated with 1 cycle of CHOP chemotherapy with resolution of cutaneous findings. However, his internal posttransplantation lymphoproliferative disorder (PTLD) progressed. Cessation of tacrolimus treatment, and additional treatment with CHOP chemotherapy, lenalidomide, bortezomib, and daratumumab were trialed without response, and the patient was referred to hospice care for refractory, rapidly progressive disease.The malignancy PTLD is a complication of immunosuppression for solid organ and hematopoietic stem cell transplants. The incidence of PTLD following OLT in adults is 2% to 3%.1 Usual sites of involvement include the lymph nodes and gastrointestinal tract, whereas cutaneous involvement is very rare.2 Multiorgan involvement occurs in 41% of cases.1The broader PTLD encompass a heterogeneous group of lymphoproliferative diseases categorized histologically. A pPTLD diagnosis is very rare. Only 21 cases of cutaneous pPTLD have been reported, and 8 of those cases involve multiorgan disease.2,3 Cutaneous pPTLD presents as red to purple papules, plaques, and nodules often involving lower extremities, head, and trunk. Interestingly, we report pPTLD in a unique annular configuration encircling an OLT scar. Only 1 report of cutaneous diffuse large B-cell lymphoma presenting with an annular configuration has been reported.4 Histologically, pPTLD presents as a diffuse infiltrate of atypical lymphoid cells with plasmablastic features of eccentric nuclei, clumpy chromatin, prominent central nucleoli, and abundant basophilic cytoplasm. The CD20 stain is negative to weak, CD138 staining is positive, and monoclonality is seen with kappa or lambda light chain restriction.Most cases of B-cell PTLD are associated with primary infection or reactivation of EBV. It is unclear if the present patient’s pathology was related to EBV. In-situ hybridization stains of skin and stomach biopsies were negative, but the patient had a history of EBV infection (positive immunoglobulin G serology), which cannot be used to definitively determine EBV cause.5Initial treatment of PTLD is immune suppression reduction, but further steps are unclear. One study suggests that immune suppression reduction combined with local therapy is insufficient to prevent disease progression, but immune suppression reduction combined with systemic chemotherapy can result in durable, complete remissions.6Cases of pPTLD are particularly aggressive with poor prognosis.7 Progression and survival statistics for pPTLD are unavailable, but data can be extrapolated from plasmablastic lymphoma arising in patients with immunosuppression because of HIV, thought to be a similar disease differentiated by immunosuppression type.6,8 In these patients, 53% reported disease progression with an average survival of 15 months.9 A negative test for EBV, gene translocations, and multisite disease further increase the risk for partial or no remission. Multisite disease is associated with increased rates of mortality compared with single-site disease.1,7A case of pPTLD is a rare and serious complication of solid organ transplant that rarely involves the skin and is associated with high recurrence and mortality. We recommend early decrease and cessation, when possible, of intensive immunosuppression and initiation of intensive treatment owing to the disorder’s highly aggressive nature.
Dermatology
A man in his 50s presented with an abdominal skin eruption of 2 days’ duration. His medical history included hepatitis C, cirrhosis, and an orthotopic liver transplant (OLT) 3 years prior that was subsequently treated with tacrolimus, 5 mg, and mycophenolate, 750 mg, twice daily. Approximately 2½ years after the transplant, the patient developed gastric outlet obstruction secondary to an infiltrative gastric wall mass. The gastric wall mass was found to be associated with plasmablastic posttransplantation lymphoproliferative disorder (pPTLD). He was subsequently instructed to stop use of mycophenolate and decrease use of tacrolimus to 0.25 mg daily, then treated with 1 cycle of CHOP chemotherapy (cyclophosphamide, doxorubicin hydrochloride, vincristine sulfate, and prednisone) and lenalidomide, resulting in symptomatic and radiologic improvement. Five weeks after treatment with CHOP chemotherapy, the patient presented with an acute-onset abdominal skin eruption without fever or pain. A computed tomographic scan of the abdomen and pelvis showed possible abdominal wall cellulitis, so treatment with piperacillin-tazobactam and vancomycin was empirically started. Physical examination revealed confluent indurated violaceous papules and plaques coalescing into an annular pattern encircling the healed OLT incisional scar with central sparing (Figure 1A). A 4-mm punch biopsy specimen was obtained for histopathological evaluation (Figure 1B and C) and bacterial and fungal cultures.A, Confluent, indurated, violaceous papules and plaques coalescing into an annular pattern encircling the well-healed orthotopic liver transplant incisional scar. B, Hematoxylin-eosin staining of the punch biopsy shows an atypical, hyperchromatic infiltrate present throughout the superficial and deep dermis. C, High magnification reveals crushed but markedly atypical-appearing lymphoid cells with nuclear hyperchromasia and scant amounts of cytoplasm. Multiple mitotic and apoptotic figures are easily identified.
what is your diagnosis?
What is your diagnosis?
Posttransplantation lymphoproliferative disorder
Deep fungal infection
Reactive granulomatous disorder
Cutaneous T-cell lymphoma
a
0
1
1
1
male
0
0
55
51-60
null
543
original
https://jamanetwork.com/journals/jamaneurology/fullarticle/2725382
A 13-year-old boy with a longstanding history of gait imbalance presented with 2 episodes of acute-onset left hemibody weakness and dysarthria without changes in sensorium in a 24-hour period. Symptoms lasted approximately 60 minutes each before completely resolving; prior to the day of the events, he had never experienced similar phenomena. Initial examination following the second event revealed mild-to-moderate dysarthria, pes planovalgus and tight achilles tendons bilaterally, and decreased vibratory and fine touch sensation from the great toe up to the mid shin bilaterally. Reflexes were absent at the patella and achilles bilaterally. Bilateral dorsiflexion weakness was present (4 of 5). There were otherwise no motor deficits, muscle atrophy, or lateralizing neurologic abnormalities.On arrival, the patient underwent a stroke/transient ischemic attack workup. Results of a comprehensive metabolic panel, complete blood cell count, erythrocyte sedimentation rate, C-reactive protein, and a coagulation panel were within normal limits. Computed tomography angiogram demonstrated no evidence of extra or intracranial dissection. Magnetic resonance imaging of the brain with and without contrast (Figure) demonstrated symmetric, bilateral, restricted diffusion, with T2 hyperintensities in the supratentorial white matter and corpus callosum. Lumbar puncture was obtained and revealed normal cell counts and no oligoclonal bands or evidence of infection.A, Diffusion-weighted imaging sequence demonstrating bilateral, symmetric, restricted diffusion of the supratentorial white matter (corresponding hypointensity noted on apparent diffusion coefficient sequences). B, T2 sequence demonstrating bilateral and symmetric increased signal in the supratenorial white matter–sparing U-fibers. The patient had reportedly always walked on the outside of his feet and had been considered clumsy when compared with other children but had no history of motor delays. Family history revealed a mother with diminished reflexes, hand tremor, and balance issues with a prior inconclusive electromyogram and nerve conduction study. A younger brother with attention-deficit/hyperactivity disorder also had similar gait abnormalities. What Is Your Diagnosis?
Mitochondrial encephalopathy, lactic acidosis, and strokelike episodes
Metachromatic leukodystrophy
X-linked Charcot-Marie-Tooth disease
Pediatric-onset multiple sclerosis
C. X-linked Charcot-Marie-Tooth disease
C
X-linked Charcot-Marie-Tooth disease
The patient had no further events following his admission and neurodiagnostic workup. His examination was consistent with peripheral nerve pathology (diminished reflexes and decrements in sensorium), which appeared to be longstanding as demonstrated by pes planovalgus and tight Achilles tendons. The patient’s superimposed acute events were felt to be reflective of a strokelike phenomenon, given the duration and pattern of neurologic symptoms.In reviewing the patient’s case, examination, and neuroimaging, it was determined that the patient’s clinical findings fit most closely with a diagnosis of Charcot-Marie-Tooth disease (CMTX). The imaging findings, particularly with prominent bilateral and symmetric restricted diffusion in the supratentorial white matter, were thought to be more characteristic of CMTX-related strokelike episode and would not be typical for leukodystrophies (specifically, metachromatic leukodystrophy, X-linked adrenoleukodystrophy, and Alexander disease). Given the symmetric pattern of involvement, mitochondrial disorders were considered although were thought to be less likely given the patient’s normal growth and development and lack of lactic acidosis and other laboratory abnormalities. Acute disseminated encephalomyelitis was considered, but the large-scale confluent lesions involving only the white matter were considered unusual as would be the symmetry of this pattern. Multiple sclerosis and neuromyelitis optica typically do not exhibit these patterns and were thought to be very low on the differential. Myelin oligodendrocyte antibody spectrum disorders can present with large T2-hyperintense lesions but are rarely symmetric or isolated to the white matter only. Although a classic mimic in nearly all central demyelinating disorders, central nervous system lymphoma was lower on the differential based on symmetry, age of patient, and lack of other findings.Ultimately, the patient was discharged at full neurologic capacity. He had an outpatient neurogenetics workup that identified a hemizygous mutation in gap junction β-1 (GJB1) c.227 t > G. Loss-of-function variants in the GJB1 gene are associated with type 1 CMTX (CMTX1) and thus presumed to be a pathogenic variant.As noted previously, CMTX1 is an inherited neuropathy caused by mutation of the GJB1 gene. This gene encodes for connexin 32, which is a gap junction protein that is expressed by Schwann cells in the peripheral nervous system.1 Owing to X-linked inheritance, this disorder mostly affects male patients, although female carries may also be symptomatic, as is possible in this case. Transient neurologic events, also described as strokelike episodes, have been reported in the literature and are often associated with symmetric white matter abnormalities.1-7 The pathophysiology underlying these events and white matter changes is unknown, although it is presumed to be associated with connexin 32 expression in oligodendrocytes in the central nervous system.8 Because most patients with CMTX1 do not have long-term central neurologic phenomenon, there is great debate regarding whether other connexin subtypes may provide compensatory support at gap junctions in patients.9,10Clinically, the strokelike episodes observed in CMTX1 are heterogenous and include transient generalized weakness resembling periodic paralysis, ataxia, dysarthria, or combinations of these deficits.1,6,7 The fluctuation in these neurologic disturbances and magnetic resonance imaging findings should prompt suspicion of the diagnosis. The central nervous system phenotype of CMTX1 disease has a favorable prognosis with respect to central nervous system function, and recognition will avoid unnecessary investigations and potentially harmful therapeutic intervention. There is no clear evidence for steroids or other immunomodulatory therapy in CMTX1-associated transient neurologic phenomenon, with clinical and imaging findings usually being self-limited.6,7This case highlights one of the rarer strokelike presentations in a patient with a neuromuscular disorder. The presence of long-standing peripheral nerve pathology should prompt interrogation of other etiologies to strokelike presentations in children and young adults.
Neurology
A 13-year-old boy with a longstanding history of gait imbalance presented with 2 episodes of acute-onset left hemibody weakness and dysarthria without changes in sensorium in a 24-hour period. Symptoms lasted approximately 60 minutes each before completely resolving; prior to the day of the events, he had never experienced similar phenomena. Initial examination following the second event revealed mild-to-moderate dysarthria, pes planovalgus and tight achilles tendons bilaterally, and decreased vibratory and fine touch sensation from the great toe up to the mid shin bilaterally. Reflexes were absent at the patella and achilles bilaterally. Bilateral dorsiflexion weakness was present (4 of 5). There were otherwise no motor deficits, muscle atrophy, or lateralizing neurologic abnormalities.On arrival, the patient underwent a stroke/transient ischemic attack workup. Results of a comprehensive metabolic panel, complete blood cell count, erythrocyte sedimentation rate, C-reactive protein, and a coagulation panel were within normal limits. Computed tomography angiogram demonstrated no evidence of extra or intracranial dissection. Magnetic resonance imaging of the brain with and without contrast (Figure) demonstrated symmetric, bilateral, restricted diffusion, with T2 hyperintensities in the supratentorial white matter and corpus callosum. Lumbar puncture was obtained and revealed normal cell counts and no oligoclonal bands or evidence of infection.A, Diffusion-weighted imaging sequence demonstrating bilateral, symmetric, restricted diffusion of the supratentorial white matter (corresponding hypointensity noted on apparent diffusion coefficient sequences). B, T2 sequence demonstrating bilateral and symmetric increased signal in the supratenorial white matter–sparing U-fibers. The patient had reportedly always walked on the outside of his feet and had been considered clumsy when compared with other children but had no history of motor delays. Family history revealed a mother with diminished reflexes, hand tremor, and balance issues with a prior inconclusive electromyogram and nerve conduction study. A younger brother with attention-deficit/hyperactivity disorder also had similar gait abnormalities.
what is your diagnosis?
What is your diagnosis?
Metachromatic leukodystrophy
Mitochondrial encephalopathy, lactic acidosis, and strokelike episodes
X-linked Charcot-Marie-Tooth disease
Pediatric-onset multiple sclerosis
c
1
1
1
1
male
0
0
13
11-20
White
544
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2726035
A 43-year-old healthy human leukocyte antigen (HLA) B27–positive man was referred to the retina clinic with progressively worsening blurry vision in both of his eyes. His right eye had experienced an episode of blurry vision 4 months previously that had spontaneously improved over a few weeks. Two weeks before presentation, he experienced blurry vision in his left eye and received a diagnosis of vasculitis based on fluorescein angiography (FA) findings. He was referred to a rheumatologist and received high-dose oral prednisone therapy for presumed autoimmune vasculitis. Over the next 2 weeks, he experienced a rapid worsening of vision in both eyes and presented to the Illinois Eye and Ear Infirmary. During an interview, he noted that the laboratory workup results had recently come back as notable for a mildly reactive fluorescent treponemal antibody absorption test. In the absence of other signs or symptoms of syphilis, he was scheduled to undergo a lumbar puncture later that day.On presentation, his visual acuity was counting fingers OD and 20/300 OS, with manifest refraction to 20/60 OS only. He had no afferent pupillary defect and his intraocular pressures were healthy. The anterior segment examination yielded normal results without evidence of anterior chamber inflammation. A dilated fundus examination was notable for mild vitritis of both eyes, mild hyperemia of the right optic nerve, and a large white lesion that encompassed the nerve and macula of both eyes. Fluorescein angiography results revealed multiple hyperfluorescent lesions in late frames (Figure 1).A, Color fundus photograph of the left eye shows a large placoid lesion in the macula. B, Late fluorescein angiography. showing multiple areas of hyperfluorescence in the macula and periphery with peripheral vascular leakage. What Would You Do Next?
Perform bilateral posterior sub-Tenon triamcinolone injections
Increase the dose of oral prednisone
Initiate antiviral therapy
Initiate intravenous penicillin therapy
Acute syphilitic posterior placoid chorioretinitis
D
Initiate intravenous penicillin therapy
The patient presented with vasculitis in the setting of HLA-B27 positivity and had been presumed to have autoimmune vasculitis. Although patients with HLA-B27 may present with isolated ocular findings, they typically have acute-onset unilateral anterior uveitis.1 Posterior uveitis may develop with concurrent anterior or panuveitis and commonly results in extensive phlebitis that is associated with optic disc leakage and cystoid macular edema.2 Isolated retinal vasculitis is rare but has been reported in patients with inflammatory bowel disease.1 Therefore, this patient’s presentation is atypical for HLA-B27-associated uveitis and should have prompted further investigation for infectious etiologies before treatment with systemic steroids. A local steroid injection, such as posterior sub-Tenon triamcinolone, is likewise contraindicated when infection has not yet been ruled out.Viral retinopathy due to herpes simplex virus, varicella-zoster virus, and cytomegalovirus may present with vitreous inflammation. However, the patient lacked retinitis, which typically begins as multifocal areas of retinal whitening in the periphery, and retinal hemorrhages, which are characteristic of cytomegalovirus retinitis.3 Therefore, antiviral therapy would not treat his symptoms.Ocular syphilis can manifest with almost any symptom and ocular finding, including retinal vasculitis.4 Centers for Disease Control and Prevention guidelines define ocular syphilis as affecting any person with clinical symptoms or signs consistent with ocular disease, including uveitis and diminished vision with any stage of syphilis.5 In this patient, a reactive fluorescent treponemal antibody absorption in the setting of decreased vision qualifies as ocular syphilis. This patient’s fundus findings and FA on presentation are typical for acute syphilitic posterior placoid chorioretinitis with the large, yellowish, placoid outer retinal lesion in the macula.6 Fluorescein angiography results show progressive hyperfluorescence in the lesion area, often accompanied by scattered focal hypofluorescence known as leopard spotting.6 Optical coherence tomography may show disruption of the ellipsoid zone, hyperreflective thickening of the retinal pigment epithelium, and subretinal fluid (Figure 2).7 The lesion areas may appear hypofluorescent on indocyanine green angiography and hyperautofluorescent on fundus autofluorescence.6Optical coherence tomography of the left eye shows a loss of the ellipsoid zone and retinal pigment epithelium stippling.Ocular syphilis should be managed according to treatment recommendations for neurosyphilis, which consist of intravenous aqueous crystalline penicillin G or intramuscular procaine penicillin with probenecid for 10 to 14 days.5 In patients who are allergic to penicillin, intravenous or intramuscular ceftriaxone for 10 to 14 days can be used, although some clinicians advocate penicillin desensitization prior to treatment with penicillin.5 Oral or topical steroids may be used concurrently with penicillin treatment as an adjunct to reduce infection-induced inflammation, but oral prednisone alone is not recommended.7 Reports from small case series suggest that intravitreal injection of triamcinolone should be avoided.6 Testing for the human immunodeficiency virus (HIV) often is recommended for patients because of the risk of concurrent infection.5The patient completed a 14-day course of intravenous penicillin therapy and noted immediate visual improvement. His rapid plasma reagin test results showed an elevated titer of 1:64, but his lumbar puncture study results were healthy. He had a negative result for HIV. At 2 months after presentation, his visual acuity was 20/40 OD and 20/20 OS with resolution of the placoid lesions on examination and reconstitution of the ellipsoid zone on optical coherence tomography.
Ophthalmology
A 43-year-old healthy human leukocyte antigen (HLA) B27–positive man was referred to the retina clinic with progressively worsening blurry vision in both of his eyes. His right eye had experienced an episode of blurry vision 4 months previously that had spontaneously improved over a few weeks. Two weeks before presentation, he experienced blurry vision in his left eye and received a diagnosis of vasculitis based on fluorescein angiography (FA) findings. He was referred to a rheumatologist and received high-dose oral prednisone therapy for presumed autoimmune vasculitis. Over the next 2 weeks, he experienced a rapid worsening of vision in both eyes and presented to the Illinois Eye and Ear Infirmary. During an interview, he noted that the laboratory workup results had recently come back as notable for a mildly reactive fluorescent treponemal antibody absorption test. In the absence of other signs or symptoms of syphilis, he was scheduled to undergo a lumbar puncture later that day.On presentation, his visual acuity was counting fingers OD and 20/300 OS, with manifest refraction to 20/60 OS only. He had no afferent pupillary defect and his intraocular pressures were healthy. The anterior segment examination yielded normal results without evidence of anterior chamber inflammation. A dilated fundus examination was notable for mild vitritis of both eyes, mild hyperemia of the right optic nerve, and a large white lesion that encompassed the nerve and macula of both eyes. Fluorescein angiography results revealed multiple hyperfluorescent lesions in late frames (Figure 1).A, Color fundus photograph of the left eye shows a large placoid lesion in the macula. B, Late fluorescein angiography. showing multiple areas of hyperfluorescence in the macula and periphery with peripheral vascular leakage.
what would you do next?
What would you do next?
Perform bilateral posterior sub-Tenon triamcinolone injections
Initiate antiviral therapy
Initiate intravenous penicillin therapy
Increase the dose of oral prednisone
c
1
1
1
1
male
0
0
43
41-50
White
545
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2726036
A woman in her 30s was referred for evaluation of bilateral papilledema and progressive visual loss in the right eye, which deteriorated to blindness in 3 months. She denied any other neurologic symptoms, such as headache, nausea, vomiting, seizures, and paresis. She had a 4-year history of Behçet disease (BD), and her condition had been stabilized with thalidomide. Her optic neuropathy of BD had been treated with high-dose intravenous methylprednisolone for 4 days followed by oral prednisone for a month without improvement.Her best-corrected visual acuity was no light perception OD and 20/20 OS. Intraocular pressure was 21 mm Hg OD and 19 mm Hg OS. Fundus examination revealed papilledema in both eyes (Figure 1A). No obvious sign of optic disc ischemia or active uveitis was observed by fundus fluorescein angiography bilaterally. Humphrey visual fields showed complete visual field loss in the right eye and peripheral visual field defects in the left eye. Her anterior segment was normal in both eyes except for relative afferent pupillary defect in the right eye. Cranial nerve examination was otherwise unremarkable. Magnetic resonance imaging of the brain showed no remarkable abnormalities except for partially empty sella (Figure 1B). She had normal blood pressure levels. Results of laboratory testing regarding systemic infection were negative, and her serum D-dimer level was normal.A, Fundus photograph of the right eye indicated papilledema, disc congestion, venous distension, and macular exudates. B, Sagittal T1-weighted magnetic resonance imaging showed partially empty sella (arrowhead).Obtain cerebral magnetic resonance venography or computed tomographic venography What Would You Do Next?
Add immunosuppressant to corticosteroid therapy
Increase the prednisone dose
Obtain cerebral magnetic resonance venography or computed tomographic venography
Perform catheter cerebral angiography
Cerebral venous thrombosis
C
Obtain cerebral magnetic resonance venography or computed tomographic venography
Optic neuropathy of BD usually occurs with systemic flare-up and responds well to steroids.1 This patient showed no signs of relapse of BD, and her vision continued to deteriorate despite the use of corticosteroids. Therefore, it is not proper to use immunosuppressants and corticosteroids (choices A and B) before a definite diagnosis. Although the patient lacked neurological symptoms other than visual loss, the bilateral papilledema and partially empty sella indicated intracranial hypertension, the possible causes of which include arteriovenous malformations, intracranial mass lesions, obstruction to venous drainage, decreased flow through arachnoid granulations, and idiopathic intracranial hypertension.2 Moreover, a negative magnetic resonance imaging result cannot fully rule out cerebral vascular abnormalities, among which cerebral venous thrombosis (CVT) accounts for 9.4% of the presumed idiopathic intracranial hypertension.3 In suspected CVT, magnetic resonance venography or computed tomographic venography (choice C) is recommended when the magnetic resonance imaging result is negative.4 Therefore, a cerebral magnetic resonance venography of the patient was performed, showing thromboses of the superior sagittal sinus and the right transverse and sigmoid sinus (Figure 2). Draining cerebrospinal fluid is suggested when intracranial hypertension results in vision-threatening papilledema.5 Therefore, a lumber puncture was performed to confirm intracranial hypertension (showing opening pressure of 30.5 cm H2O) and to remove cerebrospinal fluid. Acetazolamide was also initiated to lower the intracranial pressure. Anticoagulation therapy of heparin was started to facilitate recanalization and to prevent recurrent CVT and other venous thrombosis. Endovascular thrombolysis was performed to prevent visual loss of the left eye. The catheter cerebral angiography (choice D), which is invasive, is only considered when magnetic resonance venography or computed tomography venography results are inconclusive or an endovascular procedure is performed.4 Therefore, it was performed along with endovascular thrombolysis to visualize thrombosed veins.Cerebral magnetic resonance venography showed thromboses of the superior sagittal sinus, the right transverse sinus and the right sigmoid sinus (white arrowheads), and stenosis of the left sigmoid sinus (yellow arrowhead).Cerebral venous thrombosis is a rare cerebrovascular disease, with an annual incidence of 3 to 4 cases per million.6 Headache is the most common symptom, presenting in 90% of patients, while focal seizures, paresis, impaired consciousness, and visual disturbance are less common.6 Cerebral venous thrombosis only presenting visual loss is extremely rare.7 The lack of headaches and other neurological symptoms in this case might be caused by the gradually raised intracranial pressure and compensatory anatomical adjustments. Empty sella might be caused by the persistent intracranial hypertension, which pressed the subarachnoid space through the incomplete sellar diaphragm. The normal D-dimer level might be attributed to the prolonged duration of symptoms (more than 1 week).8 Risk factors of CVT were miscellaneous, including prothrombotic conditions, infections, trauma, cancer, and systemic diseases such as BD.6 This patient had BD and had taken thalidomide, both of which are predisposing factors of CVT.6,9One month following thrombolysis, the patient’s bilateral papilledema resolved. Three months later, the visual field of her left eye became normal, while the blindness of her right eye was irreversible. This case reminded us that when empty sella meets bilateral papilledema, intracranial hypertension should be considered even in the absence of neurologic symptoms, and CVT can be one of the possible diagnoses.
Ophthalmology
A woman in her 30s was referred for evaluation of bilateral papilledema and progressive visual loss in the right eye, which deteriorated to blindness in 3 months. She denied any other neurologic symptoms, such as headache, nausea, vomiting, seizures, and paresis. She had a 4-year history of Behçet disease (BD), and her condition had been stabilized with thalidomide. Her optic neuropathy of BD had been treated with high-dose intravenous methylprednisolone for 4 days followed by oral prednisone for a month without improvement.Her best-corrected visual acuity was no light perception OD and 20/20 OS. Intraocular pressure was 21 mm Hg OD and 19 mm Hg OS. Fundus examination revealed papilledema in both eyes (Figure 1A). No obvious sign of optic disc ischemia or active uveitis was observed by fundus fluorescein angiography bilaterally. Humphrey visual fields showed complete visual field loss in the right eye and peripheral visual field defects in the left eye. Her anterior segment was normal in both eyes except for relative afferent pupillary defect in the right eye. Cranial nerve examination was otherwise unremarkable. Magnetic resonance imaging of the brain showed no remarkable abnormalities except for partially empty sella (Figure 1B). She had normal blood pressure levels. Results of laboratory testing regarding systemic infection were negative, and her serum D-dimer level was normal.A, Fundus photograph of the right eye indicated papilledema, disc congestion, venous distension, and macular exudates. B, Sagittal T1-weighted magnetic resonance imaging showed partially empty sella (arrowhead).Obtain cerebral magnetic resonance venography or computed tomographic venography
what would you do next?
What would you do next?
Add immunosuppressant to corticosteroid therapy
Increase the prednisone dose
Perform catheter cerebral angiography
Obtain cerebral magnetic resonance venography or computed tomographic venography
d
1
1
1
1
female
0
0
4
0-10
null
546
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2726698
An 8-year-old girl was referred to the vitreoretinal service for evaluation of bilateral macular lesions noted by her pediatric ophthalmologist. Bilateral lateral rectus muscle recession for exotropia was performed 4 years before this presentation. Medical history was otherwise unremarkable. The patient was born full term after a normal pregnancy and delivery, with normal growth assessments. The patient was performing well in school.On examination, her best-corrected visual acuity was 20/40 OD and 20/40 OS, which was stable from previous visits. Her pupils were round and reactive. There was a residual intermittent exotropia of 4 prism diopters bilaterally. Findings on slitlamp examination of the anterior segment were unremarkable. A single, 1-cm, oval, lightly pigmented macule with irregular borders was noted on her right upper arm. Dilated fundus examination revealed eccentric, yellow-clear, preretinal lesions in the maculas of both eyes. Optical coherence tomography revealed hyperreflective areas on the surface of the macula with a spiculated appearance, projecting anteriorly into the vitreous. There was an increase in retinal thickness and loss of the foveal contour in both eyes (Figure).Optical coherence tomography of the right eye (A) showing an atypical epiretinal membrane with anterior projection into the vitreous and hazy borders and the left eye (B) showing a similar epiretinal membrane with mild anterior projection and hazy borders.Obtain a family medical history and genetic testingRecommend pars plana vitrectomy with epiretinal membrane peel What Would You Do Next?
Obtain a family medical history and genetic testing
Recommend pars plana vitrectomy with epiretinal membrane peel
Observe the lesions
Give an intravitreal ocriplasmin injection
Neurofibromatosis 2–related epiretinal membranes
A
Obtain a family medical history and genetic testing
After further questioning, the patient’s father had a history of neurofibromatosis type 2 (NF2) and died in his early 40s. Her younger brother was previously diagnosed with bilateral parasellar masses that extended into the right orbital apex, causing a neurogenic ptosis of the right upper eyelid. The brother subsequently tested positive for NF2 gene with deletion of exons 1 through 7. Genetic testing results obtained for the patient were positive for an abnormality in the NF2 gene with deletion of the NF2 exons 1 through 7. Observation (choice C) and surgical intervention (choice B) are both potential management options for patients with epiretinal membranes. However, an epiretinal membrane in a child should prompt further medical history, family medical history, and potential genetic testing (choice A) for systemic disorders. Ocriplasmin injection (choice D) would not be the correct treatment for removal of epiretinal membranes.An autosomal dominant inherited disorder, neurofibromatosis type 2, is caused by mutations in the NF2 gene located on chromosome 22.1 Abnormalities in the NF2 gene, which produces the tumor suppressor merlin protein, lead to the disease. Individuals with the disease are predisposed to tumors of the nervous system, including bilateral vestibular schwannomas, meningiomas, and spinal tumors. Although thought to be rare, the incidence of NF2 may be as high as 1 in 25 000 persons.2 Skin lesions are an important diagnostic criterion for neurofibromatosis type 1; however, skin lesions can also be found in patients with NF2.3 Ophthalmic manifestations can be seen including cataracts, epiretinal membranes, retinal hamartomas, and optic nerve meningiomas.Epiretinal membranes are a common finding in NF2 and are usually solitary within each eye and located in the macula. They have been described in children as young as 4 years and are likely to be congenital.4 Many epiretinal membranes in NF2 have a spiculated appearance with curled edges, projecting anteriorly into the vitreous and best visualized using optical coherence tomography.5There has been controversy about whether these epiretinal membranes could represent a mild form of combined hamartomas of the retinal and retina pigment epithelium. Although both are similar congenital macular lesions, there are certain optical coherence tomographic characteristics that can differentiate them. Combined hamartomas of the retinal and retina pigment epithelium lesions show greater retinal disorganization, photoreceptor attenuation, and loss of the foveal depression compared with NF2 epiretinal membranes.5Although observation of NF2-related epiretinal membranes is an option if the vision loss is mild, recent studies have shown gains in vision with early pars plana vitrectomy and epiretinal membrane peel.6,7 Bonnin et al7 reported operating on 13 patients with epiretinal membranes who were 12 years or younger. Of the 13 patients, 12 had improvement in visual acuity and the corrected visual acuity improved from an average of 20/160 to 20/40.The patient has continued to follow up with a pediatric neurologist, who has recommended magnetic resonance imaging of the brain to rule out central nervous system lesions. The family reported that they prefer observation of the retinal lesions and will consider surgery if the patient’s vision worsens.
Ophthalmology
An 8-year-old girl was referred to the vitreoretinal service for evaluation of bilateral macular lesions noted by her pediatric ophthalmologist. Bilateral lateral rectus muscle recession for exotropia was performed 4 years before this presentation. Medical history was otherwise unremarkable. The patient was born full term after a normal pregnancy and delivery, with normal growth assessments. The patient was performing well in school.On examination, her best-corrected visual acuity was 20/40 OD and 20/40 OS, which was stable from previous visits. Her pupils were round and reactive. There was a residual intermittent exotropia of 4 prism diopters bilaterally. Findings on slitlamp examination of the anterior segment were unremarkable. A single, 1-cm, oval, lightly pigmented macule with irregular borders was noted on her right upper arm. Dilated fundus examination revealed eccentric, yellow-clear, preretinal lesions in the maculas of both eyes. Optical coherence tomography revealed hyperreflective areas on the surface of the macula with a spiculated appearance, projecting anteriorly into the vitreous. There was an increase in retinal thickness and loss of the foveal contour in both eyes (Figure).Optical coherence tomography of the right eye (A) showing an atypical epiretinal membrane with anterior projection into the vitreous and hazy borders and the left eye (B) showing a similar epiretinal membrane with mild anterior projection and hazy borders.Obtain a family medical history and genetic testingRecommend pars plana vitrectomy with epiretinal membrane peel
what would you do next?
What would you do next?
Give an intravitreal ocriplasmin injection
Obtain a family medical history and genetic testing
Observe the lesions
Recommend pars plana vitrectomy with epiretinal membrane peel
b
0
1
1
1
female
0
0
8
0-10
null
547
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2726699
A woman in her mid-80s presented with ptosis and blurred vision in the right eye. The ptosis was first noticed 3 weeks prior to presentation and had gradually worsened. She denied any acute changes in her health and review of her systems was unremarkable. The patient had an ocular history of bilateral cataract surgery in 2016, a medical history of thyroid cancer with thyroidectomy in 1990, and stage IIIc lobular carcinoma of the right breast with mastectomy of the right breast in 2008.On examination, her visual acuity was 20/80 OD and 20/30 OS. Pupils were equal in size and reactive without an afferent pupillary defect. Intraocular pressures were 21 mm Hg OD and 16 mm Hg OS. Assessment of the extraocular motility demonstrated moderate limitation to up gaze of the right eye and full ductions of the left eye. External examination confirmed right upper eyelid ptosis without substantial proptosis. On slitlamp examination, eversion of the right upper eyelid revealed a solid, superior bulbar conjunctival mass measuring 10 mm in diameter (Figure 1A). B-scan ultrasonography demonstrated an extraconal, noncompressible lesion of orbital tissue (Figure 1B). A computed tomographic scan showed nonspecific, increased soft-tissue density in the superior and anterior orbit.A, External photograph of the right eye demonstrating an elevated conjunctival lesion. B, Ultrasonographic image of the conjunctival lesion; the arrowhead indicates the tumor location.Complete blood cell count, basic metabolic panel, and liver function tests What Would You Do Next?
Topical corticosteroid eyedrops
Conjunctival biopsy
Complete blood cell count, basic metabolic panel, and liver function tests
Serum and urine electrophoresis
Breast cancer metastasis to the conjunctiva
B
Conjunctival biopsy
The differential diagnosis for an acquired conjunctival lesion is broad.1,2 The lesion can be classified as epithelial, subepithelial, pigmented, or inflammatory. Acquired epithelial lesions include squamous cell carcinoma and papilloma. Subepithelial lesions range from benign causes, such as lacrimal gland duct cysts and myxomas, to infiltrative conditions, such as metastatic carcinoma. Pigmented lesions typically consist of nevi or malignant melanoma, and inflammatory lesions can stem from conditions such as sarcoidosis.2When a patient presents with a conjunctival lesion, diagnostic imaging is important. Given the concern for an orbital mass in this case, orbital imaging was the initial step of evaluation. Findings of the computed tomographic scan that was performed were nonspecific, as was B-scan ultrasonography. Given the lack of diagnostic information from imaging, a conjunctival biopsy was performed to evaluate the lesion, which is important given the patient’s history of both thyroid and breast cancer.3 Although a systemic workup, including complete blood cell count and basic metabolic panel as well as serum and urine electrophoresis, are important in the evaluation for lymphoma or amyloidosis, the conjunctival biopsy was the next step given its ability to provide a more specific diagnosis.4,5 Histopathologic testing results revealed tumor cells with estrogen-receptor positive, progesterone-receptor positive, and ERBB2-negative, confirming the diagnosis of metastatic breast carcinoma (Figure 2). Topical corticosteroids are useful in inflammatory conditions, but would not be indicated in this patient.Histopathologic examination of a conjunctival lesion revealing tumor cells (hematoxylin-eosin, original magnification ×400).The most common primary tumor to give rise to ocular metastasis in the female population is breast carcinoma.6 The incidence of ocular metastasis among patients with breast cancer is approximately 8% to 10%, typically occurring 2 to 5 years after initial diagnosis of the primary tumor.7 Metastatic lesions to the conjunctiva are uncommon and indicate poor prognosis.3 Symptoms include pain, chemosis, and foreign body sensation.3,6-9 In this case, the patient noticed ptosis, but was not aware of the conjunctival lesion as the lesion was not visible until the upper eyelid was everted.Most metastatic conjunctival lesions regress with external-beam radiotherapy, excisional biopsy, and/or chemotherapy.3,7 Because ocular disease presents when the systemic malignant lesion is advanced, the prognosis tends to be poor.2 The mean reported survival time after diagnosis of a conjunctival mass varies, ranging from 3 weeks to 26 months.3,6 Therefore, it is important to recognize conjunctival metastasis upon examination to provide timely and adequate therapeutic intervention.6,8 In addition, our case highlights the importance of obtaining a thorough history, because distant metastases can present many years after the initial diagnosis of nonocular malignant diseases.The patient underwent an excisional conjunctival biopsy on the same day as presentation, confirming the diagnosis of metastatic breast carcinoma. Further systemic evaluation revealed additional metastases to the cervical, lumbar, and thoracic spine; sacrum and bilateral iliac bones; adrenal glands; and lymph nodes. She underwent antiestrogen therapy with an adjuvant cyclin-kinase inhibitor. Five months after presentation of the ocular symptoms, positron emission tomographic scan showed full remission of the malignant lesion.
Ophthalmology
A woman in her mid-80s presented with ptosis and blurred vision in the right eye. The ptosis was first noticed 3 weeks prior to presentation and had gradually worsened. She denied any acute changes in her health and review of her systems was unremarkable. The patient had an ocular history of bilateral cataract surgery in 2016, a medical history of thyroid cancer with thyroidectomy in 1990, and stage IIIc lobular carcinoma of the right breast with mastectomy of the right breast in 2008.On examination, her visual acuity was 20/80 OD and 20/30 OS. Pupils were equal in size and reactive without an afferent pupillary defect. Intraocular pressures were 21 mm Hg OD and 16 mm Hg OS. Assessment of the extraocular motility demonstrated moderate limitation to up gaze of the right eye and full ductions of the left eye. External examination confirmed right upper eyelid ptosis without substantial proptosis. On slitlamp examination, eversion of the right upper eyelid revealed a solid, superior bulbar conjunctival mass measuring 10 mm in diameter (Figure 1A). B-scan ultrasonography demonstrated an extraconal, noncompressible lesion of orbital tissue (Figure 1B). A computed tomographic scan showed nonspecific, increased soft-tissue density in the superior and anterior orbit.A, External photograph of the right eye demonstrating an elevated conjunctival lesion. B, Ultrasonographic image of the conjunctival lesion; the arrowhead indicates the tumor location.Complete blood cell count, basic metabolic panel, and liver function tests
what would you do next?
What would you do next?
Topical corticosteroid eyedrops
Conjunctival biopsy
Serum and urine electrophoresis
Complete blood cell count, basic metabolic panel, and liver function tests
b
0
1
1
1
female
0
0
85
81-90
null
548
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2727508
A 53-year-old male was referred for evaluation of a nonhealing corneal ulcer. He had not received medical care in 20 years and had no known medical diagnoses, but his social history was notable for consumption of approximately 12 alcoholic beverages per day. His ocular history was notable for left-eye enucleation from recent trauma. He initially presented to his local ophthalmologist with right eye redness, pain, and photophobia for 1 week. He was diagnosed with a corneal ulcer. Bacterial (aerobic and anaerobic) and fungal cultures were obtained, and the patient started receiving topical moxifloxacin. The cultures were negative after 1 week, but the patient did not demonstrate signs of clinical improvement. A complete blood cell count, metabolic panel, and serum test results (for antinuclear antibodies, peripheral antineutrophil cytoplasmic antibodies, cytoplasmic antineutrophil cytoplasmic antibodies, rheumatoid factor, and anti–cyclic citrullinated peptide) were unremarkable. On referral, his visual acuity was 20/100 OD, and his intraocular pressure was 16 mm Hg. Anterior examination revealed a diffusely hazy cornea with a blunted light reflex. A large, well-demarcated epithelial defect with substantial underlying stromal loss was seen in the inferonasal periphery (Figure). The stroma was 40% thinned without infiltrate. The epithelium was heaped up along the lesion borders. Corneal sensation was diminished. The conjunctiva demonstrated hyperemia, which was more prominent around the corneal lesion, although there was no discharge. A dilated ophthalmoscopic examination was unremarkable. Repeated bacterial and fungal cultures were obtained.An external photograph showing a large, well-demarcated epithelial defect with underlying stromal loss.Obtain herpes simplex virus tissue culture and antigen detection testsBroaden antibiotic coverage to include vancomycin and tobramycin What Would You Do Next?
Use confocal microscopy to evaluate for cysts
Obtain herpes simplex virus tissue culture and antigen detection tests
Broaden antibiotic coverage to include vancomycin and tobramycin
Obtain serum vitamin A levels
Keratomalacia
D
Obtain serum vitamin A levels
The patient was sent to the emergency department for serum vitamin A testing (choice D), which revealed undetectable vitamin A levels (normal range, 30-80 μg/dL; to convert to micromoles per liter, multiply by 0.0349). The differential diagnosis for a nonhealing corneal ulcer may include inadequate antibiotic coverage, a nonbacterial infectious source, systemic autoimmune conditions, and epithelial wound-healing deficits.The patient’s history and presentation is consistent with vitamin A deficiency–associated keratomalacia. Patients with alcoholism may have protein and vitamin deficiencies, including vitamin A,1 because of both low dietary intake and decreased absorption. Nyctalopia is often the initial finding in vitamin A deficiency,2 although it was not present in this case. Vitamin A deficiency may also lead to ocular surface disease, such as conjunctival and corneal xerosis, and diffuse or focal keratin accumulation with a foamy appearance (Bitôt spots). There is a lack of mucin because goblet cells in the mucosal surface are greatly reduced.3 In advanced stages, keratomalacia may develop, with severe corneal ulceration and thinning, and this may ultimately result in diffuse corneal necrosis.Confocal microscopy to evaluate for cysts (choice A) to diagnose Acanthamoeba keratitis would not be the preferred answer. An infectious causative mechanism that is inadequately treated by a fourth-generation fluoroquinolone, such as Acanthamoeba keratitis, typically presents as a nonhealing corneal ulcer in a patient who wears contact lenses, often associated with radial keratoneuritis and/or a ring ulcer.Similarly, a herpes simplex virus tissue culture and antigen detection test (choice B) would not be the preferred choice. While herpes simplex virus epithelial keratitis may be considered in cases of nonhealing corneal ulcers, it is primarily a clinical diagnosis, and this patient lacked the dendritic ulcerations, subepithelial or deep stromal infiltrates, and corneal edema characteristic of this clinical entity. Decreased corneal sensation may suggest a neurotrophic causative mechanism, owing to diabetes, chemical burns, contact lens use, and chronic topical medications. However, this patient’s metabolic profile, including his glucose level, was unremarkable, and the history and examination did not reveal burns, contact lens use, or topical medications.Broadening antibiotic coverage (choice C) would not be the preferred answer. There is a low suspicion of infection, given the lack of injection, infiltrate, and discharge on examination. In addition, bacterial and fungal cultures were negative. While it is reasonable to repeat cultures, vancomycin and tobramycin may induce corneal epithelial toxicity, which may exacerbate the problem.The urgency of identifying vitamin A deficiency is associated with both systemic and ocular manifestations. Children with keratomalacia owing to vitamin A deficiency have a high mortality rate, with estimates up to 30% to 80%.4,5 Keratomalacia may progress to corneal perforation in a matter of days if left untreated.6 Furthermore, stromal loss is irreversible, resulting in permanent structural damage.7This patient started receiving high-dose oral vitamin A supplementation at 200 000 IU for the first 2 consecutive days, followed by another dose 1 week later.8 He also started receiving aggressive lubricating ointment, while decreasing topical moxifloxacin use. He had marked improvement in his right eye pain, vision, and corneal thinning. His conjunctival and corneal keratinization also improved dramatically within several days. He was referred to a primary care physician for further nutritional evaluation.
Ophthalmology
A 53-year-old male was referred for evaluation of a nonhealing corneal ulcer. He had not received medical care in 20 years and had no known medical diagnoses, but his social history was notable for consumption of approximately 12 alcoholic beverages per day. His ocular history was notable for left-eye enucleation from recent trauma. He initially presented to his local ophthalmologist with right eye redness, pain, and photophobia for 1 week. He was diagnosed with a corneal ulcer. Bacterial (aerobic and anaerobic) and fungal cultures were obtained, and the patient started receiving topical moxifloxacin. The cultures were negative after 1 week, but the patient did not demonstrate signs of clinical improvement. A complete blood cell count, metabolic panel, and serum test results (for antinuclear antibodies, peripheral antineutrophil cytoplasmic antibodies, cytoplasmic antineutrophil cytoplasmic antibodies, rheumatoid factor, and anti–cyclic citrullinated peptide) were unremarkable. On referral, his visual acuity was 20/100 OD, and his intraocular pressure was 16 mm Hg. Anterior examination revealed a diffusely hazy cornea with a blunted light reflex. A large, well-demarcated epithelial defect with substantial underlying stromal loss was seen in the inferonasal periphery (Figure). The stroma was 40% thinned without infiltrate. The epithelium was heaped up along the lesion borders. Corneal sensation was diminished. The conjunctiva demonstrated hyperemia, which was more prominent around the corneal lesion, although there was no discharge. A dilated ophthalmoscopic examination was unremarkable. Repeated bacterial and fungal cultures were obtained.An external photograph showing a large, well-demarcated epithelial defect with underlying stromal loss.Obtain herpes simplex virus tissue culture and antigen detection testsBroaden antibiotic coverage to include vancomycin and tobramycin
what would you do next?
What would you do next?
Broaden antibiotic coverage to include vancomycin and tobramycin
Obtain serum vitamin A levels
Obtain herpes simplex virus tissue culture and antigen detection tests
Use confocal microscopy to evaluate for cysts
b
0
1
1
1
male
0
0
53
51-60
null
549
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2721232
A white man in his 20s presented with numerous pink and brown papules and plaques, some with ulceration, on the head and along the midline of the neck and spine (Figure). Medical history included medulloblastoma treated with irradiation of the brain and spine at age 2 years. He also received radiation therapy for a meningioma of the posterior fossa in adulthood. He was also treated for several seemingly similar lesions on the face and scalp and had a family history of similar cutaneous lesions reported in 2 brothers and 1 niece.Figures show crusted erythematous papules and plaques adjacent to the surgical excision scar within a prior irradiation field on the head and neck (A) and erythematous and hyperpigmented papules and plaques overlying diffuse postsurgical scarring within a prior irradiation field on the back (B). What Is Your Diagnosis?
Angiosarcoma
Leukemia cutis
Squamous cell carcinoma
Basal cell carcinoma
D. Basal cell carcinoma
D
Basal cell carcinoma
Basal cell carcinoma (BCC) is the most common cutaneous cancer in the United States and has an indolent growth pattern, though the lesions may become crusted and ulcerated. Although metastasis is rare, local invasion and destruction may occur with aggressive or chronic lesions. Thus, early identification and treatment is prudent. Risk factors include fair complexion; a history of intense, intermittent sun exposure; immunosuppression; and a positive family history. Radiation exposure may be especially carcinogenic in inducing BCC, and BCC is more likely than squamous cell carcinoma to develop following radiation exposure, with the greatest risk seen in patients who were younger than 10 years when exposed.1 Furthermore, several genetic syndromes are associated with an increased risk of multiple BCCs, including basal cell nevus syndrome (BCNS).Basal cell nevus syndrome is an autosomal dominant condition caused by mutations of genes involved in the Sonic Hedgehog signaling pathway, most often mutations of PTCH1, a tumor suppressor.2,3 In addition to multiple BCCs, which typically develop during late adolescence to early adulthood, other classic manifestations include odontogenic keratocysts of the jaw, palmoplantar pits, calcification of the falx cerebri, and skeletal anomalies.4-6 Other neoplasms, specifically, medulloblastoma, meningioma, and cardiac and bilateral ovarian fibromas, are also associated with BCNS. Although diagnosed at an earlier age and in multiplicity, BCCs associated with BCNS usually have a similar natural history and predilection for sun-exposed areas as seen in patients without the syndrome. Of note, exposure to ionizing radiation is especially detrimental to individuals with BCNS, with a high tumor burden occurring in irradiated sites.2,4,7Prior to the present patient’s initial visit, therapy with 150 mg/d of vismodegib, a systemic inhibitor of the Sonic Hedgehog signaling pathway, had been discontinued owing to adverse effects of headache and muscle spasms. Given the burden of disease at presentation, therapy with vismodegib was restarted with an amended dosing schedule. One month of 150-mg/d vismodegib was alternated with 1 month off of treatment, which the patient tolerated well. Dosing frequency was eventually increased to 150 mg/d for 5 days per week. In addition, curettage of larger lesions was performed with topical application of imiquimod at home. Despite this treatment regimen, several lesions continued to enlarge, leading to painful ulceration and bleeding. At the last follow-up visit, a plan was made for the patient to participate in an immunotherapy trial for refractory disease.
Oncology
A white man in his 20s presented with numerous pink and brown papules and plaques, some with ulceration, on the head and along the midline of the neck and spine (Figure). Medical history included medulloblastoma treated with irradiation of the brain and spine at age 2 years. He also received radiation therapy for a meningioma of the posterior fossa in adulthood. He was also treated for several seemingly similar lesions on the face and scalp and had a family history of similar cutaneous lesions reported in 2 brothers and 1 niece.Figures show crusted erythematous papules and plaques adjacent to the surgical excision scar within a prior irradiation field on the head and neck (A) and erythematous and hyperpigmented papules and plaques overlying diffuse postsurgical scarring within a prior irradiation field on the back (B).
what is your diagnosis?
What is your diagnosis?
Angiosarcoma
Leukemia cutis
Squamous cell carcinoma
Basal cell carcinoma
d
0
0
0
1
male
0
0
25
21-30
White
550
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2722615
A 41-year-old woman with a history of Hodgkin disease in her adolescence was diagnosed with melanoma of the left upper thigh that was classified as American Joint Committee on Cancer (AJCC) stage IIA with (pT3aN0 [sn0/1] M0) BRAF mutation. One year later she underwent inguinal lymphadenectomy for macrometastasis followed by adjuvant immunotherapy with ipilimumab, a monoclonal antibody that targets cytotoxic T-lymphocyte antigen 4. Owing to local disease progression, treatment escalation with combined immunotherapy (ipilimumab and nivolumab, an anti–PD-1 [programmed cell death 1]) antibody was initiated. Apart from immunotherapy-induced thyroiditis, the treatment was well tolerated. However, after 11 cycles of nivolumab, the patient presented with progressive painful reddening and swelling of the left thigh. There was no deterioration of the general condition, no fever, no change in current medication, and no recent cold exposure. Clinical examination revealed diffuse erythematous induration of the upper thigh expanding to the lower left abdominal area, as well as multiple small palpable subcutaneous nonindurated nodules on the ipsilateral gluteal region and leg (Figure, A). Fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography–computed tomography (PET-CT) detected multiple FDG-active lesions in both lower extremities and the lumbar region; a previously described left iliac lymphadenopathy was partially regressed (Figure, B). A diagnostic biopsy specimen of the left thigh revealed a lobular lymphocytic panniculitis with dense infiltrate of mainly lymphocytes and plasma cells with no signs of vasculitis (Figure, C and D). Direct immunofluorescence and blood analysis showed an absence of specific autoimmune antigens (test results for α-1 antitrypsin, antinuclear antibody, anti-Sjögren syndrome–related antigen, and anti-Sjögren syndrome–related antigen B were all unremarkable).A, Localized erythematous brownish induration seen on the upper thigh. Arrowheads correlate with the most prominent clinical findings. B, Radioactive fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography–computed tomography (PET-CT) reveals multiple subcutaneous lesions in both lower extremities, the lumbar region, and on the ipsilateral lower leg with high FDG uptake highly suggestive of (in transit) metastasis. C and D, Lesional skin biopsy samples obtained from the proximal left thigh show normal epidermis and dermis with patchy infiltrate of the deep subcutaneous fat (hematoxylin-eosin stain). Green arrows indicate the plasma cells, and blue arrows indicate the lymphocytes. What Is Your Diagnosis?
Disease progression
Nivolumab-induced extensive panniculitis
Cold panniculitis
Erythema nodosum under nivolumab
B. Nivolumab-induced extensive panniculitis
B
Nivolumab-induced extensive panniculitis
Immunologic checkpoint blockade has changed the treatment landscape for advanced melanoma with impressive response and survival rates,1 but it is associated with a large spectrum of immune-related toxic effects owing to an increase of the baseline T-cell–specific immune response.2,3 Skin toxic effects represent the most common adverse effects associated with immune checkpoint inhibitors, including pruritus, rash, vitiligolike depigmentation, and lichenoid, cytotoxic, and autoimmune bullous reactions.4 Interestingly, appearance of these adverse effects, especially vitiligo, is associated with a superior outcome and treatment response.5Panniculitis is a group of inflammatory disorders of the subcutaneous fat tissue. Diagnosis can be challenging because different forms of panniculitis may present with overlapping clinical findings. Panniculitis can either be classified clinically based on the cause (eg, infection, inflammation, trauma, enzymatic destruction, deposition, or cancer) or histologically as lobular or septal panniculitis with or without concomitant vasculitis. As for drug-induced panniculitis, clinical and histopathological features of drug-induced panniculitis are indistinguishable from those associated with other agents, and only the history of previous drug intake or clinical improvement after drug treatment interruption may prove a causative relationship.6Erythema nodosum (EN) is the most common type of panniculitis located on the anterior surface of the lower extremities. Mostly idiopathic in origin, EN may indicate an underlying systematic disease such as tuberculosis, deep bacterial and/or fungal infection, inflammatory bowel disease, or cancer. Erythema nodosum represents the cutaneous manifestation of sarcoidosis. Histologically, septal panniculitis can present without vasculitis and with radial granulomas. Erythema nodosum is also reported to be the most common drug-induced panniculitis.7In the setting of immune checkpoint inhibitors used for cell-mediated immunity, few reports in the literature have described a sarcoidlike granulomatous panniculitis.7-9 The typical histological pattern includes partly granulomatous reactions with mixed septal and lobular inflammatory infiltrate with lymphocytes and epithelioid histiocytes.Herein, we report a case of lobular panniculitis without vasculitis. Lobular panniculitis is usually considered in connective tissue diseases, such as systemic lupus erythematosus, pancreatic diseases or α-1 antitrypsin deficiency. Other histopathologic differential diagnoses include infection, trauma, or subcutaneous T-cell lymphoma.10 All of these differential diagnoses were ruled out through diagnostic workup in the present case. The absence of tumor cells in the biopsy specimen ruled out disease progression.The treatment with nivolumab was continued unchanged, and the patient achieved a complete metabolic response seen in the routine follow-up 18F-FDG PET-CTs. As for the panniculitis, the lesion regressed slowly without the need for systemic steroid or treatment cessation.With regard to the treatment approach for immunotherapy-induced panniculitis, the lesions usually regress spontaneously or after treatment with nonsteroidal anti-inflammatory drugs or oral or topical steroids. Cessation of immunotherapy is not generally necessary.To our knowledge, this is the first case of an extensive lobular panniculitis in a patient treated with an anti–PD-1 antibody. Until recently, panniculitides associated with immunotherapeutic agents have not been extensively described. Because inconclusive FDG-avid lesions may simulate disease progression, recognition of these adverse effects is important, and histopathological workup is mandatory to ensure adequate treatment and prevent unnecessary treatment discontinuation.
Oncology
A 41-year-old woman with a history of Hodgkin disease in her adolescence was diagnosed with melanoma of the left upper thigh that was classified as American Joint Committee on Cancer (AJCC) stage IIA with (pT3aN0 [sn0/1] M0) BRAF mutation. One year later she underwent inguinal lymphadenectomy for macrometastasis followed by adjuvant immunotherapy with ipilimumab, a monoclonal antibody that targets cytotoxic T-lymphocyte antigen 4. Owing to local disease progression, treatment escalation with combined immunotherapy (ipilimumab and nivolumab, an anti–PD-1 [programmed cell death 1]) antibody was initiated. Apart from immunotherapy-induced thyroiditis, the treatment was well tolerated. However, after 11 cycles of nivolumab, the patient presented with progressive painful reddening and swelling of the left thigh. There was no deterioration of the general condition, no fever, no change in current medication, and no recent cold exposure. Clinical examination revealed diffuse erythematous induration of the upper thigh expanding to the lower left abdominal area, as well as multiple small palpable subcutaneous nonindurated nodules on the ipsilateral gluteal region and leg (Figure, A). Fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography–computed tomography (PET-CT) detected multiple FDG-active lesions in both lower extremities and the lumbar region; a previously described left iliac lymphadenopathy was partially regressed (Figure, B). A diagnostic biopsy specimen of the left thigh revealed a lobular lymphocytic panniculitis with dense infiltrate of mainly lymphocytes and plasma cells with no signs of vasculitis (Figure, C and D). Direct immunofluorescence and blood analysis showed an absence of specific autoimmune antigens (test results for α-1 antitrypsin, antinuclear antibody, anti-Sjögren syndrome–related antigen, and anti-Sjögren syndrome–related antigen B were all unremarkable).A, Localized erythematous brownish induration seen on the upper thigh. Arrowheads correlate with the most prominent clinical findings. B, Radioactive fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography–computed tomography (PET-CT) reveals multiple subcutaneous lesions in both lower extremities, the lumbar region, and on the ipsilateral lower leg with high FDG uptake highly suggestive of (in transit) metastasis. C and D, Lesional skin biopsy samples obtained from the proximal left thigh show normal epidermis and dermis with patchy infiltrate of the deep subcutaneous fat (hematoxylin-eosin stain). Green arrows indicate the plasma cells, and blue arrows indicate the lymphocytes.
what is your diagnosis?
What is your diagnosis?
Cold panniculitis
Disease progression
Nivolumab-induced extensive panniculitis
Erythema nodosum under nivolumab
c
1
1
1
1
female
0
0
41
41-50
null
551
original
https://jamanetwork.com/journals/jamacardiology/fullarticle/2722743
A woman in her mid-50s presented to the hospital with acute, worsening dyspnea, with shortness of breath at rest and with minimum exertion. She had undergone mechanical mitral valve replacement and aortic dissection repair 2 months prior to presentation. On physical examination, she was found to have elevated jugular venous pressure, bilateral rales in her lungs, and reduced intensity of the mitral valve closure sound, and her international normalized ratio was subtherapeutic (1.5). An echocardiographic image showed a mean gradient of 12 mm Hg across the mitral valve, suggesting valve stenosis. Fluoroscopy showed restricted mobility of the leaflets (Video 1). Transesophageal echocardiography showed a large thrombus on the mechanical mitral valve, along with a small new dissection flap in the aortic root (Figure and Video 2). A chronic dissection flap was evident in the aortic arch and descending aorta, which was unchanged from the previous aortic dissection repair.Refer the patient for aortic root dissection repairAdminister a full dose of emergency tissue plasminogen activator What Would You Do Next?
Refer the patient for aortic root dissection repair
Start intravenous heparin
Administer a full dose of emergency tissue plasminogen activator
Perform emergency percutaneous clot removal
Acute mechanical mitral valve thrombosis
B
Start intravenous heparin
The decision was made to start the patient on intravenous heparin. Repeat surgery (choice A) was considered high risk in this patient because she had New York Heart Association functional class IV symptoms, pulmonary edema, and a history of surgery 2 months ago. The new dissection flap was small, with a small leak of blood into the false lumen. The patient presented with decompensated heart failure symptoms but had no chest pain. Tissue plasminogen activator (choice C) was not given because of the small stable dissection flap seen in the aortic root. Performing an emergency percutaneous clot removal procedure (choice D) is not correct because, for practical purposes, there is no device that can perform percutaneous removal of clots formed on mechanical valves.Mechanical valve thrombosis is a relatively uncommon complication that has an annual occurrence of 1% to 2%, even with appropriate antithrombotic therapy. It can occur despite international normalized ratio in a therapeutic range at testing intervals.1 The highest risk of thrombosis is in the immediate 3 to 6 months after valve replacement, particularly in the first 10 to 30 days, regardless of location of the valve. Mitral prostheses carry an approximately 2-fold to 3-fold greater risk for thrombosis than aortic protheses do.2The clinical manifestations of mechanical valve thrombosis vary depending on whether the valvular dysfunction is obstructive or nonobstructive. In the short-term setting, obstructive valve dysfunction can present with acute decompensated heart failure signs and symptoms (as with this patient), syncope (or presyncope), pulmonary embolism, stroke, or even myocardial infarction from thromboembolism, and (rarely) sudden cardiac death.3 When the valve is nonobstructive in nature, patients may even be asymptomatic, and thrombosis becomes evident through a change in closing clicks, abnormality or restriction in valve movement, or increased gradients on echocardiograms done incidentally for other causes.Echocardiographic evidence of more than a 50% increase in the mean transvalvular gradient should prompt further imaging regardless of prosthetic location. Further imaging depends on the location of the prosthetic valve of interest. When thrombosis is suspected, transesophageal echocardiography is often the initial modality for evaluation and can be useful in detecting severe obstruction.4 Transesophageal echocardiography is often the modality of choice for mitral or tricuspid prosthetic valves, while multidetector-row computed tomography has better visualization of mechanical aortic or pulmonic valves.5 Transesophageal echocardiography and/or multidetector-row computed tomography are often done in conjunction with fluoroscopy, which allows evaluation of mechanical valve leaflet motion without the interference of soft tissue.Immediate management options depend on the degree of symptoms, valve location (right vs left), and thrombus size. Patients with left-sided mechanical valve obstruction have high rates of mortality and morbidity, and immediate management with either fibrinolytics or surgical intervention should be initiated when this is considered feasible and safe. Presence of New York Heart Association functional class IV symptoms, a large thrombus size (>5 mm), and left-sided mechanical-valve thrombi are considered high-risk features and mandate emergency interventions, such as surgery. Based on the patient’s condition, risks and benefits of a surgical vs conservative approach and fibrinolytic vs anticoagulation therapy, along with heart failure management, should be considered. Low-dose fibrinolytic therapy is recommended for poor surgical candidates and patients with New York Heart Association functional class I, II, or III symptoms, small clot sizes (≤0.8 cm2), and no or mild coronary artery disease with no other concomitant valve disease or contraindication to fibrinolytic medications, based on patient’s choice.6This patient presented with a large thrombus burden on a left-sided valve in decompensated heart failure. A usual recommendation would be to send her for valve-replacement surgery. However, a repeat open-heart operation within 2 months is considered to involve a high risk of mortality. In this patient, the presence of a concomitant dissection flap in the aortic root also increased the risk for bleeding with use of fibrinolytic therapy. After thorough discussion with the patient, we pursued a conservative approach with intravenous heparin for 3 days. Her symptoms improved, and she remained well compensated.The patient’s symptoms improved in a few days, and her mean gradient across the mechanical mitral prosthesis was reduced to 5 mm Hg. She was discharged with a planned reevaluation and repeat imaging of the aortic root dissection flap in a few months.
Cardiology
A woman in her mid-50s presented to the hospital with acute, worsening dyspnea, with shortness of breath at rest and with minimum exertion. She had undergone mechanical mitral valve replacement and aortic dissection repair 2 months prior to presentation. On physical examination, she was found to have elevated jugular venous pressure, bilateral rales in her lungs, and reduced intensity of the mitral valve closure sound, and her international normalized ratio was subtherapeutic (1.5). An echocardiographic image showed a mean gradient of 12 mm Hg across the mitral valve, suggesting valve stenosis. Fluoroscopy showed restricted mobility of the leaflets (Video 1). Transesophageal echocardiography showed a large thrombus on the mechanical mitral valve, along with a small new dissection flap in the aortic root (Figure and Video 2). A chronic dissection flap was evident in the aortic arch and descending aorta, which was unchanged from the previous aortic dissection repair.Refer the patient for aortic root dissection repairAdminister a full dose of emergency tissue plasminogen activator
what would you do next?
What would you do next?
Administer a full dose of emergency tissue plasminogen activator
Perform emergency percutaneous clot removal
Refer the patient for aortic root dissection repair
Start intravenous heparin
d
1
1
0
1
female
0
0
55
51-60
null
552
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2726813
A white man in his 70s presented with a gradually enlarging right supraorbital mass that had been neglected for 4 years. His comorbidities included congestive heart failure, atrial fibrillation, coronary artery disease, chronic obstructive pulmonary disease, hypertension, and a 50-pack-year history of tobacco abuse. Physical examination revealed a firm, nonmobile, nontender, bulging mass situated over the right frontal sinus, which expanded to the upper right eyelid. The mass covered the globe completely, resulting in total vision obstruction (Figure, A). However, the patient’s vision remained intact when the mass was elevated off the eye with great effort. His right pupil was round and reactive to light without afferent pupillary defect. No palpable lymphadenopathy was noted. A non–contrast-enhanced computed tomography (CT) scan demonstrated a large, right supraorbital mass with bony destruction of the anterior table of the right frontal sinus (Figure, B). It extended into the superior aspect of the orbit, displacing the right globe inferiorly. Air was seen within the mass (Figure, C). A fine-needle aspiration biopsy yielded inconclusive results. After medical clearance, the patient was taken to the operating room for a wedge excisional biopsy with decompression of the right supraorbital mass under monitored anesthesia care. Histopathological investigation revealed large nests of neoplastic epithelioid cells surrounded by fibrotic tissue and necroinflammatory debris. The neoplastic cells contained round nuclei with single prominent nucleoli and abundant clear to vacuolated cytoplasm (Figure, D).A, Enlarged right supraorbital mass. B, Computed tomography (CT) scan of destruction of the anterior table of right frontal sinus. C, Air pocket within the mass. D, Neoplastic epithelioid cells of the mass (original magnification ×40). What Is Your Diagnosis?
Pott puffy tumor
Mucocele
Basal cell carcinoma
Sebaceous carcinoma
D. Sebaceous carcinoma
D
Sebaceous carcinoma
Sebaceous carcinoma (SC) is an aggressive, malignant epithelial tumor with a variable presentation and occasional multicentric origin due to the widespread distribution of sebaceous glands in the skin.1-3 Sebaceous carcinoma encompasses 0.67% of all tumors of the eyelid, and is the third most common cancer of the eyelid (3%) behind basal cell carcinoma (86%) and squamous cell carcinoma (7%).2,4-6 The majority of sebaceous carcinomas occur in the intratarsal Meibomian and Zeis glands (38.7%), with other locations noted in the literature including the trunk, scalp, neck, upper limbs, lower limbs, and genitalia.5-8 An SC typically presents as a painless, firm, and rounded mass with a yellow hue due to its lipid content.2 It can also present as treatment unresponsive unilateral blepharitis or as a pedunculated lesion with keratinization and ulceration similar to a cutaneous horn or basal cell carcinoma.7 One clinical feature that would suggest SC over its benign mimics is a diffuse loss of cilia, which is typically seen in malignant lesions.1,7 Other lesions to add to the differential diagnosis include basal cell carcinoma, cutaneous horn, blepharitis, squamous cell carcinoma, Pott puffy tumor, chalazion, hordeolum, and sebaceous adenoma. Sebaceous carcinoma tends to be locally aggressive and has been known to invade the orbit and cause orbital displacement.1 Invasion of the paranasal sinuses has been noted only once to our knowledge, involving the maxillary sinus and ethmoids.2 To the our knowledge, there have been no reports of an aggressive SC that has invaded the frontal sinus alone, as seen in this case. As seen in the Figure, B, this SC invaded the anterior wall of the frontal sinus with noted air pockets, and because of its location, appeared initially similar to a Pott puffy tumor. To distinguish SC from Pott puffy tumor, a biopsy with corresponding histological analysis was required (Figure, D). Final pathological findings revealed large pleomorphic cells with brisk mitotic activity, vacuolated cytoplasm, and zones of necrosis, which are consistent with SC.1,2 Extensive necrotic areas were noted without normal epithelial or stromal tissues, suggesting that the tumor’s rapid growth outpaced its vascular supply. Oil-red-O staining can be adjunctively used to demonstrate the large lipid components of these cells.1,2It has been widely reported that advanced age is the main risk factor for developing SC.1,5,7,8 Historically, Asian descent was noted as a risk factor for the development of SC, but recent studies have suggested otherwise.1,7 Multiple systematic reviews have shown that older, white men have the highest risk of developing SC.1,5,7Traditionally, wide local excision with 5-to 6-mm surgical margins was the standard treatment for SC, along with either frozen or permanent sections.1,7,9 The noted 5-year recurrence rate with wide local excision varies between 9% and 36%.10 If there is orbital involvement, either on presentation or with recurrence, exenteration of the eye is typically performed.1 Recently, emphasis has been placed with Mohs micrographic surgery as a curative treatment for SC because of its inherent tissue conservation. One study by Snow et al10 of 49 patients noted a local cure rate of 87.8% for SC with Mohs micrographic surgery.1 As for nonsurgical treatment options, both irradiation and cryotherapy have been used in poor surgical candidates and patients unwilling to undergo surgery. Callahan et al1 noted that 2 patients were treated with radiation and had no evidence of SC recurrence at 28 and 37 months. Doxanas and Green2 question the value of radiation therapy after reviewing 40 prior cases in the literature. In the present case, the patient elected against treatment for his SC and shortly after died of cardiovascular disease.
General
A white man in his 70s presented with a gradually enlarging right supraorbital mass that had been neglected for 4 years. His comorbidities included congestive heart failure, atrial fibrillation, coronary artery disease, chronic obstructive pulmonary disease, hypertension, and a 50-pack-year history of tobacco abuse. Physical examination revealed a firm, nonmobile, nontender, bulging mass situated over the right frontal sinus, which expanded to the upper right eyelid. The mass covered the globe completely, resulting in total vision obstruction (Figure, A). However, the patient’s vision remained intact when the mass was elevated off the eye with great effort. His right pupil was round and reactive to light without afferent pupillary defect. No palpable lymphadenopathy was noted. A non–contrast-enhanced computed tomography (CT) scan demonstrated a large, right supraorbital mass with bony destruction of the anterior table of the right frontal sinus (Figure, B). It extended into the superior aspect of the orbit, displacing the right globe inferiorly. Air was seen within the mass (Figure, C). A fine-needle aspiration biopsy yielded inconclusive results. After medical clearance, the patient was taken to the operating room for a wedge excisional biopsy with decompression of the right supraorbital mass under monitored anesthesia care. Histopathological investigation revealed large nests of neoplastic epithelioid cells surrounded by fibrotic tissue and necroinflammatory debris. The neoplastic cells contained round nuclei with single prominent nucleoli and abundant clear to vacuolated cytoplasm (Figure, D).A, Enlarged right supraorbital mass. B, Computed tomography (CT) scan of destruction of the anterior table of right frontal sinus. C, Air pocket within the mass. D, Neoplastic epithelioid cells of the mass (original magnification ×40).
what is your diagnosis?
What is your diagnosis?
Pott puffy tumor
Basal cell carcinoma
Sebaceous carcinoma
Mucocele
c
1
1
1
1
male
0
0
75
71-80
White
553
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2727328
A 46-year-old African swimmer presented with a 1-month history of progressive left hearing loss and vertigo. He also reported asthenia, sleep disorders, and urinary frequency. The urinary frequency has been treated for 2 years with many drugs (tamsulosin, desmopressin, and solifenacin) without improvement. The patient had spent his childhood in eastern Africa, where he swam in Lake Tanganyika. His history did not reveal trauma, infection, or any other associated conditions at the onset of the complaints. The ear, nose, and throat examination revealed a right horizontal nystagmus and a poorly delineated hypoesthesia of the left pinna. He could not walk properly owing to his vertigo. The rest of the clinical examination findings were unremarkable. Additional examinations (ie, pure-tone audiometry, video nystagmography, tympanometry) revealed left profound hearing loss with an air-bone gap of 40 dB and central vertigo. Otoacoustic emissions were absent on the left side. There was no left acoustic reflex. Auditory brainstem responses showed asymmetric interpeak latencies of waves I through V and III through V, suggesting slower conduction on the left side. Computed tomography and cerebral magnetic resonance imaging (MRI) revealed a 24 × 20-mm hemorrhagic lesion in the left middle cerebellar peduncle associated with a thickening of the cochleovestibular nerve (Figure). The patient was referred to urology, where a 13 × 8-mm mass was discovered in the lateral wall of the prostate gland. Biopsies of prostate and brain lesions, blood tests, and urinalysis were performed.Computed tomography (CT) scan and magnetic resonance images (MRIs) of the left middle cerebellopontine angle and prostate gland. A, Axial CT scan showed a hyperdense lesion suggesting hemorrhage in the left middle cerebellar peduncle. B, The lesion extended to the base of the vestibulocochlear nerve that was thickened (arrowheads) on the Fast Imaging Employing Steady-State Acquisition (FIESTA) MRI (axial plane). C, Coronal postcontrast MRI demonstrated a T1 hyperintense lesion that was consistent with hemorrhagic lesion with no true enhancement. D, Axial pelvic MRI showed a prostatic lesion (13 × 4 mm) located in the left posterior and lateral wall (arrowhead); the prostatic volume and size were substantially increased (132 mL, 59 × 60 × 70 mm). What Is Your Diagnosis?
Prostate cancer with brain metastasis
Tuberculosis
Toxoplasmosis
Schistosomiasis
D. Schistosomiasis
D
Schistosomiasis
The cerebrospinal fluid analysis was unremarkable, but our additional examinations identified the presence of Schistosoma hematobium (schistosomiasis). The patient was treated with praziquantel and benefited from prostatectomy owing to the increased prostatic size (Figure). The vertigo progressively disappeared throughout the months of therapy.Trematode infections are transmitted when larval forms released by freshwater snails penetrate human skin during contact with infested water. The patient was probably infected during his training sessions in Lake Tanganyika. Six months after the end of the treatment, brain MRI revealed a disappearance of the lesion. Urogenital schistosomiasis is characterized by asthenia, fever, abdominal pain, and urogenital disorders.1 Neuroschistosomiasis is a rare form of the disease, affecting 3% to 5% of individuals infected with Schistosoma japonicum.1 The incidence of cerebral schistosomiasis caused by other Schistosoma species (including S hematobium, Schistosoma mansoni, Schistosoma intercalatum, and Schistosoma mekongi) is still unknown. Neurological manifestations are the result of the inflammatory response of the host to egg deposition in the brain and, depending on the location, may lead to headache, ataxia, transient flaccid paraplegia, sphincter dysfunction, sensory disturbance, and other dysfunctions.1The present case is exceptional in 2 respects. First, the clinical presentation of the disease characterized by vertigo and hearing loss developed many years after disease transmission, which is unusual. Indeed, the mean delay of the disease manifestation is approximately 1 month after the initial infection, and dormant forms of the disease are rare.2 Egg deposition on the cochleovestibular nerve explains the atypical presentation of the disease. To our knowledge, no similar case has been reported.Second, urogenital schistosomiasis (due to S hematobium infection) is rarely associated with an impairment of the central nervous system. When the disease spreads to the central nervous system, the eggs usually deposit in the spine rather than the brain.1 Moreover, as described in postmortem studies, the deposition of trematode eggs in the brain is usually asymptomatic.3 The definitive diagnosis is made with an identification of eggs in urine, while the diagnosis can be indirectly suspected by blood analyses.1 As found in the present case, physicians should take into consideration that the cerebrospinal fluid can be normal in this setting or can show nonspecific findings.3In the present case, main differential diagnoses included prostatic cancer with cerebral metastasis, tuberculosis, and toxoplasmosis. The MRI findings of cancer and metastasis are usually characterized by T1 hyposignal, T2 hypersignal, and diffusion-weighted hypersignal. In the present case, the absence of diffusion hypersignal put into question the cancer/metastasis diagnosis. Toxoplasmosis may occur with similar neurological presentation, and the MRI findings of toxoplasmosis granuloma under treatment may be characterized by T1 and Fast Imaging Employing Steady-State Acquisition (FIESTA) hypersignal with hemorrhagic presentation.4 Furthermore, the spread of the disease into the prostate gland is only anecdotally reported and mainly in immunosuppressed patients.Tuberculosis may display numerous clinical presentations, but the imaging studies commonly find small tubercles with T1 hyposignal. Although tubercles can be characterized by an annular enhanced signal on the MRI, there is no hemorrhagic finding, such as was found in the present case. In addition, we did not identify Mycobacterium tuberculosis in either urine or cerebrospinal fluid.Although schistosomiasis is rarely associated with ear, nose, and throat disorders,5 physicians should consider the diagnosis in patients who visited endemic areas and who present with head and neck neurological deficits. The disease may manifest several years after the contamination, making the diagnosis difficult and the patient history crucial.
General
A 46-year-old African swimmer presented with a 1-month history of progressive left hearing loss and vertigo. He also reported asthenia, sleep disorders, and urinary frequency. The urinary frequency has been treated for 2 years with many drugs (tamsulosin, desmopressin, and solifenacin) without improvement. The patient had spent his childhood in eastern Africa, where he swam in Lake Tanganyika. His history did not reveal trauma, infection, or any other associated conditions at the onset of the complaints. The ear, nose, and throat examination revealed a right horizontal nystagmus and a poorly delineated hypoesthesia of the left pinna. He could not walk properly owing to his vertigo. The rest of the clinical examination findings were unremarkable. Additional examinations (ie, pure-tone audiometry, video nystagmography, tympanometry) revealed left profound hearing loss with an air-bone gap of 40 dB and central vertigo. Otoacoustic emissions were absent on the left side. There was no left acoustic reflex. Auditory brainstem responses showed asymmetric interpeak latencies of waves I through V and III through V, suggesting slower conduction on the left side. Computed tomography and cerebral magnetic resonance imaging (MRI) revealed a 24 × 20-mm hemorrhagic lesion in the left middle cerebellar peduncle associated with a thickening of the cochleovestibular nerve (Figure). The patient was referred to urology, where a 13 × 8-mm mass was discovered in the lateral wall of the prostate gland. Biopsies of prostate and brain lesions, blood tests, and urinalysis were performed.Computed tomography (CT) scan and magnetic resonance images (MRIs) of the left middle cerebellopontine angle and prostate gland. A, Axial CT scan showed a hyperdense lesion suggesting hemorrhage in the left middle cerebellar peduncle. B, The lesion extended to the base of the vestibulocochlear nerve that was thickened (arrowheads) on the Fast Imaging Employing Steady-State Acquisition (FIESTA) MRI (axial plane). C, Coronal postcontrast MRI demonstrated a T1 hyperintense lesion that was consistent with hemorrhagic lesion with no true enhancement. D, Axial pelvic MRI showed a prostatic lesion (13 × 4 mm) located in the left posterior and lateral wall (arrowhead); the prostatic volume and size were substantially increased (132 mL, 59 × 60 × 70 mm).
what is your diagnosis?
What is your diagnosis?
Prostate cancer with brain metastasis
Toxoplasmosis
Schistosomiasis
Tuberculosis
c
1
1
0
1
male
0
0
46
41-50
African
554
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2729841
A preschool boy presented with asymptomatic, reddish, raised lesions over his right ear, noticed 3 months earlier, associated with multiple painless swellings on the ipsilateral side of the neck for the past 3 weeks. On examination, there were multiple, erythematous papules coalescing to form a plaque with areas of scarring over the posterior aspect of right ear, along with firm, nontender posterior cervical lymphadenopathy (Figure, A). There was no history of trauma or intervention prior to the onset of lesions. His medical history was unremarkable, as were the rest of the mucocutaneous and systemic examination findings. A skin biopsy from the plaque was submitted for histopathological examination (Figure, C and D).A, Erythematous papules coalescing to form a plaque with areas of scarring, along with ipsilateral posterior cervical lymphadenopathy. B, The lesions have healed with atrophic scarring after 6 weeks of treatment. C, Microphotograph showing unremarkable epidermis with evidence of granulomatous inflammation and central caseous necrosis in the dermis (original magnification ×100). D, Ziehl-Neelsen stain shows acid-fast bacilli (oil immersion; original magnification ×1000). What Is Your Diagnosis?
Fixed cutaneous sporotrichosis
Lupus vulgaris
Atypical mycobacterial infection
Cutaneous leishmaniasis
B. Lupus vulgaris
B
Lupus vulgaris
Histopathological examination revealed multiple foci of caseating epithelioid cell granulomas with occasional Langerhans type giant cells and moderate lymphomononuclear infiltrate (Figure, C). Ziehl-Neelsen staining for acid-fast bacilli was positive (Figure, D), while stainings for fungus (periodic acid–Schiff and Grocott stains) were negative. Fine-needle aspirate from 1 of the enlarged lymph nodes revealed loose epithelioid cell granulomas in a necrotic background. Chest radiography was normal. Intradermal testing with purified protein derivative produced an induration of 15 × 15 mm. Culture studies for leishmaniasis, fungus, and mycobacteria yielded no growth. Overall features were consistent with a diagnosis of cutaneous tuberculosis, lupus vulgaris type. Therapy was begun with antitubercular treatment (ATT) consisting of 6-month regimen of 4 drugs, known as 2 HRZE/4 HR (2 months of isoniazid [H], 10-15 mg/kg/; rifampicin [R], 10-15 mg/kg/d; pyrazinamide [Z], 20-30 mg/kg/d; and ethambutol [E], 15 mg/kg/d followed by 4 months of isoniazid [H] and rifampicin [R] for 4 months at the same doses). At 6-week follow-up, there was remarkable improvement in the lesions (Figure, B).Childhood tuberculosis is a major health problem, especially in developing countries. Though pulmonary tuberculosis is the most common form, extrapulmonary forms have steadily been on the rise. Cutaneous tuberculosis is a rare form of extrapulmonary tuberculosis with myriad of clinical presentations accounting for approximately 1% to 2% of cases.1 Though the spectrum of cutaneous tuberculosis in children is similar to that in adults, widespread disease, unusual variants, and systemic involvement are more common. The 2 most common forms of childhood cutaneous tuberculosis are lupus vulgaris and scrofuloderma.1Lupus vulgaris is a gradually progressive form of cutaneous tuberculosis, usually seen in patients with fairly good immunity. Lesions most commonly occur on the lower extremities and buttocks, and are usually solitary, though multiple lesions can occur in children. It begins as asymptomatic papules that coalesce to form soft, reddish-brown plaques, which are seen as “apple jelly” nodules on diascopy. The gradually progressive plaque generally shows an actively spreading margin at one end, with involution and scarring at the other end of the lesion, often resulting in a geographic shape. Several variants of lupus vulgaris have been described, namely, plaque form, ulcerative and mutilating forms, vegetating, tumorlike, and papulonodular forms.2 Though systemic manifestations are rare, lymph node involvement can occur, as seen in the present case. Biopsy from the active margin of the plaque showed marked epithelial hyperplasia with tuberculoid granulomas showing caseous necrosis and Langerhans giant cells in the dermis, features typical of cutaneous tuberculosis.1 The foci of cutaneous tuberculosis and tubercular involvement of concomitant draining lymph nodes represents the cutaneous equivalent of pulmonary Ghon complex. Rapid clinical response to antituberculous treatment within 6 weeks further strengthened our diagnosis. At last follow-up, continued ATT was planned for a full course of 6 months, as mandated by revised national tuberculosis control program.3Lymphocutaneous sporotrichosis was a close differential diagnosis in the present case owing to the linear arrangement of the enlarged lymph nodes and the cutaneous lesion. Although sporotrichosis in children occurs on the face more often in children than in adults,4 and can arise without trauma, presence of scarring at one end in the lesion, absence of suppurative granulomas in the biopsy, negative tissue culture, and the favorable response to ATT helped to exclude this differential diagnosis.Nontuberculous mycobacterial infections have a diverse clinical presentation. It was a differential diagnosis in the present case owing to the lymphocutaneous nature of the lesion. However, its relative rarity in the pediatric age group, well-formed granulomas seen on histopathologic analysis, and a favorable response to ATT pointed to a diagnosis of lupus vulgaris.Cutaneous leishmaniasis usually involves exposed areas, and the nose and ears are among the common sites involved. Lesions commonly begin as a solitary, small, well-circumscribed papule at the site of inoculation and slowly enlarge over several weeks into a nodule or plaque, eventually becoming ulcerated orverrucous.5 Lesions may be solitary or multiple, with presence of satellite lesions or lymphatic spread. Absence of plasma cell infiltrate in the skin biopsy specimens in the present case along with presence of caseous granuloma steered the diagnosis toward cutaneous tuberculosis.In conclusion, this patient had an uncommon presentation of lupus vulgaris at an unusual site that mimicked other common cutaneous diseases such as lymphocutaneous sporotrichosis and cutaneous leishmaniasis. Diagnosis was confirmed histopathologically. The lesions responded well to antituberculous treatment.
General
A preschool boy presented with asymptomatic, reddish, raised lesions over his right ear, noticed 3 months earlier, associated with multiple painless swellings on the ipsilateral side of the neck for the past 3 weeks. On examination, there were multiple, erythematous papules coalescing to form a plaque with areas of scarring over the posterior aspect of right ear, along with firm, nontender posterior cervical lymphadenopathy (Figure, A). There was no history of trauma or intervention prior to the onset of lesions. His medical history was unremarkable, as were the rest of the mucocutaneous and systemic examination findings. A skin biopsy from the plaque was submitted for histopathological examination (Figure, C and D).A, Erythematous papules coalescing to form a plaque with areas of scarring, along with ipsilateral posterior cervical lymphadenopathy. B, The lesions have healed with atrophic scarring after 6 weeks of treatment. C, Microphotograph showing unremarkable epidermis with evidence of granulomatous inflammation and central caseous necrosis in the dermis (original magnification ×100). D, Ziehl-Neelsen stain shows acid-fast bacilli (oil immersion; original magnification ×1000).
what is your diagnosis?
What is your diagnosis?
Fixed cutaneous sporotrichosis
Lupus vulgaris
Cutaneous leishmaniasis
Atypical mycobacterial infection
b
0
0
1
1
male
0
0
null
null
null
555
original
https://jamanetwork.com/journals/jama/fullarticle/2730013
A 53-year-old man presented with 2 weeks of back pain, stiff neck, headache, and fevers. He also reported 3 days of right knee pain and swelling. He had a history of pseudogout and kidney transplant 13 years ago. He was taking sirolimus and prednisone, with excellent graft function. His temperature on presentation was 102°F (38.9°C). He had nuchal rigidity and positive Kernig sign. There were no focal neurologic deficits and no spinal tenderness. He had a right knee effusion without erythema and with full range of motion. Laboratory workup of serum showed a white blood cell (WBC) count of 10 200/μL (neutrophils, 63.8%; lymphocytes, 19.6%; monocytes, 15.1%) and erythrocyte sedimentation rate, 63 mm/h; workup of cerebrospinal fluid showed a WBC count of 0/μL; red blood cell count, 32/μL; total protein level, 109 mg/dL; and glucose level, 59 mg/dL (3.3 mmol/L). Magnetic resonance imaging (MRI) of the spine showed cervical degenerative changes with reactive marrow (Figure 1). He was prescribed intravenous vancomycin and cefepime. Blood and cerebrospinal fluid (CSF) cultures were negative. What Would You Do Next?
Continue intravenous antibiotics
Obtain additional imaging with computed tomography (CT)
Start gabapentin
Start high-dose ibuprofen
Crowned dens syndrome
B
Obtain additional imaging with computed tomography (CT)
The key to the correct diagnosis is the presence on a CT scan of synovial crown- or halo-like calcific densities of the atlantoaxial (C1/C2) joint, surrounding the odontoid process, in a patient with history of peripheral acute calcium pyrophosphate (CPP) crystal arthritis (“pseudogout”) (Figure 2). This patient had crowned dens syndrome (CDS), ie, deposition of CPP or, less frequently, basic calcium phosphate (BCP) crystals in the atlantoaxial joint, causing symptoms and signs of cervical spine inflammation. Continuation of antibiotics was not indicated, since CSF and MRI findings were not consistent with infection. Gabapentin is effective against neuropathic but not acute inflammatory pain. Like other forms of crystal deposition disease, CDS can be treated with corticosteroids or colchicine when there is a relative contraindication to use of nonsteroidal anti-inflammatory drugs (NSAIDs, eg, ibuprofen), such as renal transplant in this patient.Axial (A), coronal (B), and sagittal (C) computed tomography scan of C1/C2 showing periodontoid calcifications.CDS was first described in 19801 and is defined as otherwise unexplained neck pain in the presence of radiographic atlantoaxial (C1/C2) calcifications. Fever, headache, meningismus, and elevated levels of inflammatory markers are common.2-6The prevalence of CDS is not clearly established. In 1 US series of 513 consecutive CT scans obtained for acute trauma,7 the prevalence of atlantoaxial calcific depositions was 12.9% overall and was 34% after age 60 years. In 2 other recent reports, 12.5%8 to 36%9 of patients with atlantoaxial calcifications had neck pain.CDS is most commonly associated with osteoarthritis but can also be observed in patients with rheumatoid arthritis and metabolic disorders such as hyperparathyroidism or hemochromatosis, which are linked to acute CPP crystal arthritis.3,6,9,10 Synovial fluid analysis from a peripheral joint aspirate can show CPP crystals, with negative or weakly positive birefringence under polarized light microscopy. The absence of crystals does not rule out CPP crystal disease, since the sensitivity of microscopic examination for CPP crystal detection ranges from 12% to 83%.6 Radiographic imaging of the knees and wrists at the time of presentation is a noninvasive way to infer diagnosis of CDS, by visualizing peripheral chondrocalcinosis.3,5The gold standard for diagnosis is CT scan of the cervical spine, focusing on C1/2 (Figure 2). CT has greater sensitivity than MRI in demonstrating the characteristic calcifications surrounding the odontoid process3-6,9 and should be performed first when CDS is likely (eg, in patients with history of acute CPP crystal arthritis). MRI can also show signs of inflammatory arthropathy suggestive of CDS in the appropriate clinical context (degenerative changes with reactive marrow) (Figure 1). However, MRI is usually ordered when there is clinical suspicion of cord compression, osteomyelitis, or malignancy.6The differential diagnosis of CDS includes infectious meningitis, retropharyngeal abscess, and diskitis-osteomyelitis of the cervical spine.3-5 Importantly, CPP calcifications, including the C1/C2 joint, are often observed in asymptomatic individuals.5,7-10 Thus, radiographic findings suggestive of CDS can coexist with infectious causes of neck pain. Therefore, even when CDS is suspected, some patients should still undergo evaluation for infection (lumbar puncture, MRI) and receive antibiotics until infection is ruled out. Unremarkable CSF studies and MRI results favor noninfectious etiologies, prompting discontinuation of antibiotics. Other considerations are tension headache, occipital neuralgia, cervicobrachial syndrome, BCP calcific longus colli tendinitis,6 polymyalgia rheumatica/giant cell arteritis when neck stiffness is associated with shoulder pain or jaw claudication,3-6 and vertebral metastases in patients with cancer.3Treatment of CDS is similar to that of peripheral acute crystal arthritis and typically consists of a short course of high-dose NSAIDs.10 Colchicine and oral corticosteroids are effective alternatives.10 Anti-inflammatory treatment results in rapid resolution of symptoms, within days to weeks.1,4,5,10Before obtaining CT of the cervical spine, knee arthrocentesis was performed to rule out septic arthritis. Synovial fluid was consistent with aseptic inflammatory arthritis, although crystals were not seen. Three-view radiographs of the hands showed bilateral chondrocalcinosis. All antibiotics were stopped. On discharge, the patient was prescribed prednisone (40 mg daily) for 5 days with rapid clinical improvement and then resumed low-dose prednisone (5 mg daily). He has not had recurrence of his symptoms after 12 months.
General
A 53-year-old man presented with 2 weeks of back pain, stiff neck, headache, and fevers. He also reported 3 days of right knee pain and swelling. He had a history of pseudogout and kidney transplant 13 years ago. He was taking sirolimus and prednisone, with excellent graft function. His temperature on presentation was 102°F (38.9°C). He had nuchal rigidity and positive Kernig sign. There were no focal neurologic deficits and no spinal tenderness. He had a right knee effusion without erythema and with full range of motion. Laboratory workup of serum showed a white blood cell (WBC) count of 10 200/μL (neutrophils, 63.8%; lymphocytes, 19.6%; monocytes, 15.1%) and erythrocyte sedimentation rate, 63 mm/h; workup of cerebrospinal fluid showed a WBC count of 0/μL; red blood cell count, 32/μL; total protein level, 109 mg/dL; and glucose level, 59 mg/dL (3.3 mmol/L). Magnetic resonance imaging (MRI) of the spine showed cervical degenerative changes with reactive marrow (Figure 1). He was prescribed intravenous vancomycin and cefepime. Blood and cerebrospinal fluid (CSF) cultures were negative.
what would you do next?
What would you do next?
Start high-dose ibuprofen
Continue intravenous antibiotics
Obtain additional imaging with computed tomography (CT)
Start gabapentin
c
1
1
0
1
male
0
0
53
51-60
White
556
original
https://jamanetwork.com/journals/jama/fullarticle/2729031
A healthy 65-year-old woman presented with 2 days of sustained throbbing, bilateral temporal headache and bilateral blurred vision. Laboratory results are shown in the Table. Given her erythrocyte sedimentation rate (ESR) of 110 mm/h and symptoms concerning for giant cell arteritis (GCA), a temporal artery biopsy was obtained. Results showed a normal caliber lumen without infiltrating inflammatory cells. What Would You Do Next?
Brain magnetic resonance imaging
Empirical methotrexate prescription
Empirical sumatriptan prescription
Serum protein electrophoresis (SPEP)
null
D
Serum protein electrophoresis (SPEP)
While the patient had a nearly 50% pretest probability of GCA,1 the negative predictive value of temporal artery biopsy is approximately 83%.2,3 The negative biopsy results required consideration of an alternative diagnosis, such as brain abscess, cerebral venous sinus thrombosis, or hypergammaglobulinemia with hyperviscosity. The high ESR, low anion gap (2 mEq/mL), and elevated globulin gap (6.6 g/dL) support a diagnosis of hypergammaglobulinemia. Empirical steroids are recommended when GCA is strongly suspected, even before temporal artery biopsy, but were not prescribed because of high suspicion for the alternative diagnosis of hypergammaglobulinemia.The ESR measures the rate at which red blood cells (RBCs) settle in the plasma of anticoagulated blood in a standardized Westergren tube (Figure). Although the ESR is considered a measure of inflammation from infection, malignancy, or rheumatologic disease, multiple noninflammatory factors affect RBC sedimentation. These factors alter the ESR via distinct mechanisms: spatial interference, electrical charge, and viscosity (Supplement).4 Increased spatial interference, defined as physical factors interfering with the ability of RBCs to settle in the Westergren tube, slows the packing of erythrocytes at the tube bottom, thereby decreasing the ESR. With extreme leukocytosis, RBC downward flow is impeded by leukocytes, which decreases the ESR.5 Conversely, decreased spatial interference, such as with cases of anemia and consequently fewer RBCs, causes an increase in the ESR because RBCs can sediment with less obstruction.4 Electrical charge refers to the slight negative surface charge that RBCs have, which causes them to repel each other, impeding erythrocyte packing and sedimentation.5,6 However, if plasma is enriched with positively charged proteins, such as immunoglobulins or fibrinogen, this electrical repulsion is blunted and RBC agglutination occurs, speeding sedimentation.4 Fibrinogen is a large, positively charged protein and the most abundant acute-phase reactant. Elevated fibrinogen is the predominant reason for high ESR in inflammatory states.4,5,7 Increased plasma viscosity, such as in cases of hypergammaglobulinemia, can slow erythrocyte sedimentation through the plasma and decrease the ESR.4,8The erythrocyte sedimentation rate (ESR) is the distance (mm) that anticoagulated blood settles into the red blood cell and plasma components over 1 hour. The middle tube demonstrates an ESR of 20 mm/h.Because noninflammatory factors influence the ESR, its specificity for establishing a diagnosis of inflammatory conditions is modest. In an analysis of 1106 patients who underwent 1174 temporal artery biopsies, an ESR value greater than 22 mm/h in men and 29 mm/h in women had sensitivity of 84.2%, specificity of 29.5%, positive predictive value of 26.4%, and negative predictive value of 86.1% for a diagnosis of GCA.9 In another study,10 using a higher ESR cutoff improved specificity, but sensitivity markedly decreased; an ESR cutoff of greater than 107 mm/h had a sensitivity of 23%, specificity of 90.1%, positive predictive value of 30%, and negative predictive value of 86.5% for a diagnosis of GCA (positive and negative predictive values calculated by the authors from data provided by Walvick)10. In a study of 1006 patients, ESR greater than 100 mm/h had poor sensitivity for infection (36%), malignancy (25%), and inflammatory disorders (21%). However, it had high specificity (99%) for elevated sickness index, defined as the presence of a significant underlying disease.6 These results suggest an ESR greater than 100 mm/h deserves diagnostic attention but is nonspecific and cannot by itself establish a diagnosis. Common causes of ESR greater than 100 mm/h include deep-seated infection, such as endocarditis or osteomyelitis; connective tissue disorders, such as GCA; and malignancies, such as multiple myeloma and Waldenström macroglobulinemia.4 The ESR cost to Medicare is $3.33.The patient’s high ESR was related to noninflammatory factors. The low anion gap and elevated globulin gap suggested elevated paraproteins. SPEP results showed a monoclonal IgM concentration of 1260 mg/dL (reference range, 48-271 mg/dL). Because γ-globulins are positively charged plasma proteins, the repelling force of the negative surface charge of RBCs is blunted in hypergammaglobulinemia, which facilitates agglutination of RBCs and rapid sedimentation. The patient’s blood viscosity relative to water was 4.1 (reference range, 1.4-1.8). Although hyperviscosity decreases the ESR, the electrical charge effect of hypergammaglobulinemia is stronger, hence the patient’s ESR of 110 mm/h.4 Bone marrow biopsy results showed 10% Κ-restricted lymphoplasmacytic cells, confirming Waldenström macroglobulinemia. The patient’s presenting symptoms of headache and blurry vision are common presentations of this disease.In scenarios with high suspicion for GCA but negative temporal artery biopsy results, a second, contralateral biopsy can be considered. Approximately 5% of these contralateral biopsies will be positive when the first was negative.2,3 C-reactive protein (CRP) is an acute-phase reactant synthesized by the liver. CRP is elevated with most inflammatory conditions but is unaffected by noninflammatory factors.7 The patient had a normal CRP level, highlighting the noninflammatory cause of her elevated ESR. Obtaining concurrent ESR and CRP levels is less cost-effective than obtaining one before the other, but may provide benefit in select cases.The patient was treated with rituximab and has been in remission for 3 years. Her IgM concentration decreased to 261 mg/dL and her ESR decreased to 32 mm/h.Although commonly used to measure inflammation, the ESR is affected by noninflammatory factors, including anemia, polycythemia, hypofibrinogenemia or hyperfibrinogenemia, and hypergammaglobulinemia.Patients with ESR greater than 100 mm/h are likely to have a clinically relevant underlying condition, but the ESR test is nonspecific.Understanding the physiologic principles of erythrocyte sedimentation can facilitate the diagnosis of underlying etiologies.
Diagnostic
A healthy 65-year-old woman presented with 2 days of sustained throbbing, bilateral temporal headache and bilateral blurred vision. Laboratory results are shown in the Table. Given her erythrocyte sedimentation rate (ESR) of 110 mm/h and symptoms concerning for giant cell arteritis (GCA), a temporal artery biopsy was obtained. Results showed a normal caliber lumen without infiltrating inflammatory cells.
what would you do next?
What would you do next?
Empirical sumatriptan prescription
Empirical methotrexate prescription
Serum protein electrophoresis (SPEP)
Brain magnetic resonance imaging
c
0
1
1
0
female
0
0
65
61-70
null
557
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2727364
A man in his 60s presented with an approximately 4-month medical history of lip enlargement with nodular growths. Skin examination was significant for multiple erythematous and flesh-colored papules and nodules symmetrically affecting the upper and lower lips with extension onto the vermilion border (Figure 1A). There were no intraoral lesions, and all other findings of his examination were within normal limits. The patient denied fevers, chills, weight loss, night sweats, cough, lymphadenopathy, abdominal pain, change in bowel habits, or other systemic symptoms. A biopsy was obtained of the vermilion lip (Figure 1B).A, Symmetric lip enlargement with infiltrative erythematous and skin-colored papules and nodules extending onto the vermilion border. B, Hematoxylin-eosin stain (original magnification ×40). What Is Your Diagnosis?
Granulomatous cheilitis
Leukemia labialis
Lip licker’s dermatitis
Pyostomatitis vegetans
B. Leukemia labialis
B
Leukemia labialis
Histopathologic examination demonstrated a nodular and diffuse dermal lymphoid infiltrate composed of small, mature lymphocytes with hyperchromatic and slightly convoluted nuclei (Figure 2A). Findings from CD20, CD79a, and LEF-1 immunostaining highlighted a neoplastic B-cell population occurring in sheets and clusters with aberrant coexpression of CD5 and CD23 (Figure 2B).A, Lip biopsy demonstrating monotonous small lymphoid cells with hyperchromatic, slightly irregular and angulated nuclei with clumped chromatin (hematoxylin-eosin, original magnification ×400). B, immunostain diffusely highlighting the lymphoid infiltrate (CD20, original magnification ×600).A subsequent complete blood cell count showed a white blood cell count of 10 600/µL with an absolute lymphocytosis of 4378 cells/µL. Flow cytometry on the peripheral blood demonstrated a monoclonal B-cell population with dim to moderate CD20 expression and aberrant expression of CD5 and CD23. The clinical, histologic, and flow cytometry findings were consistent with a diagnosis of chronic lymphocytic leukemia (CLL) presenting initially with only lip involvement.Leukemia labialis is a rare subtype of leukemia cutis that presents as multiple plaques, papules, nodules, and (rarely) ulcerative lesions confined to the lips.1 Leukemia labialis as an initial manifestation of CLL is rare.2Approximately 25% of patients with CLL present with cutaneous manifestations of their disease.3 More common presentations include purpura, pruritus, urticaria, erythroderma, vasculitis, pyoderma gangrenosum, and Sweet syndrome.4 Leukemia cutis, however, is far less frequent in CLL than in T-cell leukemias or lymphomas.5 Leukemia cutis as the initial presenting sign of CLL is even more exceptional. Cerroni et al3 examined a cohort of 42 CLL patients with leukemia cutis and found that only 7 patients (16.7%) presented with skin lesions as the first sign of systemic disease. Importantly, 6 patients presented with leukemia cutis at sites of previous herpes zoster and herpes simplex eruptions.2,3 This finding suggests that an exaggerated herpes-induced inflammatory response may play a role in the pathogenesis of this presentation.Microscopically, cutaneous CLL is characterized by a periadnexal and perivascular, nodular and/or diffuse or band-like infiltration of CD20-positive neoplastic B cells that aberrantly express CD5 and CD23. Lymphoid enhancer binding factor 1, which was positive in the case, is a recently described highly specific marker for CLL.6 Cytologically, CLL is characterized by uniform, small lymphocytes with mature clumped and hyperchromatic chromatin. Scattered prolymphocytes with vesicular chromatin and prominent nucleoli may also be seen. A marked increase in prolymphocytes portends Richter transformation and a worse prognosis. In patients without Richter transformation, leukemia cutis in CLL likely does not significantly affect overall prognosis.3,7The differential diagnoses of multiple acquired labial papules are broad and may include infiltrative disorders (eg, granulomatous, systemic amyloidosis), infections (eg, verrucae, herpes labialis), and benign neoplasms (eg, Fordyce spots). Of the listed choices, granulomatous cheilitis (GC) typically presents in younger adults and may be associated with sarcoidosis, Crohn disease, and Melkersson-Rosenthal syndrome (MRS).8 Clinically it manifests as asymptomatic labial swelling that may have a nodular texture and may be accompanied by facial palsy and plicated tongue in MRS. Histologically, GC is distinguished from leukemia cutis by the presence of nonnecrotizing granulomas. Lip licker’s dermatitis is a form of chronic irritant contact dermatitis to saliva and repeated minor trauma secondary to habitual lip licking. Examination classically demonstrates perioral erythema, scale, crusting, or fissures. Histologic changes in irritant contact dermatitis include spongiosis, a superficial perivascular chronic inflammatory infiltrate, and scattered apoptotic keratinocytes. Pyostomatitis vegetans is strongly correlated with inflammatory bowel disease and typically presents with multiple white or yellow pustules on an erythematous base involving the oral mucosa, particularly the gingiva and lip.9 The absence of gastrointestinal symptoms and pustular lesions on examination excludes this diagnosis. In addition, biopsy will demonstrate acanthosis, exocytosis of neutrophils, and scattered neutrophilic and eosinophilic microabscesses.10This case highlights a rare presentation of leukemia labialis as the initial manifestation of CLL. It is important for clinicians to recognize both common and rare presentations of leukemia cutis to avoid delay in diagnosis and treatment.
Dermatology
A man in his 60s presented with an approximately 4-month medical history of lip enlargement with nodular growths. Skin examination was significant for multiple erythematous and flesh-colored papules and nodules symmetrically affecting the upper and lower lips with extension onto the vermilion border (Figure 1A). There were no intraoral lesions, and all other findings of his examination were within normal limits. The patient denied fevers, chills, weight loss, night sweats, cough, lymphadenopathy, abdominal pain, change in bowel habits, or other systemic symptoms. A biopsy was obtained of the vermilion lip (Figure 1B).A, Symmetric lip enlargement with infiltrative erythematous and skin-colored papules and nodules extending onto the vermilion border. B, Hematoxylin-eosin stain (original magnification ×40).
what is your diagnosis?
What is your diagnosis?
Granulomatous cheilitis
Pyostomatitis vegetans
Leukemia labialis
Lip licker’s dermatitis
c
0
1
1
1
male
0
0
65
61-70
null
558
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2724217
A woman in her 50s with a right lower-eyelid yellow plaque presented with unremitting abdominal and back pain. Computed tomographic (CT) scan of the chest, abdomen, and pelvis demonstrated extensive infiltrative stranding from the thorax through the retroperitoneal space. Sclerotic changes in multiple osseous structures were noted. Idiopathic retroperitoneal fibrosis was diagnosed following biopsies. Prednisone and azathioprine were started. Biopsy of the lower-eyelid yellow plaque returned xanthelasma (Figure, A). Ureteral obstruction, chronic kidney disease, and lymphedema complicated her course. Azathioprine was stopped after 1 year. Prednisone was slowly tapered but she relapsed 3 years later. A second retroperitoneal biopsy exhibited fibroadipose tissue, foamy macrophages, and chronic inflammation with fibrosis. Azathioprine and prednisone were restarted. She required multiple surgeries for spinal stenosis and ureteral obstruction. Her kidney function continued declining. Then, owing to knee pain, a plain radiograph was obtained which demonstrated osteosclerosis of the distal femur. Consequently, the previous eyelid and retroperitoneal biopsies were reviewed. The clinicopathologic presentation was confirmed with a molecular test.A, Clinical image of a soft, nonscaly yellow-tan plaque on the medial canthus. B, Hematoxylin-eosin stain image demonstrating foamy histiocytes with Touton giant cells (original magnification ×100). C, CD68 immunohistochemical stain diffusely positive in the histiocytic infiltrate (original magnification ×100). D, Computed tomographic image with contrast demonstrating retroperitoneal stranding and fibrosis with hydronephrosis (arrowheads indicate hairy kidney appearance of fibrosis). What Is Your Diagnosis?
Necrobiotic xanthogranuloma
Xanthelasma palpebrum
Erdheim-Chester disease
Langerhans cell histiocytosis
C. Erdheim-Chester disease
C
Erdheim-Chester disease
The eyelid biopsy demonstrated expanded papillary and reticular dermis filled with foamy histiocytes in a background of lymphocytic infiltrate and scattered Touton giant cells (Figure, B). Histiocytes had positive results for CD68 (Figure, C) and factor XIIIa, and negative results for S100, CD1a. The CT findings showed extensive retroperitoneal fibrosis and a hairy kidney appearance (Figure, D). The DNA extracted from formalin-fixed, paraffin-embedded skin and retroperitoneal biopsies was amplified using allele-specific fluorescent PCR. Via capillary electrophoresis BRAF V600E mutation was detected. The retroperitoneal biopsy was similar to the eyelid demonstrating sheets of foamy histiocytes, scattered lymphocytic infiltrate, and fibrosis.Vemurafenib, 480 mg, twice daily was started following Erdheim-Chester disease (ECD) confirmation. BRAF inhibitor (BRAFi) therapy was initially tolerated for months, but despite intermittent prednisone fatigue and inflammatory arthropathy eventually prompted a drug holiday. Two dose reductions, first to 240 mg twice daily, then 240 mg daily failed to relieve her arthritis. Vemurafenib was discontinued and methotrexate initiated. Repeated CT scans showed stable disease. Her cutaneous lesion resolved.Erdheim-Chester disease was first described in 1930 by Austrian pathologist Jakob Erdheim and his student William Chester.1 Hundreds of cases highlight profound clinical variability in this L group of diseases, non–Langerhans cell histiocytosis, from bone limited disease to multisystem involvement with a poor prognosis.1-3Erdheim-Chester disease is more common in men, and most often presents in the sixth decade of life.1,2 The pathophysiology is characterized as a clonal neoplasia of histiocytes, which generate a robust Th1 cytokine profile.1 Histologically, cutaneous biopsy findings show dermal infiltration with foamy histiocytes and scattered Touton giant cells.4 In contrast, Langerhans cell histiocytosis shows positive eosinophilic histiocytes displaying kidney-bean shaped nuclei in a background of eosinophilic infiltrate. The ECD histiocytes are immunoreactive for CD68, CD163, and Factor XIIIa, and are negative for S100, CD1a, and Langerin. BRAF V600E driver mutations lend evidence for the neoplastic theory.1,5,6Patients diagnosed with necrobiotic xanthogranuloma (NXG) most commonly have an underlying monoclonal gammopathy and histologically show layered necrobiosis and peripheral multinucleated giant cells with haphazardly arranged nuclei. Clinically, both histiocytoses have a periorbital predilection for cutaneous involvement.Cutaneous involvement is present in 32% of ECD cases.4 In patients with skin involvement, it is the first sign of disease in 30%. The Xanthelasma-like lesion (XLL) is specific, as seen in this patient, and present in approximately 25%, most commonly on the inner canthus.4 The XLL can be difficult to distinguish from xanthelasma palpebrum, and was subtle in this case. Both display CD68+, CD163+, and Cd1a− histiocytes. The infiltrate in ECD involves the reticular dermis and has a higher density of multinucleated and Touton giant cells, less dermal fibrosis, and is less likely associated with dyslipidemia.4 Other cutaneous manifestations include erythematous-brownish localized or disseminated patches, papulonodules, and rarely erythroderma.4 Pediatric cases are described, some with cutaneous involvement.7 Several reports of concomitant LCH and ECD exist, and classic lesions of LCH may be present and should be sought for biopsy in any patient with ECD with cutaneous involvement.4,5BRAF V600E mutations are present in 54% to 60% of patients with ECD,6 and seen in considerably larger percentages of patients with XLL.4 Mutations are less common in other cutaneous lesions.4 An XLL biopsy can be easily undertaken, vs biopsy of bone, retroperitoneum, or other internal manifestation.Interferon-α (IFN-α) was the first-line treatment since the early 2000s, and the first agent to show increased survival.1,2 Other treatments include high-dose steroids, cladribine, imatanib, anakinra, tofacitinib, cobimetinib, methotrexate, and cyclophosphamide.1,2,8,9 Efficacy is difficult to ascertain given the relatively small numbers studied. However, since 2012 with discovery of BRAF mutations in most patients with ECD,6BRAF inhibitors have become a promising first-line modality. In the largest study to date, the LOVE study,10 vemurafanib and dabrafenib showed a high response rate of 91% in patients with BRAF-mutations. This included 76% partial response and 15% complete response.10 In addition, cobimetinib, an MEK inhibitor, had suggested benefit in patients with wild-type mutations, in combination with BRAF inhibitors in BRAF-mutants, and in those who had no improvement with BRAF-inhibition therapy.10 Relapse after interruption of BRAF inhibition is standard; however, all patients in the LOVE study subsequently responded again after reintroduction of therapy. Adverse events to BRAF inhibition were similar to prior studies of melanoma.10Dermatologists can recognize cutaneous manifestations of ECD, confirm BRAF mutations via skin biopsy, and aid in diagnosis and treatment of cutaneous adverse reactions associated with BRAF inhibition.
Dermatology
A woman in her 50s with a right lower-eyelid yellow plaque presented with unremitting abdominal and back pain. Computed tomographic (CT) scan of the chest, abdomen, and pelvis demonstrated extensive infiltrative stranding from the thorax through the retroperitoneal space. Sclerotic changes in multiple osseous structures were noted. Idiopathic retroperitoneal fibrosis was diagnosed following biopsies. Prednisone and azathioprine were started. Biopsy of the lower-eyelid yellow plaque returned xanthelasma (Figure, A). Ureteral obstruction, chronic kidney disease, and lymphedema complicated her course. Azathioprine was stopped after 1 year. Prednisone was slowly tapered but she relapsed 3 years later. A second retroperitoneal biopsy exhibited fibroadipose tissue, foamy macrophages, and chronic inflammation with fibrosis. Azathioprine and prednisone were restarted. She required multiple surgeries for spinal stenosis and ureteral obstruction. Her kidney function continued declining. Then, owing to knee pain, a plain radiograph was obtained which demonstrated osteosclerosis of the distal femur. Consequently, the previous eyelid and retroperitoneal biopsies were reviewed. The clinicopathologic presentation was confirmed with a molecular test.A, Clinical image of a soft, nonscaly yellow-tan plaque on the medial canthus. B, Hematoxylin-eosin stain image demonstrating foamy histiocytes with Touton giant cells (original magnification ×100). C, CD68 immunohistochemical stain diffusely positive in the histiocytic infiltrate (original magnification ×100). D, Computed tomographic image with contrast demonstrating retroperitoneal stranding and fibrosis with hydronephrosis (arrowheads indicate hairy kidney appearance of fibrosis).
what is your diagnosis?
What is your diagnosis?
Erdheim-Chester disease
Langerhans cell histiocytosis
Xanthelasma palpebrum
Necrobiotic xanthogranuloma
a
1
1
1
1
female
0
0
55
51-60
null
559
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2723492
A Japanese man in his 50s presented with a 6-month history of worsening pruritic hand dermatitis that progressed to involve his face, trunk, upper extremities, and feet. He had emigrated from Japan more than 10 years earlier but had no history of recent travel. He took no medications, had no significant medical or surgical history, and denied personal and family history of atopy. While he worked as a biomedical researcher, he denied any caustic exposures. On physical examination, his face, trunk, upper extremities, palms, and soles were marked by 1- to 2-mm erythematous papules and pustules coalescing into large, partially indurated and polycyclic plaques with centrifugal extension and occasional central clearing (Figure, A and B). Treatment with potent topical corticosteroids was ineffective. Punch biopsies of lesions on the shoulder (Figure, C) and foot (Figure, D) were performed.A, Annular erythematous plaque studded with papules and pustules on the cheek and temple. B, Erythematous plaques studded with papules and pustules on the shoulder. C and D, Lesional specimens under hematoxylin-eosin stain taken from the shoulder (C, original magnification ×400) and plantar foot (D, original magnification ×40). What Is Your Diagnosis?
Folliculotropic mycosis fungoides
Papulopustular rosacea
Eosinophilic pustular folliculitis
Majocchi granuloma
C. Eosinophilic pustular folliculitis (EPF)
C
Eosinophilic pustular folliculitis
Histopathologic analysis of a follicular papule on the shoulder demonstrated a perivascular and periadnexal eosinophilic and lymphocytic infiltrate concentrated at the follicular isthmus and sebaceous glands (Figure, C). A plaque on the glabrous plantar foot showed eosinophilic spongiosis and subcorneal and intraepidermal pustules (Figure, D). Tissue stains and cultures were negative for bacterial, mycobacterial, and fungal organisms. Neither lesional tissue nor blood demonstrated a monoclonal T-cell population on T-cell receptor (TCR) gene rearrangement studies. Flow cytometry did not detect an atypical B- or T-cell population in the blood. Laboratory workup was notable for an elevated absolute eosinophil count of 1100/μL (1.1 × 109/L). Complete blood cell counts, results of a comprehensive metabolic panel, and lactate dehydrogenase levels were otherwise unremarkable. Findings of human immunodeficiency virus (HIV) testing were negative. Collectively, these findings were compatible with a diagnosis of classic EPF, or Ofuji disease.After diagnosis, the patient began treatment with indomethacin, 50 mg/d. Within 1 month, he had demonstrated nearly complete resolution of all lesions with improvement in pruritus and resolution of eosinophilia. The condition was similarly well controlled by 6-month follow-up.Eosinophilic pustular folliculitis is a noninfectious inflammatory dermatosis first characterized in Japan in 1970 by Ofuji et al.1 Over 300 cases of EPF have been reported, with more than 100 cases in Japanese patients.2 Eosinophilic pustular folliculitis is classified into 3 variants: (1) classic EPF (Ofuji disease), which typically affects healthy Japanese adults; (2) immunosuppression-associated EPF, which is often associated with HIV; and (3) infancy-associated EPF.3 Classic EPF peaks in incidence during the third and fourth decades of life.4 Among Japanese patients, the male-to-female ratio approximates 5 to 1.5Clinically, classic EPF appears as recurrent papules and pustules on an erythematous base, which gradually become confluent to form indurated, annular plaques.6 Eruptions most commonly occur on the face (>80%), with frequent involvement of the trunk and extremities.2 Palmoplantar pustules occur in approximately one-fifth of patients despite the lack of follicles in these areas.2,5 There is no systemic involvement.5 Individual plaques typically last for 7 to 10 days, recur every 3 to 4 weeks, and are often pruritic.5Laboratory findings in classic EPF include leukocytosis with eosinophilia in up to 35% of patients, which normalize as skin lesions improve.2,4,6 Histologically, there is a periadnexal infiltrate of eosinophils and lymphocytes primarily involving the follicular isthmus, infundibulum, and sebaceous glands.3,5 On follicle-bearing areas, this infiltrate may form eosinophilic microabscesses, invade the pilosebaceous unit, and lead to destruction of the follicles in advanced cases.3,6 On follicle-free palmoplantar surfaces, histologic findings include eosinophilic spongiosis and subcorneal and intraepidermal pustules with eosinophils and neutrophils.7Differential diagnoses include fungal and bacterial folliculitides, parasitic infestations, drug eruption, rosacea, folliculotropic mycosis fungoides, and follicular mucinosis.3 General diagnostic workup includes complete blood cell count with differential, HIV testing, and skin punch biopsy.3Follicular mycosis fungoides can present clinically with pink indurated plaques with follicular involvement, demonstrating eosinophilic folliculitis and pustular changes on histologic analysis. However, the T-cell infiltrate often shows atypia and epidermotropism, and TCR gene rearrangement studies demonstrate a clonal T-cell population.8 Histopathologic analysis of papulopustular rosacea typically demonstrates a dense follicular and perivascular infiltrate composed of lymphocytes, plasma cells, and neutrophils without eosinophils.9 Majocchi granuloma is a clinical mimicker, but histologically, it demonstrates perifollicular granulomatous inflammation with numerous fungal organisms.10While the cause of EPF is uncertain, it has been proposed to be the result of immune dysregulation surrounding sebocytes, with excess prostaglandin D2 stimulating sebocyte release of the eosinophil chemoattractant, eotaxin-3.2 Indomethacin inhibits cyclooxygenase and arachidonic acid metabolites, including prostaglandin D2, thus disrupting this process.4,6 Oral indomethacin (25-75 mg/d) is suggested as first-line treatment for classic EPF,2-4,6 demonstrating clinical efficacy in numerous cases.2,3 The time to initial response to oral indomethacin is estimated from several days6 to 2 to 6 weeks.4 In patients with kidney failure or peptic ulcers, topical indomethacin can be considered as an alternative first-line treatment.3Eosinophilic pustular folliculitis should be considered as a diagnosis for recurrent pruritic plaques studded with papules and pustules, particularly in Japanese patients. Biopsy is necessary to make the diagnosis, and testing for HIV is required to differentiate classic EPF from immunosuppression-associated EPF in adult patients. In this case, the patient’s history, presentation, histopathologic findings, and clinical course with response to indomethacin were all characteristic of EPF. It is important to consider EPF in cases with palmoplantar involvement with eosinophilic pustulosis rather than folliculitis in these locations.
Dermatology
A Japanese man in his 50s presented with a 6-month history of worsening pruritic hand dermatitis that progressed to involve his face, trunk, upper extremities, and feet. He had emigrated from Japan more than 10 years earlier but had no history of recent travel. He took no medications, had no significant medical or surgical history, and denied personal and family history of atopy. While he worked as a biomedical researcher, he denied any caustic exposures. On physical examination, his face, trunk, upper extremities, palms, and soles were marked by 1- to 2-mm erythematous papules and pustules coalescing into large, partially indurated and polycyclic plaques with centrifugal extension and occasional central clearing (Figure, A and B). Treatment with potent topical corticosteroids was ineffective. Punch biopsies of lesions on the shoulder (Figure, C) and foot (Figure, D) were performed.A, Annular erythematous plaque studded with papules and pustules on the cheek and temple. B, Erythematous plaques studded with papules and pustules on the shoulder. C and D, Lesional specimens under hematoxylin-eosin stain taken from the shoulder (C, original magnification ×400) and plantar foot (D, original magnification ×40).
what is your diagnosis?
What is your diagnosis?
Papulopustular rosacea
Folliculotropic mycosis fungoides
Majocchi granuloma
Eosinophilic pustular folliculitis
d
0
0
0
1
male
0
0
55
51-60
Japanese
560
original
https://jamanetwork.com/journals/jamaneurology/fullarticle/2727297
A 42-year-old white woman with a history of episodic migraine with visual aura presented for evaluation of transient right upper extremity weakness and word-finding difficulty associated with headache. She admitted to having developed new intermittent vertigo and more frequent and severe throbbing migraine with transient blurry vision and nonpulsatile tinnitus several months prior. She had recently developed hearing loss with a robotic quality of auditory perception. She denied mouth or genital ulcers. A neurologic examination revealed diminished hearing to a finger rub on the right side. Fundoscopy showed several segmental retinal arterial plaques, with the right side worse than the left. A brain magnetic resonance image (MRI) with gadolinium revealed multiple white-matter lesions that were hyperintense on diffusion-weighted imaging (DWI), including some with enhancement, as well as a small DWI-negative lesion in the left thalamus and corpus callosum that was hyperintense on T2/fluid-attenuated inversion recovery magnetic resonance imaging (FLAIR; Figure 1). An MRI of the spine with gadolinium did not reveal a signal abnormality in the spinal cord. Infectious, rheumatologic, and hypercoaguable test results were negative. A lumbar puncture revealed a white blood cell count of 8 cells/μL (normal range, 0-5 cells/μL; to convert to cells × 109/L, multiply by 0.001) with 78% lymphocytes and 22% monocytes (to calculate these as proportions of 1.0, multiply by 0.01), and a protein level of 0.067 g/dL (normal range, 0.015-0.045 g/dL; to convert to grams per liter, multiply by 10). Cerebrospinal fluid (CSF) glucose and IgG index test results were normal, with culture and oligoclonal bands negative. An audiogram revealed bilateral sensorineural hearing loss, with the right side worse than the left. Retinal fluorescein angiography was obtained.A, Pink arrowheads indicate hyperintensities in the right centrum semiovale. B, The yellow arrowhead indicates a hyperintensity in the corpus callosum.Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) What Is Your Diagnosis?
Multiple sclerosis
Behçet disease
Susac syndrome
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL)
C. Susac syndrome
C
Susac syndrome
Susac syndrome is a rare disorder caused by occlusions of microvessels in the brain, retina, and inner ear, with an autoimmune causative mechanism presumed based on signs of inflammation in biopsies of the brain and CSF testing, as well as contrast enhancement on retinal and brain imaging and case reports of good response to immunosuppressive and immunomodulatory therapies.1 John O. Susac, MD, described the initial case in the mid-1970s after he encountered a young woman with a previously unreported triad of encephalopathy, branch retinal artery occlusions, and deafness.2 Since then, more than 300 cases of Susac syndrome have been reported in the literature worldwide with female individuals (aged 20-40 years) affected more frequently than male individuals.1 The prevalence of this disease may not be as rare as once thought, given that the full clinical triad rarely exists at time of initial presentation and many cases are often being misdiagnosed as multiple sclerosis.3-6At clinical onset, the most common manifestation is central nervous system symptoms, followed by visual and hearing or vestibular disturbances. Headaches are present at disease onset in more than 80% of patients and often resemble migraine. Central nervous system symptoms can include focal neurological deficits owing to transient ischemic attack or stroke; later in the disease, patients can develop global deficits (eg, encephalopathy, dementia).1,7,8 Magnetic resonance imaging of the brain typically shows punctate hyperintense deep white and gray matter lesions consistent with microinfarctions on T2/FLAIR, some of which may have restricted diffusion suggestive of acute ischemia and contrast enhancement. Involvement of the central portion of the corpus callosum, rather than the periphery (as in multiple sclerosis), as evidenced by presence of so-called snowball lesions (Figure 1), is considered a characteristic sign of Susac syndrome.1 Echocardiography is usually low yield, because the deep location of the microinfarctions, including the corpus callosum, makes cardioembolism less likely. Cerebrospinal fluid studies may show pleocytosis and elevated protein, typically without oligoclonal bands. Catheter-based cerebral angiography results are usually normal. An audiogram usually shows bilateral sensorineural hearing loss. Retinal fluorescein angiography reveals unilateral or bilateral branched retinal artery occlusions and/or arterial wall hyperfluorescence (Figure 2) owing to microvasculitis, even in patients who are visually asymptomatic.1 Fundoscopy typically shows branched retinal artery occlusions as well as retinal arterial atheromatous plaques, ischemic fluffy white patches, and/or tiny hemorrhages.8Retinal fluorescein angiogram of the right eye with leakage of fluorescein dye through the wall of a branch retinal artery (pink arrowhead) and branch retinal artery occlusive disease inferiorly and temporally (yellow arrowheads).Susac syndrome is frequently misdiagnosed as multiple sclerosis owing to the presence of white matter lesions, corpus callosum involvement, and CSF pleocytosis; however, based on current multiple sclerosis diagnostic criteria,9 the absence of oligoclonal bands, involvement of deep gray matter, and presence of retinal and auditory pathology argue against this diagnosis. Recurrent oral and genital aphthous ulcers, uveitis or retinal vasculitis, pathergy, and skin or vascular lesions, characterize Behçet disease.10 The lack of ulcers and presence of corpus callosum pathology makes the diagnosis of Behçet disease unlikely. The imaging phenotype and involvement of hearing do not support the diagnosis of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Genetic testing for CADASIL may be considered in patients with cryptogenic stroke with appropriate MRI findings; in this case, CADASIL genetic testing was not ordered based on clinical phenotype favoring Susac syndrome.There is no consensus on the treatment of Susac syndrome because of the lack of large well-designed studies addressing this rare, underreported disease. Based on the idea that Susac syndrome may have an inflammatory causative mechanism, some authors have reported variable benefit with immunosuppressive agents.5,8The patient has had improvement in auditory symptoms. She has remained clinically stable while receiving therapy with mycophenolate mofetil and a tapering dosage of oral prednisone.
Neurology
A 42-year-old white woman with a history of episodic migraine with visual aura presented for evaluation of transient right upper extremity weakness and word-finding difficulty associated with headache. She admitted to having developed new intermittent vertigo and more frequent and severe throbbing migraine with transient blurry vision and nonpulsatile tinnitus several months prior. She had recently developed hearing loss with a robotic quality of auditory perception. She denied mouth or genital ulcers. A neurologic examination revealed diminished hearing to a finger rub on the right side. Fundoscopy showed several segmental retinal arterial plaques, with the right side worse than the left. A brain magnetic resonance image (MRI) with gadolinium revealed multiple white-matter lesions that were hyperintense on diffusion-weighted imaging (DWI), including some with enhancement, as well as a small DWI-negative lesion in the left thalamus and corpus callosum that was hyperintense on T2/fluid-attenuated inversion recovery magnetic resonance imaging (FLAIR; Figure 1). An MRI of the spine with gadolinium did not reveal a signal abnormality in the spinal cord. Infectious, rheumatologic, and hypercoaguable test results were negative. A lumbar puncture revealed a white blood cell count of 8 cells/μL (normal range, 0-5 cells/μL; to convert to cells × 109/L, multiply by 0.001) with 78% lymphocytes and 22% monocytes (to calculate these as proportions of 1.0, multiply by 0.01), and a protein level of 0.067 g/dL (normal range, 0.015-0.045 g/dL; to convert to grams per liter, multiply by 10). Cerebrospinal fluid (CSF) glucose and IgG index test results were normal, with culture and oligoclonal bands negative. An audiogram revealed bilateral sensorineural hearing loss, with the right side worse than the left. Retinal fluorescein angiography was obtained.A, Pink arrowheads indicate hyperintensities in the right centrum semiovale. B, The yellow arrowhead indicates a hyperintensity in the corpus callosum.Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL)
what is your diagnosis?
What is your diagnosis?
Behçet disease
Susac syndrome
Multiple sclerosis
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL)
b
1
1
1
1
female
0
0
42
41-50
White
561
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2723593
A woman in her 20s was referred by the optician to our acute and emergency department for suspected optic neuritis in the right eye. She reported a sudden onset of photopsia and visual disturbances described as “fixed swirl circles” in the central vision of both eyes noticed on awakening 5 days prior and persisting since that time. Her medical history included migraine and polycystic ovaries. She had a flulike episode 1 week prior to the onset of symptoms. The patient was taking oral contraceptives and denied the use of recreational drugs. Ocular movements were full but the patient reported pain and discomfort during movements of the right eye. Pupils were equal and reactive to light and there was no afferent pupillary defect. Best-corrected visual acuity was 20/20 OU; color vision was 17/17 OU on Ishishara plates. The anterior segment was unremarkable and intraocular pressure was 14 mm Hg in both eyes. Dilated fundus examination revealed a healthy optic disc and no obvious abnormalities at the macula of both eyes. Near-infrared reflectance imaging and spectral-domain optical coherence tomography (OCT) scans of the right eye are shown in the Figure.A, Near-infrared reflectance imaging of acute macular neuroretinopathy in the right eye at presentation reveals hyporeflective teardrop-shaped lesions (black arrowheads). B, Simultaneous spectral-domain optical coherence tomography scan passing through the acute macular neuroretinopathy lesions in the right eye reveals increased reflectivity of the outer plexiform layer (yellow arrowheads) and attenuation of the ellipsoid zone (white arrowheads). What Would You Do Next?
Perform fluorescein angiography
Observe the patient
Perform magnetic resonance imaging of the brain
Start a course of oral corticosteroids
Acute macular neuroretinopathy (AMN)
B
Observe the patient
Sudden-onset paracentral scotoma, good visual acuity, and the presence of wedge-shaped paracentral hyporeflective lesions on infrared reflectance imaging associated with hyperreflectivity of the junction of the outer plexiform layer and the outer nuclear layer on OCT were consistent with AMN. Observation (choice B) was the appropriate next step for this case. Fluorescein angiography (choice A) would not be the preferred answer as the next step because AMN is typically not detected on fluorescein angiography. Magnetic resonance imaging of the brain (choice C) was not recommended as the next step because diagnosis of AMN per se does not require brain imaging unless there are symptoms and/or signs suggestive of central nervous system involvement. There is no proven treatment for AMN; thus, treatment with oral corticosteroids (choice D) was not indicated.First described by Bos and Deutman in 1975,1 AMN is a rare condition that typically affects young women. The clinical presentation is with flat, reddish, wedge-shaped or teardrop-shaped intraretinal lesions usually pointing toward the fovea and best seen using near-infrared reflectance imaging.2 These lesions may be very subtle, and fundus examination may be unremarkable,3 as in the present patient.The patient was referred to us with a provisional diagnosis of optic neuritis in the right eye because of pain during ocular movements and sudden-onset scotoma. However, the patient’s clinical presentation was not consistent with optic neuritis in the right eye; the patient’s visual acuity and color vision were unaffected and there was no relative afferent pupillary defect. Moreover, pain during ocular movements has been reported in association with AMN at presentation,4 although it is uncommon.The clinical course of AMN is characterized by persistence of paracentral scotoma but patients typically retain good visual acuity. Optical coherence tomography is useful to monitor the natural course of the condition. In the acute phase of the disease, AMN typically presents on OCT with hyperreflective bands at the level of the junction of the outer plexiform layer and the outer nuclear layer. Subsequent attenuation of the outer nuclear layer and ellipsoid zone are the hallmark of this condition in the postacute phase of the disease.5The pathogenesis of AMN is still a matter of debate. Several risk factors, including use of oral contraceptives, preceding flulike illness, exposure to either epinephrine or ephedrine, intake of lisdexamphetamine, use of cocaine, antecedent trauma, systemic shock, and preeclampsia, have been associated with AMN.6Fawzi et al5 first described longitudinal features of AMN seen on results of OCT and showed that the earliest finding in AMN occurs at the level of the outer plexiform layer in the context of a completely normal outer retina and before the appearance of lesions on results of near-infrared reflectance imaging. Given the early involvement of the outer plexiform layer, a local impairment of the deep retinal circulation had been suggested as the possible cause of these lesions.5 However, recent reports using OCT angiography found that flow abnormalities in the choriocapillaris colocalize with the alterations of the AMN lesions seen on results of OCT and suggested a vascular insult in the choriocapillaris as the pathogenic mechanism of the AMN lesions.7-9Acute macular neuroretinopathy should be suspected in young healthy women with a sudden onset of paracentral scotoma and unremarkable clinical examination. Recognition of AMN in these patients may avoid unnecessary investigations. Identification of risk factors for AMN and its characteristic features on near-infrared reflectance imaging and OCT are critical for the recognition of this uncommon condition.The patient was reexamined after 4 weeks. Her best-corrected visual acuity was 20/20 OU. She denied ocular pain during ocular movements and reported the persistence of paracentral scotoma.
Ophthalmology
A woman in her 20s was referred by the optician to our acute and emergency department for suspected optic neuritis in the right eye. She reported a sudden onset of photopsia and visual disturbances described as “fixed swirl circles” in the central vision of both eyes noticed on awakening 5 days prior and persisting since that time. Her medical history included migraine and polycystic ovaries. She had a flulike episode 1 week prior to the onset of symptoms. The patient was taking oral contraceptives and denied the use of recreational drugs. Ocular movements were full but the patient reported pain and discomfort during movements of the right eye. Pupils were equal and reactive to light and there was no afferent pupillary defect. Best-corrected visual acuity was 20/20 OU; color vision was 17/17 OU on Ishishara plates. The anterior segment was unremarkable and intraocular pressure was 14 mm Hg in both eyes. Dilated fundus examination revealed a healthy optic disc and no obvious abnormalities at the macula of both eyes. Near-infrared reflectance imaging and spectral-domain optical coherence tomography (OCT) scans of the right eye are shown in the Figure.A, Near-infrared reflectance imaging of acute macular neuroretinopathy in the right eye at presentation reveals hyporeflective teardrop-shaped lesions (black arrowheads). B, Simultaneous spectral-domain optical coherence tomography scan passing through the acute macular neuroretinopathy lesions in the right eye reveals increased reflectivity of the outer plexiform layer (yellow arrowheads) and attenuation of the ellipsoid zone (white arrowheads).
what would you do next?
What would you do next?
Start a course of oral corticosteroids
Perform magnetic resonance imaging of the brain
Perform fluorescein angiography
Observe the patient
d
0
1
1
1
female
0
0
25
21-30
White
562
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2723990
A 44-year-old man with a 2-week history of fevers, night sweats, chills, and petechiae presented with painless, blurry vision in the left eye. His ocular symptoms began 2 days prior when he noticed dark scotomas in his central field and decreased visual acuity. There was no recent trauma or strenuous physical activity. He had no relevant medical, ocular, or family history, and he denied use of illicit drugs or anticoagulants.Uncorrected visual acuity was 20/30 OD and 20/400 OS. Extraocular motility and intraocular pressures were normal. Results of an anterior examination was unremarkable. Dilated funduscopic examination of the left eye revealed a pink optic nerve with sharp margins, diffuse intraretinal hemorrhages and Roth spots extending from the posterior pole to the far periphery, and preretinal hemorrhage overlying the central macula (Figure, A). Spectral-domain optical coherence tomography through the preretinal macular hemorrhage demonstrated layered blood in the sub–internal limiting membrane space (Figure, B). The right fundus exhibited a similar appearance aside from the preretinal hemorrhage.A, Bedside indirect ophthalmoscopy (slightly out of focus) of the left eye with a 20-diopter lens reveals diffuse retinal hemorrhages; the white arrowhead indicates hemorrhage corresponding to Figure, B and the blue arrowheads, Roth spots in the posterior pole with a normal-appearing optic nerve. Findings extended to the far periphery. B, Spectral-domain optical coherence tomography through macular hemorrhage reveals layering of blood in the sub–internal limiting membrane space of the left retina.Obtain a fluorescein angiogram to evaluate degree of ischemia and confirm a diagnosis of central retinal vein occlusionProvide the patient with stool softeners to prevent recurrence of Valsalva retinopathyImmediately begin intravenous antibiotics before any blood draws and consult cardiology for likely bacterial endocarditisDraw a complete blood cell count with differential to evaluate for leukemic retinopathy or blood dyscrasia What Would You Do Next?
Obtain a fluorescein angiogram to evaluate degree of ischemia and confirm a diagnosis of central retinal vein occlusion
Provide the patient with stool softeners to prevent recurrence of Valsalva retinopathy
Immediately begin intravenous antibiotics before any blood draws and consult cardiology for likely bacterial endocarditis
Draw a complete blood cell count with differential to evaluate for leukemic retinopathy or blood dyscrasia
Acute myeloid leukemia
D
Draw a complete blood cell count with differential to evaluate for leukemic retinopathy or blood dyscrasia
This patient had leukocytosis (white blood cells, 61 700/μL [to convert to cells × 109/L, multiply by 0.001]; normal limits, 4500-12 000/μL) with anemia (red blood cells, 2.71 × 106/μL [to convert to cell × 1012/L, multiply by 1.0]; normal limits, 3.9-5.5 × 106/μL; hemoglobin, 8.9 g/dL [to convert to g/L, multiply by 10.0]; normal limits, 14-18 g/dL; hematocrit, 26.8% [to convert to a proportion of 1.0, convert to 0.01]; normal limits, 40%-54%) and thrombocytopenia (platelet, 3.1 × 103/μL [to convert to cells × 109/L, multiply by 1.0]; normal limits, 150-400 × 103/μL). There were increases in the absolute number of neutrophils (7400/μL [to convert to cells × 109/L,multiply by 0.001]; normal limits, 17 800-53 600/μL), monocytes (18 040/μL [to convert to cells × 109/L, multiply by 0.001]; normal limits, 300-820/μL), basophils (620/μL [to convert to cells × 109/L, multiply by 0.001]; normal limits, 10-80/μL), and eosinophils (3700/μL [to convert to cells × 109/L, multiply by 0.001]; normal limits, 40-540/μL), with a normal lymphocyte count (2470/μL [to convert to  × 109/L, multiply by 0.001]; normal limits, 1320-3570/μL). Peripheral blood smear showed 35% blasts, while bone marrow flow cytometry revealed 50% abnormal monocytes and 23% myeloblasts. All findings were consistent with a diagnosis of acute myeloid leukemia with monocytic features; cytogenetic analysis revealed an inversion on chromosome 16, which has been associated with favorable prognostic outcomes.1Ocular involvement (often asymptomatic) is present in up to 50% of patients with leukemia at time of diagnosis, with retinal lesions the most common ocular manifestation.2 Leukemic retinopathy is characterized by retinal hemorrhages in the posterior pole secondary to anemia and thrombocytopenia. These hemorrhages are most commonly intraretinal but can also be subretinal, sub–internal limiting membrane, or subhyaloid.3 Other signs of leukemic retinopathy include Roth spots (retinal hemorrhages with central white spots), cotton wool spots, retinal vein tortuosity, microaneurysms, and neovascularization.4 Infiltration of the optic nerve is rare but a poor prognostic sign if observed.Other diagnoses that can manifest similar features include central retinal vein occlusion, Valsalva retinopathy, bacterial endocarditis, hypertensive retinopathy, and diabetic retinopathy. However, the presence of Roth spots with described systemic symptoms are atypical for any of these besides bacterial endocarditis, so fluorescein angiogram for evaluation of central retinal vein occlusion (choice A) and stool softeners for Valsalva retinopathy (choice B) would not be recommended next steps. While bacterial endocarditis should have been considered for this patient, initiation of antibiotics without first obtaining blood tests to rule out another causative mechanism (choice C) would be inappropriate. If leukemic retinopathy is suspected, the patient should obtain a complete blood cell count with differential and peripheral blood smear, followed by bone marrow aspiration for confirmation. Flow cytometry immunophenotyping helps distinguish between different types of leukemia, while cytogenetic analysis can provide valuable prognostic information.Treatment for leukemic retinopathy is directed toward the underlying leukemic disease rather than the ocular manifestations. Systemic chemotherapy (often with daunorubicin, cytarabine, and thioguanine) is the mainstay treatment, sometimes combined with adjunctive targeted drug therapy.5 This may be followed by stem cell transplant. Radiation has a limited role in the treatment of acute myeloid leukemia, but external beam radiation therapy may be used if the brain and spinal fluid are involved. The ophthalmic findings and decreased vision almost always resolve with induction of chemotherapy.6In this case report, we describe a patient with an unremarkable medical or ocular history presenting with leukemic retinopathy as the initial finding of acute myeloid leukemia. This case highlights the importance of considering leukemia as a diagnosis in an otherwise healthy patient presenting with retinopathy.Standard 7 + 3 induction chemotherapy was initiated with an intravenous continuous infusion of cytarabine, 100mg/m2, administered on days 1 through 7 and an intravenous bolus of idarubicin, 12mg/m2, concurrently administered on days 1 through 3. At day 43 of induction, the patient underwent a second ophthalmic examination, which showed complete resolution of leukemic retinopathy and restored baseline visual acuity. Ophthalmic return precautions were discussed with the patient and his cancer specialist team as he continues treatment for leukemia.
Ophthalmology
A 44-year-old man with a 2-week history of fevers, night sweats, chills, and petechiae presented with painless, blurry vision in the left eye. His ocular symptoms began 2 days prior when he noticed dark scotomas in his central field and decreased visual acuity. There was no recent trauma or strenuous physical activity. He had no relevant medical, ocular, or family history, and he denied use of illicit drugs or anticoagulants.Uncorrected visual acuity was 20/30 OD and 20/400 OS. Extraocular motility and intraocular pressures were normal. Results of an anterior examination was unremarkable. Dilated funduscopic examination of the left eye revealed a pink optic nerve with sharp margins, diffuse intraretinal hemorrhages and Roth spots extending from the posterior pole to the far periphery, and preretinal hemorrhage overlying the central macula (Figure, A). Spectral-domain optical coherence tomography through the preretinal macular hemorrhage demonstrated layered blood in the sub–internal limiting membrane space (Figure, B). The right fundus exhibited a similar appearance aside from the preretinal hemorrhage.A, Bedside indirect ophthalmoscopy (slightly out of focus) of the left eye with a 20-diopter lens reveals diffuse retinal hemorrhages; the white arrowhead indicates hemorrhage corresponding to Figure, B and the blue arrowheads, Roth spots in the posterior pole with a normal-appearing optic nerve. Findings extended to the far periphery. B, Spectral-domain optical coherence tomography through macular hemorrhage reveals layering of blood in the sub–internal limiting membrane space of the left retina.Obtain a fluorescein angiogram to evaluate degree of ischemia and confirm a diagnosis of central retinal vein occlusionProvide the patient with stool softeners to prevent recurrence of Valsalva retinopathyImmediately begin intravenous antibiotics before any blood draws and consult cardiology for likely bacterial endocarditisDraw a complete blood cell count with differential to evaluate for leukemic retinopathy or blood dyscrasia
what would you do next?
What would you do next?
Obtain a fluorescein angiogram to evaluate degree of ischemia and confirm a diagnosis of central retinal vein occlusion
Draw a complete blood cell count with differential to evaluate for leukemic retinopathy or blood dyscrasia
Immediately begin intravenous antibiotics before any blood draws and consult cardiology for likely bacterial endocarditis
Provide the patient with stool softeners to prevent recurrence of Valsalva retinopathy
b
0
1
1
1
male
0
0
44
41-50
White
563
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2723991
A 65-year-old white man was referred to the clinic with a pigmented conjunctival lesion in the left eye. The referring physician suspected primary acquired melanosis (PAM). His ocular history was notable for occupational exposure to sunlight and occasional smoking. There was no personal or family history of cancer, and he denied ocular surgery or trauma. On examination, his best-corrected visual acuity was 20/20 OD/OS, and intraocular pressures were 15 mm Hg OD and 18 mm Hg OS. A slitlamp examination of the right eye showed no conjunctival pigmentation. Examination of the left eye revealed a pigmented lesion at the temporal limbus from 3 to 4 o’clock with a 2-mm extension into the cornea (Figure 1A). Some opalescence underlied the pigment on the cornea. In addition, there was an adjacent small area of flat, superficial, mobile bulbar conjunctival pigmentation. The anterior segment high-resolution optical coherence tomography (HR-OCT) of the 4-o’clock corneolimbal lesion of the left eye demonstrated thickened hyperreflective corneal epithelium with an abrupt transition from normal to abnormal (Figure 1B).A, Slitlamp photography of the left eye shows a pigmented limbal lesion at 3 to 4 o’clock; another small area of bulbar conjunctival pigmentation is noted (arrowhead). B, High-resolution optical coherence tomography of the left eye through the area of the pigmented conjunctival lesion (inset with red line in area of scan), showing epithelial hyperreflectivity and thickening with an abrupt change from normal to mildly thickened epithelium (arrowhead). What Would You Do Next?
Refer to an oncologist for metastatic workup
Biopsy the conjunctival and corneal lesion
Perform cryotherapy over the lesion
Start oral doxycycline
Ocular surface squamous neoplasia in the left eye and PAM in the left eye
B
Biopsy the conjunctival and corneal lesion
This patient was referred with a diagnosis of primary acquired melanosis. PAM is an acquired flat, noncystic pigmented lesion of the conjunctiva, cornea, or caruncle. About 96% of cases occur in fair-skinned individuals with a mean age of 56 years.1,2 PAM without atypia does not progress to melanoma, whereas PAM with severe atypia has a 13% to 46% risk of progression.3 In this case, after referral for PAM, the subtle opalescence on clinical examination indicated a possible diagnosis of concurrent ocular surface squamous neoplasia (OSSN).Ocular surface squamous neoplasia includes a spectrum of dysplasia, ranging from mild to invasive carcinoma of conjunctiva, limbus, and cornea.4 It typically presents as a unilateral limbal lesion, which can appear papillomatous, gelatinous, or leukoplakic. When on the cornea, it may have an opalescent appearance. In the United States, OSSN is commonly found in white men older than 60 years and is infrequently associated with pigment. In Africa, the disease is seen in black individuals, and OSSN often has accompanying pigment.5 The lesions rarely ever metastasize, and as such, a metastatic workup for this lesion is not indicated (choice A).While this disease is typically diagnosed clinically, HR-OCT can assist in arriving at the correct diagnosis, especially in atypical cases. With this imaging, we could detect that this patient’s lesion was in fact OSSN coexisting with and, in a manner of speaking, hiding in PAM. The results of the patient’s HR-OCT imaging showed epithelial hyperreflectivity with thickening and an abrupt transition from normal to abnormal epithelium, which are classic for OSSN.6 PAM on HR-OCT appears very different, with only a linear basal epithelial hyperreflective band and normal epithelium with no thickening.7The HR-OCT suggested that this was not simply a small area of PAM; thus, a biopsy was performed to confirm these findings (choice B). The biopsy confirmed the HR-OCT findings of corneal OSSN and PAM. The corneal epithelium disclosed faulty maturational sequencing with pleomorphic nuclei that extended to near-full thickness with overlying superficial maturation. Morphologically benign melanin deposition was present within these cells (Figure 2). This case demonstrates how HR-OCT imaging can assist in the diagnosis of conjunctival lesions in complex ocular surface conditions, and gave an ad hoc optical biopsy of the ocular surface.Examination of fragmented, tangentially sectioned corneal epithelium shows faulty maturational sequencing with pleomorphic nuclei that extends to near-full thickness with overlying superficial maturation (asterisk). Morphologically benign melanin deposition is present in these cells (arrowhead); hematoxylin-eosin, original magnification ×600.Ocular surface squamous neoplasia is generally a slow-growing and treatable tumor. Surgery and chemotherapy are the main treatments. Surgery is approached using a no-touch technique with wide margins and adjuvant cryotherapy. Cryotherapy (choice C) alone is not ideal for the treatment of OSSN. Doxycycline (choice D), while helpful for meibomian gland disease, is not a treatment for OSSN or PAM. Topical chemotherapy is now often used as primary therapy.8 Both surgical excision and topical therapy have similar outcomes in terms of recurrence rates.9 The most commonly used agents are interferon–alfa 2b, 5-fluorouracil, and mitomycin C.10After the biopsy confirmed the HR-OCT findings of a corneal OSSN, the patient was treated with a cycle of topical 5-fluorouracil, 1%, 4 times daily for 1 week with 3 weeks off. After 2 cycles, the lesion resolved clinically and by HR-OCT, in that the epithelial thickening and hyperreflectivity were resolved. A total of 4 cycles were given. The patient will be followed up carefully for recurrence of the OSSN clinically and with HR-OCT.
Ophthalmology
A 65-year-old white man was referred to the clinic with a pigmented conjunctival lesion in the left eye. The referring physician suspected primary acquired melanosis (PAM). His ocular history was notable for occupational exposure to sunlight and occasional smoking. There was no personal or family history of cancer, and he denied ocular surgery or trauma. On examination, his best-corrected visual acuity was 20/20 OD/OS, and intraocular pressures were 15 mm Hg OD and 18 mm Hg OS. A slitlamp examination of the right eye showed no conjunctival pigmentation. Examination of the left eye revealed a pigmented lesion at the temporal limbus from 3 to 4 o’clock with a 2-mm extension into the cornea (Figure 1A). Some opalescence underlied the pigment on the cornea. In addition, there was an adjacent small area of flat, superficial, mobile bulbar conjunctival pigmentation. The anterior segment high-resolution optical coherence tomography (HR-OCT) of the 4-o’clock corneolimbal lesion of the left eye demonstrated thickened hyperreflective corneal epithelium with an abrupt transition from normal to abnormal (Figure 1B).A, Slitlamp photography of the left eye shows a pigmented limbal lesion at 3 to 4 o’clock; another small area of bulbar conjunctival pigmentation is noted (arrowhead). B, High-resolution optical coherence tomography of the left eye through the area of the pigmented conjunctival lesion (inset with red line in area of scan), showing epithelial hyperreflectivity and thickening with an abrupt change from normal to mildly thickened epithelium (arrowhead).
what would you do next?
What would you do next?
Start oral doxycycline
Refer to an oncologist for metastatic workup
Perform cryotherapy over the lesion
Biopsy the conjunctival and corneal lesion
d
0
1
1
1
male
0
0
65
61-70
White
564
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2721041
A 70-year old man with a history of soft contact lens wear presented for a general eye examination. Visual acuity was 20/20 OU with normal intraocular pressure. Several giant follicles were noted on the right inferior palpebral conjunctiva, and a few smaller follicles were noted in the left inferior fornix. Papillae were also noted bilaterally on the superior palpebral conjunctiva. He did not report any redness or discharge but did note mildly increased irritation mainly from soft contact lens wear. He had endorsed proper contact lens care and hygiene. He also noted an upper respiratory tract illness 4 weeks before presentation. Initially, he received a giant papillary conjunctivitis diagnosis because of soft contact lens intolerance and a recommendation to take a contact lens break. Because of the papillary reaction, prednisolone acetate, 1%, twice a day for 2 weeks was prescribed.After 3 months, the symptoms had improved in the left eye, but conjunctival follicles were still present in the right inferior palpebral conjunctiva (Figure 1). The patient subsequently underwent testing for chlamydia and gonorrhea, and the result was negative. At the 1-year follow-up, the examination findings were unchanged. During this follow-up, Lyme disease and Epstein-Barr virus serologic tests were performed in addition to repeated chlamydia testing. The results were negative, except for a positive Epstein-Barr virus IgG.Slitlamp photograph showing prominent large follicles (arrowhead) in the right inferior palpebral conjunctiva.Start a longer trial of topical steroid with topical antihistamineOrder chest radiography and an angiotensin-converting enzyme serum level test What Would You Do Next?
Start an empirical trial of oral azithromycin
Start a longer trial of topical steroid with topical antihistamine
Proceed with conjunctival biopsy
Order chest radiography and an angiotensin-converting enzyme serum level test
Conjunctival follicular lymphoma
C
Proceed with conjunctival biopsy
An empirical trial of oral azithromycin (choice A) would be less preferred because of the 2 negative chlamydia testing results. A longer trial of topical steroid with topical antihistamine (choice B) would be less favorable because symptoms and follicle appearance were not consistent with allergic conjunctivitis. Chest radiography and angiotensin-converting enzyme level test (choice D) would not be recommended as the patient’s more translucent follicles did not have the typical appearance of the yellow or tan conjunctival nodules found in ocular sarcoidosis. Because of the chronic nature and abnormal, large nodular appearance of the follicles, conjunctival biopsy was pursued for definitive diagnosis after a negative infectious serologic testing result. This biopsy showed centrocytes consistent with follicular lymphoma (Figure 2). Flow cytometry demonstrated a CD 10–positive monotypic B cell population. Fluorescence in situ hybridization testing detected a t(14;18) translocation. He was subsequently referred to the hematology-oncology service for systemic workup, including hepatitis B and C serologic tests, antinuclear antibody panel test, serum protein electrophoresis, Chlamydophila psittaci serologic test, serum κ or λ light chains, whole-body positron emission tomography–computed tomography, and magnetic resonance imaging of the brain and orbits. The systemic workup results were negative, except for mild cervical lymphadenopathy on the positron emission tomography–computed tomography. After discussion with the patient and because of the lymphoma’s localized indolent course, he elected to undergo observation with follow-up every 3 months.Hematoxylin-eosin stain showing nodular aggregates of centrocytes, with small, slightly rounded lymphoid cells (asterisks) (original magnification ×10).Follicular lymphomas are described pathologically as follicles with cellular pleomorphism and the absence of sharply demarcated germinal centers.1 Ocular adnexal lymphoproliferative lesions range from benign hyperplasia to malignant lymphomas that affect the orbit, eyelid, or conjunctiva. Most of these lesions are found unilaterally in the orbit, and most neoplastic lesions are of monoclonal B cell origin.2 Conjunctival lymphoid lesions are described as mobile salmon-pink or flesh-colored patches in the fornix or bulbar conjunctiva.2-5 Symptoms include irritation, blurred vision, and epiphora and can present as a mass, ptosis, or diplopia.3A greater concern is systemic involvement, with bilateral presentation of ocular adnexal lymphomas. In large institutional case reviews, the incidence of systemic lymphoma findings at the time of local ocular diagnosis or up to 5 years afterward was between 20% to 30%.3,6 Extraocular sites included lymph nodes, bone marrow, abdomen, brain, and lungs. A predictive factor in systemic involvement was the location in the conjunctival fornix or midbulbar conjunctiva.3 With or without treatment, the mortality rate from systemic lymphoma in the long term was less than 5%. With radiation treatment, recurrence of disease was low (0%-20%) in the first 1 to 5 years after the biopsy.6-8The differential diagnosis for chronic conjunctivitis is extensive, including allergic reaction, adverse effect from ophthalmic medications or contact lenses and their cleaning solutions, viral and bacterial etiologies, and blepharoconjunctivitis.9,10 Infectious etiologies include herpesviruses, adenoviruses, cytomegalovirus, Epstein-Barr virus, molluscum contagiosum virus, coxsackieviruses, Chlamydia trachomatis, Moraxella species, and Bartonella henselae. Important elements of medical history and system review include duration, laterality, recent illness, history of allergies, and pet exposure.10 An uncertain diagnosis supports the performance of tissue biopsy for a definitive identification.Ophthalmic and oncologic follow-up for this patient has continued. His examination findings 1 year after the biopsy have been unchanged. To date, no extraocular involvement of follicular lymphoma has been found.
Ophthalmology
A 70-year old man with a history of soft contact lens wear presented for a general eye examination. Visual acuity was 20/20 OU with normal intraocular pressure. Several giant follicles were noted on the right inferior palpebral conjunctiva, and a few smaller follicles were noted in the left inferior fornix. Papillae were also noted bilaterally on the superior palpebral conjunctiva. He did not report any redness or discharge but did note mildly increased irritation mainly from soft contact lens wear. He had endorsed proper contact lens care and hygiene. He also noted an upper respiratory tract illness 4 weeks before presentation. Initially, he received a giant papillary conjunctivitis diagnosis because of soft contact lens intolerance and a recommendation to take a contact lens break. Because of the papillary reaction, prednisolone acetate, 1%, twice a day for 2 weeks was prescribed.After 3 months, the symptoms had improved in the left eye, but conjunctival follicles were still present in the right inferior palpebral conjunctiva (Figure 1). The patient subsequently underwent testing for chlamydia and gonorrhea, and the result was negative. At the 1-year follow-up, the examination findings were unchanged. During this follow-up, Lyme disease and Epstein-Barr virus serologic tests were performed in addition to repeated chlamydia testing. The results were negative, except for a positive Epstein-Barr virus IgG.Slitlamp photograph showing prominent large follicles (arrowhead) in the right inferior palpebral conjunctiva.Start a longer trial of topical steroid with topical antihistamineOrder chest radiography and an angiotensin-converting enzyme serum level test
what would you do next?
What would you do next?
Proceed with conjunctival biopsy
Order chest radiography and an angiotensin-converting enzyme serum level test
Start a longer trial of topical steroid with topical antihistamine
Start an empirical trial of oral azithromycin
a
0
1
1
1
male
0
0
70
61-70
null
565
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2722669
A white man in his 30s employed as a metal worker presented with a 2-month history of slightly blurry vision in his right eye. He denied photopsia or floaters and did not note any visual field defects. His best-corrected visual acuity was 20/25 OD and 20/20 OS. He had no history of ocular trauma, drug use, or familial ocular diseases.The anterior segment of both eyes was normal on slitlamp examination, but the fundus examination of the right eye showed multiple refractile small crystals scattered on the posterior pole (Figure 1A). Isolated crystals were also present in the vitreous core. Optical coherence tomography of the macula revealed the crystals to be located preretinally on the surface of the inner limiting membrane with sporadic crystals within the cortical vitreous (Figure 1B).A, Fundus photography at presentation shows multiple refractile crystals at the posterior pole (black arrowheads). B, The corresponding optic coherence tomography at presentation shows these crystals adhering to the internal limiting membrane and floating in the vitreous (white arrowheads).Apart from this, biomicroscopy, fluorescein angiography, and optical coherence tomography of the posterior pole showed no abnormalities. The left eye was unremarkable, and an radiographic image of the right eye socket revealed no foreign bodies.Screen for intravenous abuse of opioids and other substances What Would You Do Next?
Request a genetic workup
Examine the retinal periphery
Measure oxalate levels in blood and urine
Screen for intravenous abuse of opioids and other substances
Crystalline retinopathy associated with a chronic retinal detachment
B
Examine the retinal periphery
Preretinal crystals are a rare sign of a chronic retinal detachment. In 1998, Ahmed et al1 published a series of 11 cases. All deposits were found no deeper than the inner limiting membrane, and the crystals themselves did not seem to cause any loss of visual acuity. More recently, Habib et al2 analyzed subretinal fluid from a patient with preretinal crystals secondary to a chronic retinal detachment. Owing to the small sample in the study by Habib et al,2 a biochemical assay was not possible, but a microscopic analysis with polarized light showed a visual resemblance of the crystals to calcium oxalate. The current hypothesis is that the crystals originate from the subretinal space, migrate into the vitreous, and then settle at the vitreoretinal interface, predominantly at the macula because of gravitation. There is no evidence that crystalline deposits lower the visual prognosis; however, the retinal detachment might progress.1-4In this case, the examination of the retinal periphery revealed an inferotemporal chronic retinal detachment with pigmented borders. Figure 2 shows the lesion 1 month after subsequent laser photocoagulation.Fundus photography of the periphery; 1 month after laser coagulation (black arrowheads), the inferotemporal chronic retinal detachment was stable.Other causes of refractile crystalline deposits include a variety of differential diagnoses, ranging from systemic disorders to primary ocular disorders. A genetic workup (choice A) for conditions like Sjögren-Larsson syndrome, cystinosis, or Bietti crystalline dystrophy is most likely futile, because these diseases usually present themselves in both eyes. Measuring oxalate levels (choice C) would not be recommended, because oxalosis similarly presents itself bilaterally, and oxalosis crystals are typically located along the retinal arterioles,3 where no crystals were detected in this case. A screening for intravenous abuse of opioids and other substances (choice D) would not yield relevant results, because drug-induced crystalline retinopathies, such as talc retinopathy, present with intraretinal instead of preretinal crystals. The preretinal crystal distribution likewise excludes other possible ocular causes of retinal crystals, such as calcified macular drusen and calcium or cholesterol emboli.3Possible treatments were discussed with the patient, and a barrier laser procedure was performed to wall off the retinal detachment. The detachment remained stable, and the patient’s visual acuity had improved spontaneously to 20/20 OD by a 1-month follow-up visit.
Ophthalmology
A white man in his 30s employed as a metal worker presented with a 2-month history of slightly blurry vision in his right eye. He denied photopsia or floaters and did not note any visual field defects. His best-corrected visual acuity was 20/25 OD and 20/20 OS. He had no history of ocular trauma, drug use, or familial ocular diseases.The anterior segment of both eyes was normal on slitlamp examination, but the fundus examination of the right eye showed multiple refractile small crystals scattered on the posterior pole (Figure 1A). Isolated crystals were also present in the vitreous core. Optical coherence tomography of the macula revealed the crystals to be located preretinally on the surface of the inner limiting membrane with sporadic crystals within the cortical vitreous (Figure 1B).A, Fundus photography at presentation shows multiple refractile crystals at the posterior pole (black arrowheads). B, The corresponding optic coherence tomography at presentation shows these crystals adhering to the internal limiting membrane and floating in the vitreous (white arrowheads).Apart from this, biomicroscopy, fluorescein angiography, and optical coherence tomography of the posterior pole showed no abnormalities. The left eye was unremarkable, and an radiographic image of the right eye socket revealed no foreign bodies.Screen for intravenous abuse of opioids and other substances
what would you do next?
What would you do next?
Request a genetic workup
Examine the retinal periphery
Measure oxalate levels in blood and urine
Screen for intravenous abuse of opioids and other substances
b
1
0
1
1
male
0
0
35
31-40
White
566
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2723092
A previously healthy African American man in his 20s presented to the emergency department after referral by an optometrist for “bleeding in the back of the eye.” The patient noted that 2 months prior he began experiencing throbbing headaches in the back of his head that were more painful and associated with lightheadedness when laying down. Two weeks prior to presentation, he started noticing black spots in his peripheral vision and progressive blurring of vision in both eyes. The patient denied recent travel, cough, gastrointestinal or genitourinary tract symptoms, ulcers, aching joints, tinnitus, or transient visual obscurations. He reported owning cats.On presentation, the patient’s blood pressure was 210/150 mm Hg, and he had a history of acute kidney failure and non–ST-elevation myocardial infarction. On ophthalmic examination, his uncorrected near visual acuity was 20/25 OD and 20/30 OS. Extraocular motility, Ishihara color plates, intraocular pressure, pupils, and results of slitlamp examination were within normal limits. Ophthalmoscopy showed Frisen grade 5 optic nerve head swelling in both eyes with extensive flame hemorrhages and nerve fiber layer infarcts surrounding both nerves and retinal exudates extending from the nerves temporally into both fovea’s (Figure 1). The vessels were of normal course and caliber. There were scattered peripheral dot-and-blot hemorrhages. Results of Humphrey visual field testing were reliable and showed bilateral concentric constriction and enlarged blind spots.Grade 5 optic nerve head edema. A, Fundus photograph of the right eye shows Frisen grade 5 optic disc edema, flame hemorrhages, and peripapillary exudation into the macula. B, Fluorescein angiogram of the right eye in the early venous phase shows blockage from flame hemorrhages, peripapillary microaneurysms, leakage from optic disc capillaries, and absence of vascular leakage at arcades.Control the systemic blood pressure and workup underlying possible cause of the hypertensionPerform magnetic resonance imaging and magnetic resonance venogram of the brain and orbits, then perform lumbar puncture What Would You Do Next?
Control the systemic blood pressure and workup underlying possible cause of the hypertension
Perform blood tests for neuroretinitis
Perform magnetic resonance imaging and magnetic resonance venogram of the brain and orbits, then perform lumbar puncture
All of the above
Papilledema secondary to intracranial venous sinus stenosis, exacerbated by hypertensive emergency
D
All of the above
Optic nerve edema can result from several causes. In this patient, hypertensive papillitis, neuroretinitis, and papilledema from intracranial hypertension were all in the differential diagnosis. Therefore, all of the above testing (choice D) was warranted to ensure a timely and accurate diagnosis. In the setting of hypertensive emergency, the patient was admitted for control of blood pressure and treatment of systemic sequelae. Hypertensive papillitis can present with macular exudation and optic nerve head edema.1 However, concluding that systemic hypertension was the sole underlying cause of the optic nerve head swelling would be inadequate management (choice A). No further update is available for suspected renal artery stenosis as an underlying cause for his malignant hypertension, as the patient is lost to follow-up.Although macular exudation can occur with severe papillitis or papilledema, given the patient’s demographics, cats, and fundoscopic appearance, blood tests were warranted to rule out neuroretinitis (choice B).2 Results of fluorescein angiography demonstrated angiographic leakage of the optic nerve head capillaries with absence of macular leakage characteristic of neuroretinitis (Figure 1). However, results of testing were negative for tuberculosis, syphilis, Bartonella quintana, Bartonella henselae, Lyme disease, sarcoidosis, and toxoplasmosis.The position-dependent headaches and constricted fields were concerning for intracranial hypertension, warranting imaging of the brain and orbits (choice C). Results of magnetic resonance imaging showed no intracranial pathologic findings; results of magnetic resonance venography revealed stenosis at the junction of the right transverse and sigmoid sinuses (Figure 2). Results of lumbar puncture demonstrated an opening pressure of 50 cm H2O. Acetazolamide sodium, 1 g twice daily, was started in the interim prior to urgent cerebral angiography and stenting of the venous sinus stenosis.Magnetic resonance venogram revealing stenosis at the junction of the right transverse and sigmoid sinuses (arrowhead).Given the severity of the optic nerve head edema, we postulated that the patient’s intracranial hypertension may have been exacerbated by the systemic hypertension. Elevated intracranial pressure with papilledema and concomitant hypertensive emergency is a rare clinical scenario, described only once previously to our knowledge,3 and the nerve edema from either is not readily distinguishable from the other.4 An incomplete workup can easily result in treatment of only 1 of the conditions, leading to continued visual deterioration and possible permanent damage from delay in appropriate treatment, showing again that choice D is necessary. A series studying risk factors for severe vision loss in patients with idiopathic intracranial hypertension showed that, while most patients had good long-term visual outcomes, 62% of patients with hypertension became significantly visually impaired, suggesting that systemic hypertension is a significant risk factor.5 Systemic hypertension may be underdiagnosed in patients with elevated intracranial pressure, compromising their visual potential. It is vital to obtain a complete workup (choice D) in patients with optic nerve head edema to ensure timely and accurate diagnosis.Three and a half months after the patient underwent venous sinus stenting, his vision was restored, with formal perimetry showing full visual fields. On fundus examination, the papilledema had decreased significantly bilaterally to Frisen grade 2 with resolved flame hemorrhages, no nerve fiber layer infarcts, absence of dot-and-blot hemorrhages, and decreased exudation into the macula.
Ophthalmology
A previously healthy African American man in his 20s presented to the emergency department after referral by an optometrist for “bleeding in the back of the eye.” The patient noted that 2 months prior he began experiencing throbbing headaches in the back of his head that were more painful and associated with lightheadedness when laying down. Two weeks prior to presentation, he started noticing black spots in his peripheral vision and progressive blurring of vision in both eyes. The patient denied recent travel, cough, gastrointestinal or genitourinary tract symptoms, ulcers, aching joints, tinnitus, or transient visual obscurations. He reported owning cats.On presentation, the patient’s blood pressure was 210/150 mm Hg, and he had a history of acute kidney failure and non–ST-elevation myocardial infarction. On ophthalmic examination, his uncorrected near visual acuity was 20/25 OD and 20/30 OS. Extraocular motility, Ishihara color plates, intraocular pressure, pupils, and results of slitlamp examination were within normal limits. Ophthalmoscopy showed Frisen grade 5 optic nerve head swelling in both eyes with extensive flame hemorrhages and nerve fiber layer infarcts surrounding both nerves and retinal exudates extending from the nerves temporally into both fovea’s (Figure 1). The vessels were of normal course and caliber. There were scattered peripheral dot-and-blot hemorrhages. Results of Humphrey visual field testing were reliable and showed bilateral concentric constriction and enlarged blind spots.Grade 5 optic nerve head edema. A, Fundus photograph of the right eye shows Frisen grade 5 optic disc edema, flame hemorrhages, and peripapillary exudation into the macula. B, Fluorescein angiogram of the right eye in the early venous phase shows blockage from flame hemorrhages, peripapillary microaneurysms, leakage from optic disc capillaries, and absence of vascular leakage at arcades.Control the systemic blood pressure and workup underlying possible cause of the hypertensionPerform magnetic resonance imaging and magnetic resonance venogram of the brain and orbits, then perform lumbar puncture
what would you do next?
What would you do next?
Perform blood tests for neuroretinitis
Control the systemic blood pressure and workup underlying possible cause of the hypertension
All of the above
Perform magnetic resonance imaging and magnetic resonance venogram of the brain and orbits, then perform lumbar puncture
c
1
0
1
1
male
0
0
25
21-30
African American
567
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2717998
A white woman in her 70s with advanced Alzheimer disease was referred to the hematology clinic for evaluation of a high hemoglobin level (169 g/L; normal range, 120-160 g/L) and red blood cell count (5.67 × 1012/L; normal range, 3.8-4.8 × 1012/L) as well as a generalized itch that was worse after a bath. On examination, she had a florid, erythematous macular eruption over the trunk and limbs (Figure, A) but no hepatosplenomegaly or lymphadenopathy. In addition to the high hemoglobin level, mild lymphocytosis (absolute lymphocyte count, 6.2 × 109/L, range 1.5-4.0 × 109/L) was noted with the lymphocyte morphology, suggesting reactive changes. Skin biopsy specimen (Figure, B) showed a normal epidermis with a pericapillary infiltrate of small lymphocytes restricted to the dermis and no leukocytoclastic vasculitis, fungal organisms, or dermal mucin. The absence of cellular atypia and epidermal involvement suggested a diagnosis of lupus, or gyrate or annular erythema, and the need for clinicopathological correlation.A, Photograph shows erythroderma. B, Skin biopsy specimen shows a pericapillary infiltrate of small lymphoid cells restricted to the dermis (hematoxylin-eosin, original magnification ×200). C, Blood smear shows a typical lymphocyte (May-Grünwald Giemsa stain, original magnification ×400).Treatment began with a topical emollient, steroid creams, and oral antihistamines. Skin biopsy was repeated 2 months after the original procedure owing to a suboptimal clinical response, but the histological appearances were unchanged. The hemoglobin level remained high, and the lymphocyte count had increased to 9.0 × 109/L (Figure, C). What Is Your Diagnosis?
T-cell prolymphocytic leukemia
Advanced cutaneous T-cell lymphoma/Sézary syndrome
Polycythemia rubra vera
Cutaneous lupus erythematosus
A. T-cell prolymphocytic leukemia
A
T-cell prolymphocytic leukemia
The blood smear test showed an excess of medium-sized lymphocytes, each with a round to oval nucleus, basophilic cytoplasm, and prominent nucleolus (Figure, C), which typify prolymphocytes. Immunophenotyping identified the expression of CD4 on lymphocytes along with the pan–T-cell antigens CD2, CD3, CD5, and CD7, but no CD8, B-cell, or natural killer cell markers, supporting a diagnosis of T-cell prolymphocytic leukemia (T-PLL).T-cell prolymphocytic leukemia, a rare lymphoid malignancy affecting older people (median age, 61 years) can be misdiagnosed as an alternative mature T-cell neoplasm or even a benign disorder, particularly in patients with an atypical presentation.1 Common features of T-PLL include a high white blood cell count, bone marrow failure, and splenomegaly or lymphadenopathy, but the presence of pseudopolycythemia, mild lymphocytosis, erythroderma, and pruritus made advanced cutaneous T-cell lymphoma (CTCL)/Sézary syndrome a diagnostic consideration.1-3 However, the malignant cells characteristic of CTCL have a hyperconvulated, cerebriform nucleus, which distinguishes these cells from prolymphocytes. Moreover, the absence of epidermotropism and cellular atypia in the skin biopsy results are characteristic of T-PLL. The preservation of CD7 expression on circulating and dermal prolymphocytes further supports a diagnosis of T-PLL over CTCL or adult T-cell lymphocytic leukemia/lymphoma,2,3 an alternative possibility. The patient’s race, low expression intensity of CD25 on neoplastic cells, and the absence of eosinophilia also argue against adult T-cell lymphocytic leukemia/lymphoma.The important learning point here is that with no unique antigen expression profile (except in a quarter of cases that coexpress CD4 and CD8), the morphological recognition of circulating prolymphocytes is key to diagnosing T-PLL.1 Thus, a failure to integrate the skin biopsy findings with blood cytomorphology could have easily become a diagnostic pitfall, particularly because the cytogenetic analysis was normal and noncontributory to the diagnosis. Fluorescence in situ hybridization probes failed to detect inversion in chromosome 14 (q11.2q32.1), the most frequent chromosomal abnormality in T-PLL that is associated with TCL1 overexpression.1-4 Nevertheless, TCRB and TCRG gene rearrangements confirmed the T-cell population to be clonal. Additional learning points relate to the original reason for referral to hematology: the patient was suspected to have polycythemia rubra vera (PRV) owing to a high hemoglobin level and itch, but patients with PRV generally do not have a rash.5 It is likely that fluid loss due to erythroderma caused pseudopolycythemia in the patient; indeed, the Janus kinase (JAK) 2 analysis was negative for mutations frequently found in PRV.6,7 Cutaneous lupus erythematosus was unlikely because parts of the body exposed to the sun were spared, and typical histological features (lymphoid involvement of the dermoepidermal junction, epithelial layer degeneration, or dermal mucin deposits) were absent,8 as were serum autoantibodies commonly associated with connective tissue disorders.Most patients with T-PLL require antileukemic therapy at presentation. Conventional chemotherapy and steroids have limited efficacy, but the anti-CD52 monoclonal antibody alemtuzumab can achieve responses rates of 70% to 90% when administered intravenously.1,2 The disease response to alemtuzumab is of prognostic significance and supersedes the use of pretreatment variables, including total white blood cell count, lymphocyte doubling time, and TCL1 expression intensity, as an important determinant of survival.1 However, responses are frequently not durable, and to optimize outcomes in eligible patients, consolidative treatment with an autologous or allogeneic stem cell transplant requires consideration.1,2 Recent reports have indicated the effectiveness of the B–cell lymphoma 2 antagonist venetoclax9 and JAK3 inhibitors10 (following identification of JAK/signal transducer and activator of transcription [STAT] pathway mutations) in T-PLL, but the optimal positioning of newer drugs in the treatment algorithms requires investigation.2The patient’s advanced cognitive impairment precluded a trial of alemtuzumab therapy, and management focused exclusively on symptom palliation. Eight months after the initial clinic visit, she presented with right-sided periorbital swelling owing to abnormal growth of soft tissue in the temporal and zygomatic region that was compressing the eyeball and worsening lymphocytosis (absolute lymphocyte count, 13 × 109/L). Disease progression was rapid, and she died.
Oncology
A white woman in her 70s with advanced Alzheimer disease was referred to the hematology clinic for evaluation of a high hemoglobin level (169 g/L; normal range, 120-160 g/L) and red blood cell count (5.67 × 1012/L; normal range, 3.8-4.8 × 1012/L) as well as a generalized itch that was worse after a bath. On examination, she had a florid, erythematous macular eruption over the trunk and limbs (Figure, A) but no hepatosplenomegaly or lymphadenopathy. In addition to the high hemoglobin level, mild lymphocytosis (absolute lymphocyte count, 6.2 × 109/L, range 1.5-4.0 × 109/L) was noted with the lymphocyte morphology, suggesting reactive changes. Skin biopsy specimen (Figure, B) showed a normal epidermis with a pericapillary infiltrate of small lymphocytes restricted to the dermis and no leukocytoclastic vasculitis, fungal organisms, or dermal mucin. The absence of cellular atypia and epidermal involvement suggested a diagnosis of lupus, or gyrate or annular erythema, and the need for clinicopathological correlation.A, Photograph shows erythroderma. B, Skin biopsy specimen shows a pericapillary infiltrate of small lymphoid cells restricted to the dermis (hematoxylin-eosin, original magnification ×200). C, Blood smear shows a typical lymphocyte (May-Grünwald Giemsa stain, original magnification ×400).Treatment began with a topical emollient, steroid creams, and oral antihistamines. Skin biopsy was repeated 2 months after the original procedure owing to a suboptimal clinical response, but the histological appearances were unchanged. The hemoglobin level remained high, and the lymphocyte count had increased to 9.0 × 109/L (Figure, C).
what is your diagnosis?
What is your diagnosis?
Cutaneous lupus erythematosus
Advanced cutaneous T-cell lymphoma/Sézary syndrome
T-cell prolymphocytic leukemia
Polycythemia rubra vera
c
0
1
1
1
female
0
0
75
71-80
White
568
original
https://jamanetwork.com/journals/jamacardiology/fullarticle/2718495
A man in his early 40s was admitted to the hospital because of acute chest pain. Laboratory test results were notable for high serum concentrations of IgG4 (5650 mg/dL; to convert to grams per liter, multiply by 0.01) and C-reactive protein (78.1 mg/L; to convert to nanomoles per liter, multiply by 9.524). Coronary computed tomographic angiography was performed (Figure, A and B). It revealed a large aneurysm (90 mm × 48 mm) with a thrombosis in the left anterior descending artery (LAD) and a perivascular soft-tissue mass involving the patent proximal right coronary artery (RCA) and left circumflex artery, which showed homogeneous moderate enhancement on contrast-enhanced images. Coronary computed tomographic angiography with cinematic rendering (Figure, C) showed multiple mass lesions around the RCA and LAD. Invasive coronary angiography results were unable to visualize the LAD but did identify a hypervascular mass in the proximal RCA.A, Contrast-enhanced axial computed tomographic image revealing perivascular soft-tissue masses involving the left circumflex and right coronary arteries (arrowheads). B, Maximum intensity projection reformatted image of coronary computed tomographic angiography revealing perivascular soft-tissue masses involving the left circumflex and right coronary arteries (arrowheads) and a large aneurysm with a thrombus involving the left anterior descending artery (asterisk). C, A 3-dimensional rendering demonstrated mass lesions involving the left anterior descending and right coronary arteries (arrowheads). What Would You Do Next?
High-dose corticosteroid therapy
High-dose immunosuppressant therapy
Percutaneous coronary intervention
Coronary artery bypass graft surgery
IgG4-associated coronary artery aneurysms
D
Coronary artery bypass graft surgery
A unique immune-mediated fibroinflammatory disorder, IgG4-related disease is characterized by elevated serum IgG4 levels and tumefied lesions that may concurrently or consecutively affect a wide variety of organs.1-3 It is most common in middle-aged or elderly men. Diagnosis of IgG4-related disease is based on histological findings that show IgG4-positive plasma cell and lymphocytic infiltration, fibrosis with storiform features, and obliterative phlebitis. Elevated serum IgG4 levels also indicate IgG4-related disease.Cases of IgG4-associated coronary artery involvement have rarely been reported. They can result in ischemic heart disease, as in this case, or sudden cardiac death. Because IgG4-associated coronary inflammation occurs in the adventitia, it can result in inflammatory pseudotumor, perivascular fibrosclerotic thickening, luminal stenosis, and aneurysmal dilatation. The typical mistletoe sign in coronary computed tomographic angiography is indicative of the diagnosis of IgG4-associated coronary artery disease,4 as this case shows. Positron emission tomography and computed tomography are also useful in evaluation and follow-up of IgG4-related disease with coronary artery involvement; these can show hypermetabolic coronary artery masses and identify alternative biopsy sites.5The optimal therapeutic approach for IgG4-related disease with coronary artery involvement is unclear. Steroid therapy is regarded as a first-line treatment that can prevent the further development of active inflammatory disease by suppressing lymphocyte activation to reduce inflammation.1-3 However, already existing lesions usually do not recover completely.1-3 Thus, surgical intervention is needed in some patients.The patient underwent coronary artery bypass graft surgery. Intraoperative findings included multiple coronary pseudoaneurysms with thrombosis. Pathological examinations of the mass resected from the LAD confirmed the diagnosis of IgG4-related disease. The patient was discharged with full symptomatic relief. A second coronary computed tomographic angiography 1 year later showed that 3 coronary bypass grafts were patent and the proximal native RCA was completely obstructed. No major adverse cardiac events were reported during the 1-year follow-up examination.
Cardiology
A man in his early 40s was admitted to the hospital because of acute chest pain. Laboratory test results were notable for high serum concentrations of IgG4 (5650 mg/dL; to convert to grams per liter, multiply by 0.01) and C-reactive protein (78.1 mg/L; to convert to nanomoles per liter, multiply by 9.524). Coronary computed tomographic angiography was performed (Figure, A and B). It revealed a large aneurysm (90 mm × 48 mm) with a thrombosis in the left anterior descending artery (LAD) and a perivascular soft-tissue mass involving the patent proximal right coronary artery (RCA) and left circumflex artery, which showed homogeneous moderate enhancement on contrast-enhanced images. Coronary computed tomographic angiography with cinematic rendering (Figure, C) showed multiple mass lesions around the RCA and LAD. Invasive coronary angiography results were unable to visualize the LAD but did identify a hypervascular mass in the proximal RCA.A, Contrast-enhanced axial computed tomographic image revealing perivascular soft-tissue masses involving the left circumflex and right coronary arteries (arrowheads). B, Maximum intensity projection reformatted image of coronary computed tomographic angiography revealing perivascular soft-tissue masses involving the left circumflex and right coronary arteries (arrowheads) and a large aneurysm with a thrombus involving the left anterior descending artery (asterisk). C, A 3-dimensional rendering demonstrated mass lesions involving the left anterior descending and right coronary arteries (arrowheads).
what would you do next?
What would you do next?
High-dose corticosteroid therapy
High-dose immunosuppressant therapy
Percutaneous coronary intervention
Coronary artery bypass graft surgery
d
1
1
1
1
male
0
0
42
41-50
null
569
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2724226
A 53-year-old African American man with a history of bipolar disorder presented to the emergency department with an enlarging right-sided tongue mass that had caused mild discomfort and halitosis over the course of 1 week. He denied recent injury, fever, chills, odynophagia, or difficulty breathing. Laboratory examination was significant for leukocytosis (white blood cell count, 15 700/μL) and hypercalcemia (total calcium level, 13.1 mg/dL) (to convert white blood cells to ×109 per liter, multiply by 0.001; to convert calcium to millimoles per liter, multiply by 0.25). Physical examination was notable for a 4- to 5-cm pedunculated, ulcerative mass on the dorsal oral aspect of the tongue near midline, anterior to the foramen cecum, without palpable cervical lymphadenopathy. Contrast-enhanced computed tomography (CT) demonstrated a rim-enhancing, centrally hypodense lesion in the right dorsal tongue abutting the lingual septum with a somewhat ill-defined border (Figure 1); there was no associated osseous erosion of the mandible or hard palate, no tongue prolapse to suggest hypoglossal nerve palsy, no involvement of the soft palate, and no cervical lymphadenopathy by radiological size criteria. An excisional biopsy of the anterior aspect of the mass was performed at bedside, and the patient was discharged home with instructions to follow up in the otolaryngology clinic.A, Sagittal contrast-enhanced computed tomography (CT) demonstrates an exophytic mass on the dorsal aspect of the tongue. B, Axial contrast-enhanced CT demonstrates a peripherally enhancing mass of the oral tongue, to the right of midline. What Is Your Diagnosis?
Squamous cell carcinoma
Oral eosinophilic ulcer
Abscess
Pleomorphic rhabdomyosarcoma
D. Pleomorphic rhabdomyosarcoma
D
Pleomorphic rhabdomyosarcoma
Fluorodeoxyglucose F18–labeled positron-emission tomography demonstrated the lesion to be intensely avid (standardized uptake value, 16.4) (Figure 2).A, Axial attenuation-corrected fluorodeoxyglucose F18 (FDG)-labeled positron-emission tomography single-photon emission computed tomography image after the administration of 12.1 mCi of FDG demonstrates an intensely avid oral tongue mass (standardized uptake value, 16.4); heterogeneity of activity suggests areas of necrosis. B, Fused image illustrates anatomical overlap between the region of uptake and rim-enhancing lesion seen on computed tomography.Rhabdomyosarcoma (RMS) is an aggressive cancer of mesenchymal origin, most common in the pediatric population, with 35% occurring in the head and neck region1; the tongue is an uncommon site of involvement. World Health Organization classification of RMS is based on histologic findings and includes 4 groups: embryonal, alveolar, pleomorphic, and spindle cell/sclerosing.2 Pleomorphic RMS is rare and is seen almost exclusively in adults.3 Wide surgical resection is the mainstay of treatment for localized pleomorphic RMS, followed by postoperative radiotherapy because this RMS subtype is sometimes regarded as chemoresistant.2,4 In patients with metastatic disease, palliative radiation therapy and chemotherapy may be attempted.2 Pleomorphic RMS heralds poor prognosis by virtue of its histologic characteristics as well as the affected age group.4 Overall survival rates in the adult population with RMS of all sites and all histologic traits range from 40% to 54% at 5 years.5In the present case, a partial glossectomy was performed, and the mass was excised with 1.5-cm circumferential margins, followed by bilateral selective neck dissection of cervical chain nodal levels I through IV. The tongue was reconstructed using local tissue rearrangement rather than flap reconstruction, given the size of the defect and the intact tongue base. The definitive diagnosis of pleomorphic RMS (negative for FOX01 rearrangement) was rendered on pathologic examination, with a pathologic stage classification of pT3N0. Although the hypercalcemia was initially attributed to the RMS, the patient was subsequently found to have an atypical parathyroid adenoma, which was also excised.The imaging-based differential diagnosis of pleomorphic RMS of the tongue includes squamous cell carcinoma (SCC), traumatic ulcerative granuloma with stromal eosinophilia, and infectious ulcer, in addition to infected dermoid (if the lesion is midline) or venous malformation (if there are phleboliths). Squamous cell carcinoma represents more than 90% of oral cavity cancers and arises from squamous epithelial cells of the oral tongue. When centrally necrotic, SCC presents as a peripherally enhancing lesion with central hypoattenuation. However, given the thickness of the lesion in the present case, one would expect that there would be metastatic cervical lymphadenopathy.6 Moreover, the patient had none of the risk factors commonly associated with the development of SCC, such as tobacco or alcohol use; infection with human immunodeficiency virus, Epstein-Barr virus, or human papilloma virus; familial history; or occupational exposure.7Traumatic ulcerative granuloma with stromal eosinophilia is a differential consideration, given the development of a solitary, ulcerated lesion on the dorsal tongue surface over a short period of time. The pathophysiology of this benign, self-limiting condition is poorly understood, but it is thought to be related to aberrant wound-healing and is of clinical importance because it can mimic malignant entities such as SCC or mucoepidermoid carcinoma.8 This entity is usually seen in individuals with a history of trauma or iatrogenic tongue injury and demonstrates rapid healing after incisional biopsy, presumably due to induction of normal healing pathways,9 features not present in the present case.Infectious ulcers of the oral tongue may be due to cytomegalovirus, tuberculosis, or fungal agents, and are suggested by details in the clinical history, such as immune status, travel history, or the presence of multisystem disease.10 The pedunculated and masslike appearance of the present lesion make infectious ulcer less likely.
General
A 53-year-old African American man with a history of bipolar disorder presented to the emergency department with an enlarging right-sided tongue mass that had caused mild discomfort and halitosis over the course of 1 week. He denied recent injury, fever, chills, odynophagia, or difficulty breathing. Laboratory examination was significant for leukocytosis (white blood cell count, 15 700/μL) and hypercalcemia (total calcium level, 13.1 mg/dL) (to convert white blood cells to ×109 per liter, multiply by 0.001; to convert calcium to millimoles per liter, multiply by 0.25). Physical examination was notable for a 4- to 5-cm pedunculated, ulcerative mass on the dorsal oral aspect of the tongue near midline, anterior to the foramen cecum, without palpable cervical lymphadenopathy. Contrast-enhanced computed tomography (CT) demonstrated a rim-enhancing, centrally hypodense lesion in the right dorsal tongue abutting the lingual septum with a somewhat ill-defined border (Figure 1); there was no associated osseous erosion of the mandible or hard palate, no tongue prolapse to suggest hypoglossal nerve palsy, no involvement of the soft palate, and no cervical lymphadenopathy by radiological size criteria. An excisional biopsy of the anterior aspect of the mass was performed at bedside, and the patient was discharged home with instructions to follow up in the otolaryngology clinic.A, Sagittal contrast-enhanced computed tomography (CT) demonstrates an exophytic mass on the dorsal aspect of the tongue. B, Axial contrast-enhanced CT demonstrates a peripherally enhancing mass of the oral tongue, to the right of midline.
what is your diagnosis?
What is your diagnosis?
Pleomorphic rhabdomyosarcoma
Squamous cell carcinoma
Oral eosinophilic ulcer
Abscess
a
1
1
1
1
male
0
0
53
51-60
African American
570
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2725435
A 17-year-old girl with a history of chronic sinusitis presented with left-sided epiphora, purulent drainage, and pain and right-sided jaw pain. She had previously been treated with antibiotics and mupirocin nasal irrigation, with temporary improvement. The patient and her identical twin sister had required multiple surgical procedures in the past for osseous lesions in the upper and lower jaws, including an aggressive resection via bilateral Caldwell-Luc approach 2 years previously; a computed tomographic scan from that initial presentation is shown in the Figure, A. At the current presentation, examination using flexible nasal endoscopy revealed bulging of the left lateral nasal wall; imaging revealed a 14-mm, dense, expansile, sclerotic mass with moderate calcifications obstructing the left nasolacrimal duct; a similar 2.3 × 1.8–cm lesion of the right mandibular body; and evidence of previous sinus surgery (Figure, B). Because of these findings, left-sided endoscopic dacryocystorhinostomy and sinus surgery was performed. Histologic findings are shown in the Figure, C and D.A, Contrast-enhanced computed tomographic image at initial presentation. B, Non–contrast-enhanced computed tomographic image at 2 years after bilateral Caldwell-Luc resection showing mass in left nasolacrimal duct. C, Photomicrograph depicting a proliferation of highly cellular, fibrous connective tissue (arrow) and cementum-like hard tissue (arrowhead) (hematoxylin-eosin, original magnification ×100). D, Photomicrograph showing whorls of spindle cells (arrow) with relatively acellular calcified masses of cemento-osseous material (arrowhead) (hematoxylin-eosin, original magnification ×200). What Is Your Diagnosis?
Albers-Schönberg disease
Gigantiform cementoma
Fibrous dysplasia
Polyostotic ossifying fibroma
B. Gigantiform cementoma
B
Gigantiform cementoma
Microscopic study showed a lesion formed by whorling, swirling spindle cells with islands of basophilic cementum that appeared concentrically laminated. There were also fragments of bone with marrow spaces containing fibrous tissue and toothlike fragments. On immunohistochemical staining, lesional cells were positive for vimentin and negative for epithelial membrane antigen, cytokeratin, smooth-muscle actin, CD31, CD34, S-100, and β-catenin. The pathologic studies confirmed the diagnosis of familial gigantiform cementoma.Gigantiform cementoma is a rare form of a fibro-cemento osseous lesion that is usually inherited in an autosomal dominant fashion with variable penetrance; when this occurs in families, it is known as familial gigantiform cementoma (FGC). In the present case, both the patient and her identical twin were found to have a de novo, balanced 1;13 translocation mutation presumably causing osseous dysplasia affecting the maxilla and mandible. Although there was no family history of GC for this patient and her twin, there have been reports of FGC in other families in the literature, with discussion of the varying characteristics of this disease.1-4Familial gigantiform cementoma lesions have distinct radiologic and histologic features. Growth typically appears in 3 stages: initial onset, rapid expansion, and suppression of growth.1,3 During initial onset, FGC lesions resemble other osseous dysplasias on radiography, with lesions demonstrating a mixture of radiolucent and radiopaque appearance. However, as the lesions mature, they become more opacified and can disrupt dentition though local invasion. A higher incidence of pathologic fracture has been reported in patients with FCG owing to disruption of dentition, cortical thinning, and subsequent compromise of mandibular integrity.5,6 On maturation, the lesions stabilize and can appear as central radiopaque lesions with a radiolucent rim.7 On histologic examination, these lesions show whorls of spindle cells with basophilic acellular lamellated masses of calcified tissue resembling cementum, which were seen in our patient.8Treatment for FGC is individualized and ranges from conservative management to radical resection of lesions with possible use of bone flap for reconstruction.1,7-10 Although most patients require multiple debulking procedures, simple recontouring without complete excision is not considered to be sufficient treatment owing to the risk of recurrence. However, given the functional cosmetic, speech, and swallowing challenges of lesions involving the jaws, the risk of recurrence must be weighed against the risks of surgical resection.
General
A 17-year-old girl with a history of chronic sinusitis presented with left-sided epiphora, purulent drainage, and pain and right-sided jaw pain. She had previously been treated with antibiotics and mupirocin nasal irrigation, with temporary improvement. The patient and her identical twin sister had required multiple surgical procedures in the past for osseous lesions in the upper and lower jaws, including an aggressive resection via bilateral Caldwell-Luc approach 2 years previously; a computed tomographic scan from that initial presentation is shown in the Figure, A. At the current presentation, examination using flexible nasal endoscopy revealed bulging of the left lateral nasal wall; imaging revealed a 14-mm, dense, expansile, sclerotic mass with moderate calcifications obstructing the left nasolacrimal duct; a similar 2.3 × 1.8–cm lesion of the right mandibular body; and evidence of previous sinus surgery (Figure, B). Because of these findings, left-sided endoscopic dacryocystorhinostomy and sinus surgery was performed. Histologic findings are shown in the Figure, C and D.A, Contrast-enhanced computed tomographic image at initial presentation. B, Non–contrast-enhanced computed tomographic image at 2 years after bilateral Caldwell-Luc resection showing mass in left nasolacrimal duct. C, Photomicrograph depicting a proliferation of highly cellular, fibrous connective tissue (arrow) and cementum-like hard tissue (arrowhead) (hematoxylin-eosin, original magnification ×100). D, Photomicrograph showing whorls of spindle cells (arrow) with relatively acellular calcified masses of cemento-osseous material (arrowhead) (hematoxylin-eosin, original magnification ×200).
what is your diagnosis?
What is your diagnosis?
Fibrous dysplasia
Polyostotic ossifying fibroma
Gigantiform cementoma
Albers-Schönberg disease
c
0
1
1
1
female
0
0
17
11-20
null
571
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2726012
A woman in her 60s reported right upper lip submucosal masses that had been present for several weeks. She denied associated pain, bleeding, or recent trauma to the lip. She also denied any other constitutional symptoms. Her social history was significant for a 45–pack-year smoking history and social drinking. On physical examination, the patient had right upper lip submucosal masses without any tenderness or mucosal abnormality. The remainder of the otolaryngic examination was unremarkable. A computed tomographic scan of the neck with contrast demonstrated multiple hyperenhancing nodules; the largest was 3 × 7 × 6 mm, with no local bony erosion or cervical lymphadenopathy (Figure 1).Computed tomographic (CT) scans of hyperenhancing nodules of the upper lip (red arrows). What Is Your Diagnosis?
Multifocal basal cell adenoma
Basal cell adenocarcinoma
Multifocal canalicular adenoma
Adenoid cystic carcinoma
C. Multifocal canalicular adenoma
C
Multifocal canalicular adenoma
Canalicular adenoma is an uncommon salivary gland tumor arising almost exclusively from the minor salivary glands of the oral cavity. It is usually asymptomatic and slow growing, and accounts for less than 1% to 3% of all salivary neoplasms.1,2 Canalicular adenoma is most commonly diagnosed as an upper lip nodule found in women in their seventh decade of life, as exemplified by our patient.1 Other locations in which canalicular adenoma can be found include the hard palate and buccal mucosa.1 It is the third most common benign salivary gland tumor, with a recurrence rate of approximately 5%.1,2 Canalicular adenoma was previously thought to be a variant of basal cell adenoma; however, its unique clinical and pathologic features warrant defining it as its own entity.3 For example, while canalicular adenoma is seen almost exclusively in the oral cavity, basal cell adenoma has a predilection for the major salivary glands, especially the parotid gland.2-4 This locational difference is the first hint in differentiating between these 2 entities.Histologically, canalicular adenoma tends to be a well-circumscribed solitary nodule consisting of anastomosing cords formed by columnar cells arranged in a loose connective tissue stroma.1,2 As seen in this patient, however, approximately 15% of canalicular adenomas are multifocal, a feature that must not be misinterpreted as invasive growth, a defining feature of salivary gland carcinomas such as adenoid cystic carcinoma or basal cell adenocarcinoma. By immunohistochemistry, canalicular adenoma is consistently positive for S100 and SOX10, variably positive for p63, and negative for p40.1,2 Although basal cell adenoma is also composed of basaloid cells growing as nests and cords, it exhibits a more cellular stroma and has 2 cell populations: basaloid myoepithelial cells and ductal cells; the myoepithelial cells can be highlighted by immunohistochemistry for smooth muscle actin, calponin, glial fibrillary acidic protein, and others.5 More important, canalicular adenoma is composed of 1 population of ductal cells, and does not stain for myoepithelial elements that are found in basal cell adenoma, basal cell adenocarcinoma, and adenoid cystic carcinoma.4 Surgical resection is the treatment of choice for canalicular adenoma.1 In contrast, malignant neoplasms such as adenoid cystic carcinoma may need further management with radiotherapy and frequent monitoring, making differentiation imperative.6Clinically, canalicular adenoma may mimic a mucocele, which is a collection of mucus in the soft tissue after rupture of a salivary gland duct.4,7 Both can present as well-demarcated blue-tinted masses or even match the color of normal oral mucosa.7 Unlike canalicular adenoma, mucoceles are often associated with a history of trauma, which our patient denied.7 In addition, mucoceles are often diagnosed earlier in life, around the second decade, and are found equally in males and females.7 Mucoceles are found predominately in the lower labial mucosa with virtually no presentation in the upper labial mucosa, making this diagnosis unlikely for our patient.7 Histologically, the differentiation is simple, with mucocele histologic characteristic showing mucin in a subepithelial vesicle.7In summary, histologic characteristics and immunohistochemistry are crucial for distinguishing canalicular adenoma from other diagnoses. Canalicular adenoma is occasionally multifocal, a feature that must be distinguished from the truly invasive growth that characterizes similar-appearing malignant neoplasms. Although surgical resection is the mainstay of treatment for upper lip masses, differentiation is necessary in cases in which the diagnosis of adenoid cystic carcinoma necessitates concurrent use of radiotherapy. Other factors such as patient sex, decade of life, and location of mass are also helpful in the initial formation of a differential diagnosis.After surgical excision, pathologic examination revealed a nodular proliferation of basaloid cells (Figure 2A) made up of nodules ranging from less than 1 to 7 mm. In the background minor salivary tissue, there were several additional microscopic, incipient nodules with an identical appearance (Figure 2B). The nodules were composed of nests and interconnecting cords of uniform cells with bland, elongated nuclei with vesicular chromatin and delicate nucleoli. The tumor cells were set in a loose myxoid stroma (Figure 2C). Although the tumor was multinodular, no convincing invasive growth was seen. By immunohistochemistry the tumor was diffusely positive for S100 and SOX10, focally positive for p63, and negative for p40.Staining of excised mass reveals nodular proliferation of basaloid cells (hematoxylin-eosin, original magnification x40) (A); microscopic, incipient nodules (black arrowheads) in the background minor salivary gland tissue (hematoxylin-eosin, original magnification x100) (B); and tumor cells in loose myxoid stroma (hematoxylin-eosin, original magnification x200) (C).
General
A woman in her 60s reported right upper lip submucosal masses that had been present for several weeks. She denied associated pain, bleeding, or recent trauma to the lip. She also denied any other constitutional symptoms. Her social history was significant for a 45–pack-year smoking history and social drinking. On physical examination, the patient had right upper lip submucosal masses without any tenderness or mucosal abnormality. The remainder of the otolaryngic examination was unremarkable. A computed tomographic scan of the neck with contrast demonstrated multiple hyperenhancing nodules; the largest was 3 × 7 × 6 mm, with no local bony erosion or cervical lymphadenopathy (Figure 1).Computed tomographic (CT) scans of hyperenhancing nodules of the upper lip (red arrows).
what is your diagnosis?
What is your diagnosis?
Multifocal basal cell adenoma
Multifocal canalicular adenoma
Adenoid cystic carcinoma
Basal cell adenocarcinoma
b
1
0
0
1
female
0
0
65
61-70
null
572
original
https://jamanetwork.com/journals/jama/fullarticle/2727553
A 22-year-old man presented with an 8-year history of painful and slightly pruritic skin lesions on his right flank. The number of lesions and degree of pain had alternately increased and decreased over the years. Occasionally the lesions would rupture and leak clear fluid. One year prior, he was hospitalized for a “skin infection” on his right flank and treated with intravenous antibiotics. The patient, a construction demolition worker, was otherwise healthy and took no medications. He had 1 lifetime female sexual partner with whom he used barrier contraception. Physical examination revealed an asymmetric cluster of lesions on the right flank measuring 14 × 12 cm at the widest margins (Figure 1). The cluster contained verrucous papules and plaques in the center and vesicles filled with clear and milky-white fluid in the periphery. Crust was partially covering some of the lesions. The surrounding skin was erythematous without warmth or induration. The remainder of the physical examination was normal except for stretch marks on the lateral abdomen.Left, Vesicular and verrucous skin lesions on patient’s right flank. Right, Close-up view of lesions. What Would You Do Next?
Biopsy the lesions
Prescribe oral valacyclovir
Prescribe topical cantharadin
Recommend a gluten-free diet
Lymphangioma circumscriptum
A
Biopsy the lesions
The key to the correct diagnosis in this case is a long-standing history of painful grouped vesicular and verrucous lesions in a single anatomical location, consistent with lymphangioma circumscriptum. The infection experienced by this patient was likely a secondary cellulitis, which is common in lymphangioma circumscriptum and usually caused by Staphylococcus aureus. Definitive diagnosis of lymphangioma circumscriptum requires biopsy and histopathological analysis. The disorder often appears similar to herpes simplex, herpes simplex vegetans, and herpes zoster, which can all initially be treated with valacyclovir. Of these viral lesions, only herpes simplex vegetans (a rare form of herpes simplex virus cutaneous infection in immunocompromised patients that is often recalcitrant to first-line treatment) presents with long-standing verrucous lesions. Clinical manifestations include papular eruptions, verrucous lesions, and erosive vegetative plaques, most often in the genitocrural area. Dermatitis herpetiformis, which initially can be managed with a gluten-free diet, presents as a chronic relapsing vesiculopapular rash but typically involves extensor surfaces in a symmetric distribution. Molluscum contagiosum can be treated with topical cantharadin and may have a verrucuous appearance, especially in immunocompromised patients; however, vesicles would not be present.Lymphangioma circumscriptum is a congenital cutaneous lymphatic malformation characterized by translucent vesicles resembling “frog spawn.”1 The vesicles harbor clear or serosanguinous fluid composed of varying degrees of lymph and blood.2,3 Vesicle rupture commonly occurs, and repetitive rupture can induce epidermal hyperplasia and hyperkeratosis, leading to a verrucous appearance.2,3 Lesions with a hemorrhagic component can become darkly pigmented.2,3 Other symptoms include lymphorrhea (leakage of lymphatic fluid), pruritus, and pain.2-4In the United States, congenital lymphatic malformations occur in 1.2 to 2.8 per 1000 live births. Lymphangioma circumscriptum is the most common congenital lymphatic malformation, although its true incidence is unknown.1 Approximately 90% of lymphangioma circumscriptum cases manifest by age 2 years, and the remainder typically appear before age 30 years.1,5 Lymphangioma circumscriptum lesions function as a “closed system,” entirely separate from the normal lymphatic system.5 During lymphatic development, abnormal cisterns with a muscular coating form in the subcutaneous tissue.1,5 These cisterns develop connections to ectopic lymphatic vessels of the papillary (superficial) dermis via vertical channels.2 As the deep cisterns contract, lymph and pressure are transmitted upward to the dermal lymphatics, which dilate to form subepidermal vesicles.1,3The differential diagnosis for lymphangioma circumscriptum includes infectious conditions (herpes simplex, herpes simplex vegetans, herpes zoster, verruca, condyloma acuminatum, and molluscum contagiosum), other benign lesions (dermatitis herpetiformis, hemangioma, angiokeratoma, epidermal nevus, and lymphangiectasia), and malignancies (metastatic carcinoma, angiosarcoma, and melanoma).4,6 Definitive diagnosis of lymphangioma circumscriptum is made histopathologically. The superficial dermis contains dilated lymphatic channels with a flattened endothelial lining (Figure 2). The overlying epidermis is often hyperkeratotic and acanthotic, especially in verrucous lesions.1,5Biopsy sample of patient’s right flank lesions showing increased number of dilated lymphatic vessels containing lymph in the superficial dermis (hematoxylin-eosin, original magnification ×4).Lymphangioma circumscriptum is benign, and treatment is not required in asymptomatic patients. Indications for treatment include lymphorrhea, recurrent infections, cosmetic concerns, bleeding, and pain.3 Surgical excision is the most common and effective treatment because it is the only treatment that can eliminate the deep subcutaneous cisterns.4 However, lymphangioma circumscriptum often has larger dermal and subcutaneous components than are clinically apparent, making complete excision difficult. All subcutaneous tissue superficial to deep fascia must be excised to encompass the deep cisterns, often necessitating extensive excision and reconstruction with flaps or skin grafts. Preoperative magnetic resonance imaging, ultrasound, or lymphangiography is recommended to delineate the extent of the lesions, and surgical excision may not be optimal for patients with extensive lesions.2,4,5 Reported recurrence rates are around 9% after a single excision and 5% after reexcision.7A commonly used alternative to excision is carbon dioxide (CO2) laser, especially in cases involving a large surface area.2 CO2 laser vaporizes tissue down to the mid-dermis, eliminating the surface lymphatics. Unlike surgical excision, the laser does not eliminate the deep cisterns but can impede their activity by sealing off the vertical communicating channels necessary for the upward transmission of pressure and vesicle formation.2,5 Although CO2 laser is less invasive than excision, it is an ablative procedure requiring extensive local anesthesia and can result in significant wounds.8 A systematic review of lymphangioma circumscriptum treated with CO2 laser reported a complete lesional recurrence rate of 10% and partial lesional recurrence rate of 50%.2 Additional treatment methods include pulsed dye laser, electrocoagulation, sclerosing agents, imiquimod, electrocautery, cryosurgery, and radiotherapy.3-5,9,10 Irrespective of the treatment modality, lymphangioma circumscriptum has a high recurrence rate, and patients should be counseled accordingly.The patient’s biopsy confirmed a diagnosis of lymphangioma circumscriptum. He was referred for CO2 laser treatment but declined because of the high recurrence rate and cost of treatment.
General
A 22-year-old man presented with an 8-year history of painful and slightly pruritic skin lesions on his right flank. The number of lesions and degree of pain had alternately increased and decreased over the years. Occasionally the lesions would rupture and leak clear fluid. One year prior, he was hospitalized for a “skin infection” on his right flank and treated with intravenous antibiotics. The patient, a construction demolition worker, was otherwise healthy and took no medications. He had 1 lifetime female sexual partner with whom he used barrier contraception. Physical examination revealed an asymmetric cluster of lesions on the right flank measuring 14 × 12 cm at the widest margins (Figure 1). The cluster contained verrucous papules and plaques in the center and vesicles filled with clear and milky-white fluid in the periphery. Crust was partially covering some of the lesions. The surrounding skin was erythematous without warmth or induration. The remainder of the physical examination was normal except for stretch marks on the lateral abdomen.Left, Vesicular and verrucous skin lesions on patient’s right flank. Right, Close-up view of lesions.
what would you do next?
What would you do next?
Prescribe oral valacyclovir
Recommend a gluten-free diet
Biopsy the lesions
Prescribe topical cantharadin
c
0
1
0
1
female
0
0
22
21-30
White
573
original
https://jamanetwork.com/journals/jama/fullarticle/2725892
A 31-year-old gravida 2, para 1 patient of 36 weeks’ gestation had undergone a laparoscopic Roux-en-Y gastric bypass (RYGB) 4 years ago, resulting in prepregnancy weight loss of 70 kg, or 128% of her excess weight. She awakened with severe lower back pain, abdominal pain, nausea, and vomiting. She came to the emergency department with 10/10 abdominal pain but no fever, chills, vaginal bleeding, or rupture of membranes. Her abdominal examination revealed diffuse tenderness and guarding. She was initially diagnosed as having preterm labor and was given antibiotics, steroids, an epidural catheter for pain management, and intravenous fentanyl as needed. She delivered a healthy baby without complications, but her severe abdominal pain, nausea, and vomiting persisted. Thirty-six hours after her initial presentation, she still had rebound tenderness in the left side of the abdomen. She was afebrile, blood pressure was 104/77 mm Hg, and heart rate was 152/min. White blood cell count was 10.7 ×109/L; anion gap, 13 mEq/L; and lactate dehydrogenase level, 131.7 U/L (2.2 μkat/L). A computed tomography (CT) scan of the abdomen and pelvis with oral and intravenous contrast revealed mesenteric vascular compromise (Figure 1); after the scan, the patient had hematemesis. What Would You Do Next?
Esophagogastrojejunoscopy
Exploratory laparotomy
Highly selective mesenteric angiography
Diagnostic laparoscopy
Internal hernia with gangrenous bowel
B
Exploratory laparotomy
The key to the correct diagnosis in this case is a patient with unstable vital signs, an acute abdomen, hematemesis, and CT findings of internal hernia with mesenteric vascular compromise. Operative findings revealed an internal hernia through the Petersen space, with tight twisting of the bowel resulting in infarction and gangrene in the mid-Roux limb and in the ileum. She required vasopressive drugs to maintain blood pressure while in the operating room. Because of her hemodynamic instability, after resection of the necrotic bowel her intestines were left discontinuous and the abdomen open. She was returned to the operating room 36 hours later and her intestines were reconnected.Internal hernia after gastric bypass remains a challenging complication. Potential spaces created during the operation include the jejunojejunostomy mesenteric defect; the Petersen space, which lies between the Roux mesentery and transverse mesocolon; and the mesocolonic defect when retrocolic Roux placement is performed (Figure 2; Video). How to best prevent internal hernia after gastric bypass is still not known.1-3 An internal hernia should be suspected in any patient who has undergone gastric bypass and presents with severe abdominal pain, a bowel obstruction, or both. When internal hernia is suspected, the evaluation should include a careful history, physical examination, and, frequently, a CT scan. The characteristic appearance of an internal hernia on CT scan is the presence of a mesenteric swirl, small-bowel sequestration (majority of the small bowel confined to 1 quadrant), beak-like appearance of the mesenteric vein, and location of the jejunojejunostomy on the right side of the abdomen instead of the middle.4Computed tomography scan (left) and correlative illustration (right) depicting common radiologic findings indicative of internal hernia following Roux-en-Y gastric bypass.Pregnancy complicates the assessment of internal hernia and can delay intervention. Because symptoms of internal hernia may be mistaken for labor pain, CT scanning to establish the diagnosis is often avoided, even when internal hernia is suspected, because of the risk of exposing the fetus to ionizing radiation.5 The prevalence of internal hernia in pregnancy after RYGB may be as high as 8% and should prompt clinicians to consider internal hernia in women who present with preterm labor and a history of gastric bypass.6 Risk factors for internal hernia include weight loss of 50 kg or more or 90% or greater excess weight loss and third trimester gestation.7,8 Although successful laparoscopic repair is possible, maternal mortality, fetal mortality, or both have been reported in 4% to 6% of pregnancies complicated by internal hernia.5-7 Women of childbearing age considering RYGB should be informed of the possible complications of internal hernia generally but also in the setting of future pregnancy, and its signs and symptoms, including abdominal pain, nausea, and emesis, should be emphasized so women with these issues will understand the urgency of having them evaluated.
General
A 31-year-old gravida 2, para 1 patient of 36 weeks’ gestation had undergone a laparoscopic Roux-en-Y gastric bypass (RYGB) 4 years ago, resulting in prepregnancy weight loss of 70 kg, or 128% of her excess weight. She awakened with severe lower back pain, abdominal pain, nausea, and vomiting. She came to the emergency department with 10/10 abdominal pain but no fever, chills, vaginal bleeding, or rupture of membranes. Her abdominal examination revealed diffuse tenderness and guarding. She was initially diagnosed as having preterm labor and was given antibiotics, steroids, an epidural catheter for pain management, and intravenous fentanyl as needed. She delivered a healthy baby without complications, but her severe abdominal pain, nausea, and vomiting persisted. Thirty-six hours after her initial presentation, she still had rebound tenderness in the left side of the abdomen. She was afebrile, blood pressure was 104/77 mm Hg, and heart rate was 152/min. White blood cell count was 10.7 ×109/L; anion gap, 13 mEq/L; and lactate dehydrogenase level, 131.7 U/L (2.2 μkat/L). A computed tomography (CT) scan of the abdomen and pelvis with oral and intravenous contrast revealed mesenteric vascular compromise (Figure 1); after the scan, the patient had hematemesis.
what would you do next?
What would you do next?
Highly selective mesenteric angiography
Exploratory laparotomy
Esophagogastrojejunoscopy
Diagnostic laparoscopy
b
1
1
0
1
female
0
0
31
31-40
White
574
original
https://jamanetwork.com/journals/jama/fullarticle/2724790
A 54-year-old man presented for evaluation of new-onset pancytopenia. He had a distant history of viral myocarditis and was taking lisinopril, carvedilol, furosemide, aspirin, and a multivitamin. No new medications had been started in the past year. Twelve years prior, he underwent uncomplicated Roux-en-Y gastric bypass (RYGB) surgery. Review of systems was notable for fatigue and negative for fever, night sweats, weight loss, dyspnea, or abnormal bleeding. Examination revealed temperature of 36.7°C; heart rate, 91/min; blood pressure, 120/70 mm Hg; and body mass index, 20.1 (calculated as weight in kilograms divided by height in meters squared). He was well-appearing with conjunctival pallor but no petechiae, no hepatosplenomegaly, and a normal neurologic examination.Laboratory evaluation showed a white blood cell count of 1.7 × 103/μL with an absolute neutrophil count of 0.17 × 103/μL; hemoglobin level, 7 g/dL with mean corpuscular volume of 103 fL (reticulocyte index, 0.94); and platelet count, 116 × 103/μL. Results of a complete blood cell count from 8 months prior were normal. Results of serum protein electrophoresis were normal, as were levels of lactate dehydrogenase, ferritin, folate, vitamin B12, and thyroid-stimulating hormone. A computed tomography (CT) scan of the abdomen showed no lymphadenopathy, masses, or hepatosplenomegaly. Peripheral blood smear showed occasional pseudo Pelger-Huet cells and rare teardrop cells. Histopathologic examination of bone marrow aspirate revealed a normocellular marrow and dyserythropoiesis with vacuolized and dysplastic erythroid precursors (Figure, panel A), left-shifted granulocytic maturation with vacuolized granulocytic precursors, ring sideroblasts (Figure, panel B), and no increase in blasts. Cytogenetic studies from the bone marrow did not reveal any abnormalities.Histopathologic examination of bone marrow aspirate from patient (A, Wright-Giemsa, original magnification ×1000; B, Prussian blue iron, original magnification ×1000).Check serum trace element levels, including iron, copper, and zincObtain periodic outpatient complete blood cell counts and treat with supportive transfusions as clinically indicatedPerform endoscopy of upper and lower gastrointestinal tract What Would You Do Next?
Check serum trace element levels, including iron, copper, and zinc
Obtain periodic outpatient complete blood cell counts and treat with supportive transfusions as clinically indicated
Perform endoscopy of upper and lower gastrointestinal tract
Start erythropoiesis stimulating agent
Pancytopenia secondary to copper deficiency
A
Check serum trace element levels, including iron, copper, and zinc
The key to the correct diagnosis in this case is the patient’s history of gastric bypass surgery, a risk factor for micronutrient deficiencies including copper deficiency. As part of the workup for pancytopenia or suspected myelodysplastic syndrome (MDS), potentially reversible causes of bone marrow failure such as nutritional deficiencies must be ruled out. Workup for nutritional deficiency should be performed before serologic and endoscopic evaluation for celiac disease. In this patient, iron and zinc levels were normal; however, serum copper level was less than 5 µg/dL (reference range, 70-175). In asymptomatic cases of pancytopenia due to copper deficiency, a trial of copper repletion should precede treatment with blood transfusions or erythropoiesis-stimulating agent.Copper deficiency rarely develops in patients without a history of malabsorption, as the average intake of 1 to 1.6 mg/d typically exceeds the estimated requirement of 0.9 mg/d.1,2 However, RYGB surgery and other bariatric procedures including partial gastrectomy are underrecognized causes of copper deficiency. These procedures bypass portions of the stomach and proximal small intestine, the major sites of copper absorption in the gut. The incidence of copper deficiency after RYGB varies and is estimated to range from 4% to 19% in the first 2 to 5 years after the procedure. However, case reports have described symptomatic copper deficiency occurring more than 10 years after RYGB.2,3 In addition to copper, deficiencies in levels of iron, zinc, vitamin B12, vitamin D, and thiamine have been reported after RYGB.4Other conditions associated with copper deficiency include celiac disease and excessive zinc ingestion. Copper absorption in celiac disease is directly impaired via malabsorption due to small intestinal inflammation. In cases of excess zinc ingestion, copper absorption is indirectly impaired due to an increase in enterocyte metallothionein levels leading to increased fecal excretion of copper.5,6 Copper deficiency has also been reported after prolonged enteral and parenteral nutrition administration when copper is not supplemented.7Patients with prolonged copper deficiency can develop both neurologic and hematologic abnormalities. Neurologic manifestations of copper deficiency include myeloneuropathic changes that present as an unsteady gait, distal muscle weakness, or sensory deficits in the extremities.2,3 Notably, this patient did not have any neurologic findings. Hematologic manifestations of copper deficiency include normocytic or macrocytic anemia and neutropenia.7 Copper is a cofactor in multiple ferroxidases, several of which are involved in hematopoiesis via iron storage, oxidation, and transport.2,7 Thus, many of the hematologic consequences of copper deficiency are linked to iron homeostasis. Bone marrow histopathology can show ringed sideroblasts, hypocellular marrow, and vacuolized erythroid and granulocytic precursors.7 Such dysplastic changes may be interpreted as manifestations of MDS; however, the diagnosis of MDS requires persistent cytopenias in addition to at least 1 of 3 histopathologic criteria from bone marrow aspirate, biopsy, or both: (1) dysplasia in at least 10% of cells from erythroid, granulocytic, or megakaryocytic lineages; (2) 5% to 19% blast cells; or (3) MDS-specific cytogenetic abnormalities such as 5q deletion and monosomy 7.8 Diagnosis of MDS also requires exclusion of other potential etiologies of marrow dysplasia, including HIV infection, acquired sideroblastic anemia, and deficiencies in vitamin B12 or copper. This is particularly important in patients predisposed to micronutrient malabsorption. After RYGB, patients generally undergo scheduled monitoring of micronutrients such as copper. If a copper deficiency develops, then supplementation is typically initiated with oral or intravenous elemental copper.4 Although neurologic deficits frequently do not respond to copper supplementation, hematologic changes are generally fully reversible.2Supplementation was initiated with oral copper gluconate (4 mg 3 times daily). Within 2 months, serum copper level normalized and blood counts improved (hemoglobin, 13.2 g/dL; white blood cell count, 8.5 × 103/μL; absolute neutrophil count, 5.6 × 103/mL; and platelet count, 202 × 103/mL). The copper gluconate dosage was subsequently reduced and maintained at 2 mg daily without recurrence of cytopenias.
General
A 54-year-old man presented for evaluation of new-onset pancytopenia. He had a distant history of viral myocarditis and was taking lisinopril, carvedilol, furosemide, aspirin, and a multivitamin. No new medications had been started in the past year. Twelve years prior, he underwent uncomplicated Roux-en-Y gastric bypass (RYGB) surgery. Review of systems was notable for fatigue and negative for fever, night sweats, weight loss, dyspnea, or abnormal bleeding. Examination revealed temperature of 36.7°C; heart rate, 91/min; blood pressure, 120/70 mm Hg; and body mass index, 20.1 (calculated as weight in kilograms divided by height in meters squared). He was well-appearing with conjunctival pallor but no petechiae, no hepatosplenomegaly, and a normal neurologic examination.Laboratory evaluation showed a white blood cell count of 1.7 × 103/μL with an absolute neutrophil count of 0.17 × 103/μL; hemoglobin level, 7 g/dL with mean corpuscular volume of 103 fL (reticulocyte index, 0.94); and platelet count, 116 × 103/μL. Results of a complete blood cell count from 8 months prior were normal. Results of serum protein electrophoresis were normal, as were levels of lactate dehydrogenase, ferritin, folate, vitamin B12, and thyroid-stimulating hormone. A computed tomography (CT) scan of the abdomen showed no lymphadenopathy, masses, or hepatosplenomegaly. Peripheral blood smear showed occasional pseudo Pelger-Huet cells and rare teardrop cells. Histopathologic examination of bone marrow aspirate revealed a normocellular marrow and dyserythropoiesis with vacuolized and dysplastic erythroid precursors (Figure, panel A), left-shifted granulocytic maturation with vacuolized granulocytic precursors, ring sideroblasts (Figure, panel B), and no increase in blasts. Cytogenetic studies from the bone marrow did not reveal any abnormalities.Histopathologic examination of bone marrow aspirate from patient (A, Wright-Giemsa, original magnification ×1000; B, Prussian blue iron, original magnification ×1000).Check serum trace element levels, including iron, copper, and zincObtain periodic outpatient complete blood cell counts and treat with supportive transfusions as clinically indicatedPerform endoscopy of upper and lower gastrointestinal tract
what would you do next?
What would you do next?
Start erythropoiesis stimulating agent
Perform endoscopy of upper and lower gastrointestinal tract
Obtain periodic outpatient complete blood cell counts and treat with supportive transfusions as clinically indicated
Check serum trace element levels, including iron, copper, and zinc
d
1
1
1
1
male
0
0
54
51-60
White
575
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2717576
A woman in her 20s presented with almost equally sized, round erythematous plaques with central pallor and confluent blisters at the periphery on both forearms. Older lesions showed central crusts. According to the patient, the lesions recurrently appeared during the last 9 years, with a symptom-free period of 2 years (Figure, A). She could not identify or recall a specific cause for these skin lesions and denied self-inflicting them. She had been treated with antibiotics (cefuroxime) as well as with topical steroids (dexamethasone) but denied any lasting effect. Her family history was without abnormalities. She reported being vaccinated against varicella zoster virus and was not immunocompromised.A, A clinical photograph shows an annular erythematous plaque with crusts, bullae, and central pallor. B, Histopathologic specimen shows normal stratum corneum, confluent necrosis of the upper epidermal layers, some multinucleated keratinocytes in the adjacent epidermis, and papillary edema with a subepidermal blister. A moderately dense perivascular lymphocytic infiltrate with eosinophilic and neutrophilic granulocytes is present in the upper dermis (hematoxylin-eosin, original magnification x200).A skin biopsy of the forearm (Figure, B), direct immunofluorescence, and blood samples were obtained. What Is Your Diagnosis?
Linear IgA bullous dermatosis
Cryothermic dermatitis artefacta
Herpes virus infection
Bullous pemphigoid
B. Cryothermic dermatitis artefacta
B
Cryothermic dermatitis artefacta
Histological examination revealed necrotic keratinocytes in the upper and middle epidermal layers up to complete epidermal necrosis with subepidermal blistering and a moderately dense perivascular lymphocytic infiltrate with some eosinophilic and neutrophilic granulocytes (Figure, B). Multinucleated keratinocytes and papillary dermal edema were also present. Direct immunofluorescence was negative.The diagnosis of cryothermic dermatitis artefacta was based on the characteristic clinical and histopathological findings together with the inconsistency of the patient’s history. In cryothermic dermatitis artefacta, lesions are typically circumscribed and localized on accessible body parts, such as the arms. The diagnosis was supported by negative immunofluorescence and negative laboratory results for antibodies against bullous pemphigoid antigen 180. The patient did not show any signs of infection, such as fever or general malaise. She did not have any diagnosed psychological disorders and denied self-inflicting these injuries. A thorough physician-patient consultation took place to discuss the findings, and psychological care was recommended. The diagnosis of cryothermic dermatitis artefacta is challenging because the lesions resemble distinct dermatoses, such as linear IgA bullous dermatosis, herpes virus infections, and bullous pemphigoid. Herpes virus infections can also produce multinucleated keratinocytes. However, herpes simplex and herpes zoster virus infections typically do not recur limited to the arms. Linear IgA bullous dermatosis may present with annular plaques with peripherally localized bullae mostly located on the lower abdomen, thighs, and groin. Linear IgA bullous dermatosis is histopathologically characterized by a subepidermal blister with an underlying predominantly neutrophilic dermal infiltrate and by the linear deposition of IgA at the dermoepidermal junction in direct immunofluorescence. The epidermal roof is uninvolved. Typically, a fixed drug eruption recurs in the same location after reexposure to a specific medication, which is usually not the case with cryothermic dermatitis artefacta. Histopathological necrotic keratinocytes are preferentially located at the basal and lower epidermal layers and there is vacuolar degeneration at the dermal-epidermal junction.There are pathognomonic clues for correct diagnosis. Patients often evade questioning and deny self-inflicting the skin lesions. They frequently have underlying psychological problems, such as borderline personality disorders. Clinical examination of dermatitis artefacta often shows geometrically formed lesions. They can have varying morphologies and appear in different stages of healing at the same time (ie, fresh bullae, crusts, scars).1 The time course of skin changes seems arbitrary.2A biopsy specimen should be obtained preferentially from the erythematous nonulcerated periphery of the lesion. Histopathologically, necrotic keratinocytes develop downward beginning from the outer and/or upper epidermal layers. If the biopsy specimen is obtained toward the center of the lesion, full–thickness epidermal necrosis could be present. In the dermis, variably dense lymphocytic infiltrate is present, often with eosinophilic and neutrophilic granulocytes. Multinucleated keratinocytes also have been reported in the setting of self-inflicted injuries caused by extreme heat.3Cryothermic dermatitis artefacta belongs to a broad spectrum of factitious diseases with lesions, which are most often self-induced by patients, and several cases of the condition in young adults have been reported in the literature.1,2,4 Aerosol sprays typically serve as blister-inducing agents. In a study, blisters were reproduced on a volunteer’s healthy skin by using a deodorant spray.5 Spraying from a close proximity of about 5 centimeters for at least 15 seconds can induce a temperature difference of about 60°C under laboratory conditions.6 “Frosties” are a special type of cryothermic dermatitis artefacta that adolescents and young adults with risk-taking behavior use to produce skin lesions, especially as a trial of courage within peer groups. This social behavior seems to be popular in South Australia.7 In the United States, a recent trend among adolescents referred to as the salt and ice challenge has produced similar clinical findings. Participants pour salt on their bodies (preferably on the arms) and ice is then placed on top, resulting in an injury similar to frostbite.8,9Cryothermic dermatitis artefacta should be part of the differential diagnosis for young patients with recurrent bullous eruptions and necrotic or multinucleated keratinocytes as well as lymphocytic infiltrate in the histological findings.
Dermatology
A woman in her 20s presented with almost equally sized, round erythematous plaques with central pallor and confluent blisters at the periphery on both forearms. Older lesions showed central crusts. According to the patient, the lesions recurrently appeared during the last 9 years, with a symptom-free period of 2 years (Figure, A). She could not identify or recall a specific cause for these skin lesions and denied self-inflicting them. She had been treated with antibiotics (cefuroxime) as well as with topical steroids (dexamethasone) but denied any lasting effect. Her family history was without abnormalities. She reported being vaccinated against varicella zoster virus and was not immunocompromised.A, A clinical photograph shows an annular erythematous plaque with crusts, bullae, and central pallor. B, Histopathologic specimen shows normal stratum corneum, confluent necrosis of the upper epidermal layers, some multinucleated keratinocytes in the adjacent epidermis, and papillary edema with a subepidermal blister. A moderately dense perivascular lymphocytic infiltrate with eosinophilic and neutrophilic granulocytes is present in the upper dermis (hematoxylin-eosin, original magnification x200).A skin biopsy of the forearm (Figure, B), direct immunofluorescence, and blood samples were obtained.
what is your diagnosis?
What is your diagnosis?
Bullous pemphigoid
Linear IgA bullous dermatosis
Herpes virus infection
Cryothermic dermatitis artefacta
d
0
0
1
1
female
0
0
25
21-30
null
576
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2720312
An adolescent girl with polycystic ovarian syndrome (PCOS) presented for evaluation of hyperpigmented skin lesions affecting her neck, upper limbs, and trunk. The lesions had started about 6 months previously as recurrent crops of pruritic erythematous papules that subsided leaving behind brownish-bluish black hyperpigmentation after each episode.On examination, there were multiple, brown, hyperpigmented patches symmetrically affecting her neck, axillae, antecubital fossa, and intermammary and inframammary areas (Figure, A). The patches showed a reticulate pattern at the peripheries. Intermammary patches had central erythema and a few subsiding papules. Additionally, there were dark brown to bluish-gray patches with reticulation on the lower back and dorsum of the neck (Figure, B). These patches also had a few resolving erythematous papules in the periphery. There was no history suggestive of allergic rhinitis or bronchial asthma. Her IgE levels and thyroid status were normal. Her fasting blood glucose and basal insulin levels were not raised. There was no family history of similar lesions. The rest of the skin, nails, and mucosae were all normal. Potassium hydroxide mount analysis from skin scrapings did not reveal fungal hyphae or spores. A skin biopsy was performed from lesions on the lower back, and the specimen was submitted for histopathologic analysis (Figure, C and D).A, Illustrated are multiple brown hyperpigmented patches symmetrically affecting the neck, axillae, antecubital fossa, and intermammary and inframammary areas. The patches show a reticulate pattern at the peripheries. The intermammary patches have central erythema and a few overlying edematous papules. B, Bluish-gray pigmented patches with prominent reticulation on lower back. C, Histopathologic lesional specimen shows mild epidermal hyperkeratosis, acanthosis, multiple foci of spongiosis, and an enhanced basal layer pigmentation. There is scant lichenoid and moderate perivascular lymphomononuclear and polymorphonuclear infiltrate, basal cell vacuolization, and melanin incontinence (hematoxylin-eosin). D, Histopathologic lesional specimen at higher power reveals spongiosis, occasional neutrophilic exocytosis (yellow arrowheads), basal cell vacuolar degeneration, and necrotic keratinocytes (black arrowheads) (hematoxylin-eosin). What Is Your Diagnosis?
Confluent and reticulated papillomatosis
Prurigo pigmentosa
Dowling-Degos disease
Lichen planus pigmentosus
A. Prurigo pigmentosa (PP)
A
Confluent and reticulated papillomatosis
The epidermis showed mild hyperkeratosis and acanthosis. There was scant lichenoid and moderate perivascular, lymphomononuclear, and polymorphonuclear inflammatory infiltrate. Additionally, basal cell degeneration and prominent melanin incontinence was observed (Figure, C). Higher power revealed spongiosis, occasional neutrophilic exocytosis, basal cell vacuolar degeneration, and necrotic keratinocytes (Figure, D). A diagnosis of PP was made. She was prescribed oral minocycline, 100 mg once a day, after her antinuclear antibody profile was found to be normal. At last follow-up, the patient had completed 3 months of minocycline therapy, and there had been no recurrence of erythematous papules. The pigmentation was persistent.Flexural hyperpigmentation can be observed in diverse dermatoses. Prurigo pigmentosa is an inflammatory dermatosis reported predominantly in young female patients of Asian descent.1 It initially presents with recurrent crops of reticulated, pruritic, erythematous, crusted or urticarial papules and papulovesicles.2 The nape of the neck, central chest (prominently intermammary and submammary areas), and lumbosacral region are commonly involved.3 The lesions resolve with broad reticulate hyperpigmentation. The early pruritic lesions can mimic dermatitis herpetiformis, scabies, and prurigo simplex, and feature spongiosis and superficial perivascular neutrophilic infiltrate in dermis. The established lesions demonstrate neutrophilic exocytosis, keratinocyte necrosis, and interface dermatitis. The resolving lesions show occasional neutrophilic exocytosis, focal interface dermatitis, and prominent dermal melanin incontinence.4 Lesions in multiple stages often coexist.5The hyperpigmented stages of PP can simulate various other dermatoses: confluent and reticulated papillomatosis of Gougerot and Carteaud (CARP) and acanthosis nigricans (AN) are important differentials. CARP typically affects the axillae, neck, central chest, and interscapular region and presents with asymptomatic, hyperpigmented ichthyosiform patches or thin plaques, with a characteristic peripheral reticulation. Histopathologic findings demonstrate undulating hyperkeratosis, papillomatosis, acanthosis, an enhanced basal layer melanization, and mild superficial perivascular lymphomononuclear infiltrate.1 AN can occur in the setting of PCOS. It usually affects the nape and flexural areas of neck, antecubital fossae, axillae, and groin with velvety, smooth hyperpigmentation. Histopathologic findings of AN can resemble those of the CARP, though changes in CARP are milder, and hyperkeratosis and basal layer hyperpigmentation are more prominent features than acanthosis in AN.6In the present patient, the involvement of the neck and upper limb flexures and demonstration of hyperkeratosis and acanthosis suggested a possible overlap between PP, CARP, and AN, especially in the setting of PCOS. AN lacks the characteristic peripheral reticulation and overlying scaling or crusting that is observed in CARP and PP. Moreover, both CARP and AN lack the neutrophilic exocytosis and interface dermatitis, which are classically seen in PP. The hyperpigmentation in both CARP and AN is epidermal and lacks the dermal melanin incontinence observed in PP. The lesions of CARP typically resolve after treatment, whereas the dermal pigmentation of PP may take years to resolve, though the inflammatory papules subside with treatment.7 The history of prior pruritic, reticulated erythematous papules at all sites, prominent interface dermatitis, and melanin incontinence favored the diagnosis of PP in the present case. It has been suggested that PP and CARP represent different manifestation along a similar spectrum of disease and that PP is a more inflammatory variant of CARP in ethnic skin.8The flexural or inverse variant of lichen planus pigmentosus (LPP) has been described,9 but it prominently lacks a prior inflammatory phase. The reticulate pattern in LPP demonstrates thin delicate reticulation, which is usually discovered only on a closer look, whereas the reticulation in PP is better appreciated from a distance.2 Histopathologic findings in LPP can simulate those seen in late stages of PP, though flexural LPP lacks the neutrophilic exocytosis and acanthosis.9The lesions in Dowling-Degos disease are hyperpigmented macules demonstrating delicate reticulation that classically affect major flexures. Comedolike papules, perioral pitted scars, and palmar pits can be observed. The histopathologic finding show follicular plugging, an antlerlike pattern of rete pegs, intense basal layer pigmentation, and conspicuously absent melanin incontinence.10To conclude, flexural hyperpigmentation can have various causes and manifestations. Involvement of typical sites, history of prior inflammatory papules, a characteristic peripheral reticular pattern of hyperpigmentation, and corroborative histopathologic results helped to establish the diagnosis of PP in the present case.
Dermatology
An adolescent girl with polycystic ovarian syndrome (PCOS) presented for evaluation of hyperpigmented skin lesions affecting her neck, upper limbs, and trunk. The lesions had started about 6 months previously as recurrent crops of pruritic erythematous papules that subsided leaving behind brownish-bluish black hyperpigmentation after each episode.On examination, there were multiple, brown, hyperpigmented patches symmetrically affecting her neck, axillae, antecubital fossa, and intermammary and inframammary areas (Figure, A). The patches showed a reticulate pattern at the peripheries. Intermammary patches had central erythema and a few subsiding papules. Additionally, there were dark brown to bluish-gray patches with reticulation on the lower back and dorsum of the neck (Figure, B). These patches also had a few resolving erythematous papules in the periphery. There was no history suggestive of allergic rhinitis or bronchial asthma. Her IgE levels and thyroid status were normal. Her fasting blood glucose and basal insulin levels were not raised. There was no family history of similar lesions. The rest of the skin, nails, and mucosae were all normal. Potassium hydroxide mount analysis from skin scrapings did not reveal fungal hyphae or spores. A skin biopsy was performed from lesions on the lower back, and the specimen was submitted for histopathologic analysis (Figure, C and D).A, Illustrated are multiple brown hyperpigmented patches symmetrically affecting the neck, axillae, antecubital fossa, and intermammary and inframammary areas. The patches show a reticulate pattern at the peripheries. The intermammary patches have central erythema and a few overlying edematous papules. B, Bluish-gray pigmented patches with prominent reticulation on lower back. C, Histopathologic lesional specimen shows mild epidermal hyperkeratosis, acanthosis, multiple foci of spongiosis, and an enhanced basal layer pigmentation. There is scant lichenoid and moderate perivascular lymphomononuclear and polymorphonuclear infiltrate, basal cell vacuolization, and melanin incontinence (hematoxylin-eosin). D, Histopathologic lesional specimen at higher power reveals spongiosis, occasional neutrophilic exocytosis (yellow arrowheads), basal cell vacuolar degeneration, and necrotic keratinocytes (black arrowheads) (hematoxylin-eosin).
what is your diagnosis?
What is your diagnosis?
Lichen planus pigmentosus
Dowling-Degos disease
Confluent and reticulated papillomatosis
Prurigo pigmentosa
c
0
1
1
1
female
0
1
15
11-20
Black
577
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2720742
An elementary school-aged girl with cystic fibrosis (CF) presented with a 2-year history of a relapsing-remitting, evanescent, salmon-pink maculopapular cutaneous eruption associated with joint pain. Each episode of joint pain and cutaneous eruption lasted 24 to 48 hours with episodes occurring anywhere from every other day to once weekly. The patient’s mother was unable to identify any possible triggers because the episodes occurred independent of pulmonary CF exacerbations or antibiotic use. The patient’s joint pain and cutaneous symptoms resolved completely between each episode and were not associated with fever. Review of photos taken during the patient’s flares revealed a variable eruption consisting of oval-shaped, salmon-pink macules on the trunk, urticarial-appearing papules on the dorsal hands, and flagellate arrays of petechial macules on the upper medial arms (Figure 1). This eruption was intermittently pruritic but otherwise was asymptomatic. Her joint pain recurred with each episode of cutaneous eruption and primarily affected the wrist, knee, and ankle joints bilaterally. She had been treated with naproxen during these flares, which improved her joint pain but not her cutaneous symptoms. A complete blood cell count and comprehensive metabolic panel as well as C-reactive protein and erythrocyte sedimentation rate obtained during a flare were normal. Titers for parvovirus, Epstein–Barr virus, and antistreptolysin O were also negative.A, Approximately 1-cm, oval-shaped, salmon-pink macules on the right lower abdomen and right lateral thigh. B, 3- to 4-mm pink urticarial-appearing papules on the dorsal hand. C, Flagellate arrays of nonblanching 1- to 2-mm petechial macules on the upper medial arm. What Is Your Diagnosis?
Systemic-onset juvenile idiopathic arthritis
Cystic fibrosis-associated episodic arthritis
Hypertrophic pulmonary osteoarthropathy
Serum sickness-like reaction
B. Cystic fibrosis-associated episodic arthritis
B
Cystic fibrosis-associated episodic arthritis
Histopathologic examination revealed a normal epidermis overlying a mild perivascular and interstitial lymphocytic infiltrate with rare neutrophils (Figure 2). Examination also demonstrated negative direct immunofluorescence results for IgG, IgM, IgA, C3, and fibrinogen.Histopathologic findings of cystic fibrosis-associated episodic arthritis show normal epidermis overlying a mild perivascular and interstitial lymphocytic infiltrate with rare neutrophils (hematoxylin-eosin stain, original magnification ×400).Cystic fibrosis-associated episodic arthritis (CFEA) is the more common of 2 arthropathies in patients with CF; the less common variety is termed hypertrophic pulmonary osteoarthropathy (HPOA).1-3 Cystic fibrosis-associated episodic arthritis is frequently associated with cutaneous manifestations including an erythematous maculopapular eruption, purpura of the legs, erythema nodosum, and cutaneous vasculitis.2-5 Patients have joint pain, swelling, and limited movement. Cystic fibrosis-associated episodic arthritis can be quite disabling, but causes no radiographic evidence of joint destruction.1-4 Cystic fibrosis-associated episodic arthritis flares typically occur independent of pulmonary CF exacerbations. In addition, they have a sudden onset and last 5 to 7 days before remitting spontaneously. Throughout the course, patients remain afebrile and constitutionally well.1,2 Rheumatoid factor and antinuclear antibodies are negative.2 Though circulating immune complexes detected by C1q binding are found in up to 30% of adult patients with CF, they are largely undetectable in the peripheral blood during CFEA flares.1,2,5 In addition, synovial fluid from CFEA-affected joints is sterile with little evidence of inflammation.2 However, synovial biopsies from several cases have shown IgM on immunofluorescence results, sometimes also with IgG, C3, and fibrinogen.2The pathophysiology of CFEA is poorly understood. Despite the inability to consistently detect serum immune complexes during flares, CFEA is postulated to be immunologically mediated.1 Seventy percent of CF patients have positive antineutrophil cytoplasmic antibodies against bactericidal/permeability-increasing protein, which is linked to colonization with Pseudomonas aeruginosa and may be pathogenic.6 Thus, the pathophysiology of CFEA may be comparable to the suspected underlying mechanism of bowel-associated dermatosis-arthritis syndrome, in which immunoglobulin against microorganisms in a blind loop of bowel binds complement to form immune complexes that deposit in the skin and joints, causing arthropathy and papulopustular eruptions.4,5 Another suggested etiology is an underlying vasculitis; however, only a subset of patients with CFEA have evidence of vasculitis on clinical examination, histopathologic analysis, or serology results.2Treatment of CFEA is supportive because episodes are self-limiting and respond to nonsteroidal anti-inflammatory drugs.2,4 In severe cases, prednisolone or intravenous antibiotics may be helpful.2The differential diagnosis in this case includes HPOA, Still disease, serum sickness-like reaction, and hyper IgD syndrome (HIDS). Hypertrophic pulmonary osteoarthropathy is characterized by symmetrical joint tenderness and swelling, and is often associated with gynecomastia or mastalgia.2 However, HPOA generally affects older patients with significantly worse pulmonary function, flares in concert with pulmonary CF exacerbations, and demonstrates periosteal new bone formation on radiographs.2,3,5 Because HPOA flares with pulmonary CF exacerbations, it responds to antibiotic therapy and is postulated to be a reactive arthritis.2 Furthermore, most patients with HPOA have circulating immune complexes.3,5 Our patient’s young age, lack of advanced pulmonary disease, and cutaneous flares independent of CF exacerbations were inconsistent with HPOA. Systemic-onset juvenile idiopathic arthritis, also known as Still disease, can present with an evanescent, nonpruritic, salmon-colored morbilliform eruption. Histopathologic analysis reveals a perivascular and interstitial neutrophilic infiltrate similar to that found in this patient.7 However, the patient’s normal inflammatory markers, lack of fever, lymphadenopathy, and hepatosplenomegaly were not consistent with Still disease.7 Serum sickness-like reaction has also been reported in patients with CF, particularly following antibiotic exposure.8 Patients present with an urticarial eruption, fever, lymphadenopathy, arthralgias, myalgias, and proteinuria. This patient’s constellation of symptoms and presentation independent of antibiotic use was not supportive of this diagnosis. Hyper IgD syndrome presents with a morbilliform or urticarial eruption and arthropathy similar to those found in this patient with CFEA.9,10 Unlike CFEA, patients with HIDS present in the first year of life with recurrent episodes of fever associated with severe abdominal pain, tender lymphadenopathy, and splenomegaly.9,10 Direct immunofluorescence of lesional skin in HIDS demonstrates perivascular deposition of IgD and C3 complexes.10 Overall, CFEA clinically mimics several other entities, causing it to remain an underrecognized and rarely reported condition.
Dermatology
An elementary school-aged girl with cystic fibrosis (CF) presented with a 2-year history of a relapsing-remitting, evanescent, salmon-pink maculopapular cutaneous eruption associated with joint pain. Each episode of joint pain and cutaneous eruption lasted 24 to 48 hours with episodes occurring anywhere from every other day to once weekly. The patient’s mother was unable to identify any possible triggers because the episodes occurred independent of pulmonary CF exacerbations or antibiotic use. The patient’s joint pain and cutaneous symptoms resolved completely between each episode and were not associated with fever. Review of photos taken during the patient’s flares revealed a variable eruption consisting of oval-shaped, salmon-pink macules on the trunk, urticarial-appearing papules on the dorsal hands, and flagellate arrays of petechial macules on the upper medial arms (Figure 1). This eruption was intermittently pruritic but otherwise was asymptomatic. Her joint pain recurred with each episode of cutaneous eruption and primarily affected the wrist, knee, and ankle joints bilaterally. She had been treated with naproxen during these flares, which improved her joint pain but not her cutaneous symptoms. A complete blood cell count and comprehensive metabolic panel as well as C-reactive protein and erythrocyte sedimentation rate obtained during a flare were normal. Titers for parvovirus, Epstein–Barr virus, and antistreptolysin O were also negative.A, Approximately 1-cm, oval-shaped, salmon-pink macules on the right lower abdomen and right lateral thigh. B, 3- to 4-mm pink urticarial-appearing papules on the dorsal hand. C, Flagellate arrays of nonblanching 1- to 2-mm petechial macules on the upper medial arm.
what is your diagnosis?
What is your diagnosis?
Serum sickness-like reaction
Cystic fibrosis-associated episodic arthritis
Hypertrophic pulmonary osteoarthropathy
Systemic-onset juvenile idiopathic arthritis
b
0
1
0
1
female
0
0
2
0-10
null
578
original
https://jamanetwork.com/journals/jamaneurology/fullarticle/2719276
A man in his late 60s with a diagnosis of type 2 diabetes for more than 10 years that was complicated by neuropathy and retinopathy presented with persistent chorea in the left upper extremity for about 1 month despite aggressive correction with insulin. He was alert and oriented with healthy vital signs. No evidence of cognitive impairments was present on bedside testing results. The patient denied any recent infections and risk for human immunodefiency virus (HIV) infection. Neurological examination results were significant for choreiform and hemiballistic movements involving the left upper extremity. His home medications were carefully reviewed to rule out any medications that could cause chorea. A review of his previous records indicated an admission to an outside facility 3 weeks beforehand with severe hyperglycemia and a hemoglobin A1c level of 14.1% (to convert to the proportion of total hemoglobin, multiply by 0.01). Despite an aggressive correction of blood glucose levels and metabolic correction, his choreiform movements had remained unresolved. On admission, a complete electrolyte and toxicology panel, including calcium, magnesium, and phosphorus, yielded normal results. The patient’s serum glucose level was 159 mg/dL (to convert to millimoles per liter, multiply by 0.0555) and his hemoglobin A1c was 10.9% without ketoacidosis. Test results for antinuclear antibodies, lupus anticoagulants, and antiphospholipid antibodies were negative. Serum thyrotropin levels, liver function test results, and parathyroid hormone levels were healthy. Copper studies, a peripheral smear, and a routine electroencephalogram yielded normal results. A general screening for cancers yielded negative results. The patient underwent brain magnetic resonance imaging (MRI) (Figure), which showed a T1 hyperintensity in the right caudate head and a lentiform nucleus with no restricted diffusion. Oral administration of olanzapine, 2.5 mg daily, in divided doses with careful monitoring of blood glucose levels resulted in the complete resolution of chorea.Magnetic resonance imaging shows T1 hyperintensity in the right caudate head (A) and a lentiform nucleus with no restricted diffusion (B). What Is Your Diagnosis?
Postinfectious/autoimmune chorea
Paraneoplastic chorea
Diabetic striatopathy
Acute stroke
C. Diabetic striatopathy
C
Diabetic striatopathy
The patient’s presentation of a unilateral subacute onset of choreiform movement that was associated with pathognomonic MRI findings in the setting of long-standing diabetes led us to the diagnosis of diabetic striatopathy. Diabetic striatopathy is a constellation of neurologic abnormalities that is characterized by sudden-onset hemiballism or hemichorea and striatal abnormalities on neuroimaging in a poorly controlled patient with diabetes.1 Delayed onset of hemichorea after an episode of hyperglycemia with or without ketoacidosis has also been reported.2 Dyskinesias, including hemichorea-hemiballism, occur because of the dysfunction of the corpus striatum and are usually reversible with the restoration of homeostasis. Diabetic striatopathy is an underdiagnosed complication of diabetes that is more frequently encountered in type 2 diabetes than type 1 diabetes.3 Chorea has also been reported after hypoglycemic episodes in people with diabetes.4 The main mode of treatment includes the correction of the metabolic derangement by fluids and insulin infusion. Patients with refractory disease require additional drugs, including γ-aminobutyric acid–ergic, neuroleptic, or dopamine-depleting agents as in this case.5 In 90% of the patients, clinical symptoms completely resolve with or without radiologic resolution, varying from 2 to 28 days. As noted in the workup, metabolic disorders, such as hypernatremia and hyponatremia, hypercalcemia and hypocalcemia, hypomagnesemia, hyperthyroidism, and hypoparathyroidism and hyperparathyroidism can cause chorea and they need to be ruled out.Sydenham chorea (SC) is a nonsuppurative sequela of group A streptococcus infection and rheumatic fever.6 This is typically seen in children and rarely presents in adults. There was no history or other evidence of streptococcal infection in the patient. Moreover, the unilateral presentation of chorea makes SC quite unlikely in this patient. Chorea can be a key manifestation of systemic lupus erythematosus, Sjögren syndrome, and antiphospholipid syndrome. Chorea that are associated with autoimmune disorders are thought to share a pathophysiology with postinfectious SC.7 A subacute form of chorea has been reported in a variant of Creutzfeldt-Jakob disease,8 HIV infection, and slow-growing tumors. Magnetic resonance imaging in this case ruled out slow-growing tumors. The lack of cognitive changes, healthy electroencephalogram results, and the clinical course ruled out the new variant form of Creutzfeldt-Jacob disease and HIV infection in this patient.Paraneoplastic chorea has been reported with renal cancer, small cell cancer of the lung, and Hodgkin and non-Hodgkin lymphomas. Nontraditional movements, including chorea, have been reported as paraneoplastic manifestations of anti–N-methyl-d-aspartate antibody–secreting tumors. In this patient, unilateral chorea and negative screen results made it unlikely that the chorea was of paraneoplastic origin. Wilson disease is a disorder of copper excretion caused by a mutation in the ATP7B gene that results in relentless multisystemic copper accumulation. Investigation demonstrates low ceruloplasmin levels in most patients (normal in 5-15% of cases), low serum copper levels, increased 24-hour urine copper excretion, and abnormal liver function test results that were not seen in this patient. Brain MRI usually shows bilateral hyperintensities in the basal ganglia or claustrum on T2-weighted images or signal abnormalities in the midbrain. Chorea is rare in Wilson disease and is usually bilateral.Chorea can occur as a consequence of acute ischemic or hemorrhagic stroke or in the context of low-grade ischemic changes in the basal ganglia without obvious infarction.9 The neostriatum and the subthalamic nucleus have been frequently implicated in strokes that cause hemichorea. Hemichorea or hemiballism occurs in less than 1% of patients with a stroke.10 In this patient, neuroimaging results showed no findings of restricted diffusion that was concerning for a stroke. Most patients with chorea following a stroke show a spontaneous resolution of chorea within 1 or 2 years. However, prolonged and persistent chorea that is disabling that requires pharmacotherapy or surgery has been reported.
Neurology
A man in his late 60s with a diagnosis of type 2 diabetes for more than 10 years that was complicated by neuropathy and retinopathy presented with persistent chorea in the left upper extremity for about 1 month despite aggressive correction with insulin. He was alert and oriented with healthy vital signs. No evidence of cognitive impairments was present on bedside testing results. The patient denied any recent infections and risk for human immunodefiency virus (HIV) infection. Neurological examination results were significant for choreiform and hemiballistic movements involving the left upper extremity. His home medications were carefully reviewed to rule out any medications that could cause chorea. A review of his previous records indicated an admission to an outside facility 3 weeks beforehand with severe hyperglycemia and a hemoglobin A1c level of 14.1% (to convert to the proportion of total hemoglobin, multiply by 0.01). Despite an aggressive correction of blood glucose levels and metabolic correction, his choreiform movements had remained unresolved. On admission, a complete electrolyte and toxicology panel, including calcium, magnesium, and phosphorus, yielded normal results. The patient’s serum glucose level was 159 mg/dL (to convert to millimoles per liter, multiply by 0.0555) and his hemoglobin A1c was 10.9% without ketoacidosis. Test results for antinuclear antibodies, lupus anticoagulants, and antiphospholipid antibodies were negative. Serum thyrotropin levels, liver function test results, and parathyroid hormone levels were healthy. Copper studies, a peripheral smear, and a routine electroencephalogram yielded normal results. A general screening for cancers yielded negative results. The patient underwent brain magnetic resonance imaging (MRI) (Figure), which showed a T1 hyperintensity in the right caudate head and a lentiform nucleus with no restricted diffusion. Oral administration of olanzapine, 2.5 mg daily, in divided doses with careful monitoring of blood glucose levels resulted in the complete resolution of chorea.Magnetic resonance imaging shows T1 hyperintensity in the right caudate head (A) and a lentiform nucleus with no restricted diffusion (B).
what is your diagnosis?
What is your diagnosis?
Diabetic striatopathy
Postinfectious/autoimmune chorea
Acute stroke
Paraneoplastic chorea
a
1
1
1
1
male
0
0
68
61-70
null
579
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2718839
A 50-year-old woman with hypertension, diabetes, and allergies presented to the ophthalmology clinic for an evaluation of uveitis after experiencing 9 months of eye redness and pain that started in her right eye and involved both eyes within a few months. The patient had a history of asthma and had recently been hospitalized after radiologic findings of pulmonary infiltrates. The patient was diagnosed with pneumonia, myocarditis, and hypereosinophilic syndrome and received high-dose intravenous corticosteroids. At hospital discharge, the patient’s medications were changed to prednisone, 11 mg orally once daily, and azathioprine, 100 mg orally once daily.Although the severity of the ocular symptoms was variable, there was no improvement with the described medication regimen or with currently prescribed topical corticosteroids given 4 times daily in both eyes. The patient denied rash, headache, and joint pain. Serologic testing ruled out infectious causes of inflammation.On examination, best-corrected visual acuity was 20/25 OU. Intraocular pressure for the right eye was 16 mm Hg and for the left eye was 24 mm Hg. Slitlamp examination showed trace hyperemia of the right eye and 2+ diffuse scleral hyperemia of the left eye (Figure). Corneas were clear bilaterally, and sensitivity was symmetric and full. There was no cell or flare in the anterior chamber, and she had clear lenses with normal irises. The fundus examination was unremarkable bilaterally with no vitreous cell or haze, a flat macula, a cup-disc ratio of 0.3, and a peripheral retina without holes or tears.Increase oral corticosteroids dosage to 40 mg daily What Would You Do Next?
Increase oral corticosteroids dosage to 40 mg daily
Perform scleral biopsy
Increase to hourly topical corticosteroid use
Increase azathioprine dosage to 500 mg daily
Churg-Strauss syndrome (eosinophilic granulomatosis with polyangiitis)
B
Perform scleral biopsy
This patient’s lack of improvement with high-dose corticosteroids makes it unlikely that increasing oral corticosteroid dosage (choice A) or topical corticosteroid use (choice C) would be effective to manage the ocular inflammation. Azathioprine treatment at a dosage of 1 mg/kg/d may be initiated for ocular inflammation, but the patient’s symptoms remained uncontrolled at a dosage of 100 mg/d. Increasing the azathioprine dosage to a maximum of 3 mg/kg/d can be used,1 but a dosage adjustment to 500 mg/d (choice B) places the patient at risk for significant adverse effects without initially reexamining the reason why the present therapy failed.The 1990 criteria of the American College of Rheumatology require 4 of 6 items for the diagnosis with Churg-Strauss syndrome: (1) asthma, (2) a blood eosinophil level more than 10%, (3) mononeuropathy or polyneuropathy, (4) pulmonary infiltrates, (5) paranasal sinus abnormality, and (6) extravascular eosinophils noted on biopsy findings.2 Results of pathologic examination of the scleral biopsy sample revealed a substantially increased number of eosinophils. These results provided the fourth criterion to establish the diagnosis of Churg-Strauss in this patient.Inflammation in the eye can be a sign of various systemic diseases. Scleritis can manifest as hyperemia in the anterior portion of the eye or result in the false appearance of a white, quiet eye in the posterior portion. Pain with eye movement differentiates this diagnosis from episcleritis and should trigger a more thorough questioning of surgical history and review of systems.3 Rheumatoid arthritis and systemic lupus erythematosus are known to be associated with scleritis, but the seronegative spondyloarthropathies, relapsing polychondritis, and granulomatosis with polyangiitis must be considered.4Churg-Strauss syndrome is a rare disease with small-vessel vasculitis that manifests in multiple organ systems, and patients may seek care from many medical specialists. The syndrome can vary in presentation because there is no pathognomonic ocular finding. Inflammation may present as scleritis, uveitis, or posterior vasculitis. Inducing remission of inflammation is critical for these patients because death can occur secondary to heart failure.Nonsteroidal antiinflammatory medications are first-line agents prescribed for scleritis to reduce pain, prevent macular edema, and enhance mydriasis.5,6 In contrast, patients with Churg-Strauss syndrome are treated with corticosteroids and cytotoxic agents. Initial management of systemic manifestations may include high doses of pulsed corticosteroids such as 15 mg/kg intravenously infused for 60 minutes daily for 2 to 3 days. Completion of this treatment can be followed by prednisone given at 1 mg/kg/d, which is tapered by 5 mg each week for 3 to 4 weeks, then is slowly tapered to 1 mg each week until the medication can either be stopped or a low-maintenance dosage is given to control the asthma.7Cyclophosphamide is conventionally used for management of Churg-Strauss syndrome. Some physicians have replaced oral cyclophosphamide prescribed at 2 mg/kg/d with pulsed therapy such as 0.6 g/m2 every 2 weeks and then monthly for a year.8 Medications such as methotrexate and mycophenolate mofetil can maintain remission.If ocular or other systemic disease processes in a patient with Churg-Strauss syndrome is treated in isolation with such therapies as nonsteroidal anti-inflammatories or antimetabolites alone, the underlying cause is not being addressed, and this delay can be fatal.The patient’s inflammation dramatically improved after 2 infusions of cyclophosphamide, 1 g/m2. No further infusions were given owing to abdominal pain and myalgias. Mycophenolate mofetil was started at 1.5 g orally daily and increased by 500 mg every 5 days until a maximum dosage of 3 g/d, at which time oral prednisone was tapered. For the past 4 years, the patient has had no inflammation after stopping prednisone. Tapering the mycophenolate was attempted, but inflammation returned while receiving approximately 500 mg of mycophenolate daily. The patient is presently doing well while receiving maintenance therapy of mycophenolate, 1 g/d.
Ophthalmology
A 50-year-old woman with hypertension, diabetes, and allergies presented to the ophthalmology clinic for an evaluation of uveitis after experiencing 9 months of eye redness and pain that started in her right eye and involved both eyes within a few months. The patient had a history of asthma and had recently been hospitalized after radiologic findings of pulmonary infiltrates. The patient was diagnosed with pneumonia, myocarditis, and hypereosinophilic syndrome and received high-dose intravenous corticosteroids. At hospital discharge, the patient’s medications were changed to prednisone, 11 mg orally once daily, and azathioprine, 100 mg orally once daily.Although the severity of the ocular symptoms was variable, there was no improvement with the described medication regimen or with currently prescribed topical corticosteroids given 4 times daily in both eyes. The patient denied rash, headache, and joint pain. Serologic testing ruled out infectious causes of inflammation.On examination, best-corrected visual acuity was 20/25 OU. Intraocular pressure for the right eye was 16 mm Hg and for the left eye was 24 mm Hg. Slitlamp examination showed trace hyperemia of the right eye and 2+ diffuse scleral hyperemia of the left eye (Figure). Corneas were clear bilaterally, and sensitivity was symmetric and full. There was no cell or flare in the anterior chamber, and she had clear lenses with normal irises. The fundus examination was unremarkable bilaterally with no vitreous cell or haze, a flat macula, a cup-disc ratio of 0.3, and a peripheral retina without holes or tears.Increase oral corticosteroids dosage to 40 mg daily
what would you do next?
What would you do next?
Increase to hourly topical corticosteroid use
Perform scleral biopsy
Increase azathioprine dosage to 500 mg daily
Increase oral corticosteroids dosage to 40 mg daily
b
0
1
1
1
female
0
0
50
41-50
null
580
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2718840
A woman in her early 60s presented with an 8-month history of central scotoma and blurry vision in her right eye. Her medical history was significant for a motor vehicle crash that occurred almost a year earlier, with no direct head trauma or eye injury. On examination, her best-corrected visual acuity was 20/500 OD and 20/30 OS. Anterior segment examination was remarkable for pseudophakia on the right eye and mild cataract in the left. Fundus examination was remarkable for a slightly blunted foveal reflex in the right eye. Spectral-domain optical coherence tomography (SD-OCT) showed segmental atrophy of the inner retina (Figure 1A). Fluorescein angiography was unrevealing (Figure 1B).A, Spectral-domain optical coherence tomography of the right eye. B, Fluorescein angiogram of the right eye. What Would You Do Next?
Optical coherence tomography angiography
Automated visual field
Full-field electroretinogram
Microperimetry
Inner retinal atrophy after traumatic Purtscher retinopathy
A
Optical coherence tomography angiography
Purtscher retinopathy was initially described in 1910 as a trauma-related retinopathy.1 When nontraumatic, the condition is typically described as Purtscher-like retinopathy.2 Purtscher retinopathy is an occlusive microvasculopathy thought to be caused by microembolization of the chorioretinal vessels by fat, air, or leukoaggregate emboli.2 Both entities present with visual disturbances along with the typical acute phase findings of Purtscher-flecken, cotton-wool spots, retinal hemorrhages, and papilledema.2 Purtscher-flecken refers to the characteristic vessel-sparing, polygonal, white retinal lesions that develop parafoveally.2 Fluorescein angiography (FA) shows hypofluorescence corresponding to the cotton-wool spots, retinal hemorrhages in the early phases, and nonperfusion and leakage of retinal microvasculature in the late phases.3 The results of SD-OCT often show retinal edema and subretinal fluid. With time, typical Purtscher retinopathy fundus and FA findings often resolve,3 and inner retinal atrophy might be observed on SD-OCT.On further questioning, the patient mentioned that during the motor crash she had sustained a year earlier, she experienced multiple long-bone fractures, severe blood loss, and cerebral fat embolism. On regaining consciousness 10 days after the crash she reported visual loss in both eyes. Fundus examination at that time had revealed multiple emboli and papilledema in both eyes, leading to the diagnosis of Purtscher retinopathy. On the present examination almost a year later, the FA results appeared to be normal, while optical coherence tomography angiography (OCT-A) showed loss of both superficial and deep retinal capillaries in the area corresponding to the visual field deficit (Figure 2).Optical coherence tomography-angiography of the right eye revealing substantially decreased deep retinal vessel density (arrowhead).Automated visual field and microperimetry may quantify the already-known visual dysfunction but would not aid as much as OCT-A in corroborating that the current issue is a sequela of the prior Purtscher occlusive vascular retinopathy.4 Full-field electroretinogram is not thought to be the test of choice in macular dysfunction.Although FA has been the method most often used for imaging retinal and choroidal vessels, the newer OCT-based angiography has been gaining traction, owing to its noninvasive nature. Despite the known limitations of OCT-A, at times it can be superior to classic FA. In 2016, Tokimitsu et al5 demonstrated deep retinal capillary dropout with OCT-A despite rather normal-appearing FA findings in a patient with central scotoma decades after the initial Purtscher retinopathy diagnosis. Three subsequent reports also demonstrated loss of superficial and deep retinal capillaries on OCT-A similar to our case,6-8 stressing the importance of OCT-A in cases of chronic Purtscher retinopathy.The patient was extensively informed about her condition and the prognosis. Progression would be unlikely, given the ischemic nature of the disease. Her visual acuity remained stable at 20/500 OD and 20/30 OS 4 months later and she was advised to follow up in our clinic the next year.
Ophthalmology
A woman in her early 60s presented with an 8-month history of central scotoma and blurry vision in her right eye. Her medical history was significant for a motor vehicle crash that occurred almost a year earlier, with no direct head trauma or eye injury. On examination, her best-corrected visual acuity was 20/500 OD and 20/30 OS. Anterior segment examination was remarkable for pseudophakia on the right eye and mild cataract in the left. Fundus examination was remarkable for a slightly blunted foveal reflex in the right eye. Spectral-domain optical coherence tomography (SD-OCT) showed segmental atrophy of the inner retina (Figure 1A). Fluorescein angiography was unrevealing (Figure 1B).A, Spectral-domain optical coherence tomography of the right eye. B, Fluorescein angiogram of the right eye.
what would you do next?
What would you do next?
Automated visual field
Full-field electroretinogram
Microperimetry
Optical coherence tomography angiography
d
1
0
1
1
female
0
0
62
61-70
null
581
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2719486
A 24-year-old South Asian man presented with acute-onset pain and decreased vision in his left eye. His ocular history was significant for high myopia, a long-standing traumatic retinal detachment in his right eye, and laser-assisted in situ keratomileusis (LASIK) surgery of the left eye 8 months earlier. Three months earlier, the patient experienced an episode of elevated intraocular pressure (IOP) of the left eye, which was treated with travoprost ophthalmic solution, 0.004%, and oral acetazolamide and resolved. The patient’s presenting visual acuity was counting fingers OD (baseline) and 20/200 OS. His initial IOPs were right eye 16 mm Hg and left eye 46 mm Hg by TonoPen. Examination of the left eye indicated normal gonioscopy results and unremarkable anterior and posterior segment examination findings except for the presence of corneal edema without keratic precipitates. The patient started treatment with dorzolamide hydrochloride, timolol maleate, brimonidine tartrate, latanoprost, and oral acetazolamide.The next day, the left eye IOP improved to 28 mm Hg as measured by Goldmann applanation tonometry (GAT), with a clear cornea and return of visual acuity to 20/20. Despite continuing maximum medical therapy for 1 week, the patient’s examination findings remained unchanged. Because of a concern for possible trabeculitis, he also started treatment with topical prednisolone acetate 4 times daily and oral valacyclovir hydrochloride. Two days later, the patient returned to the clinic with decreased vision in his left eye, which showed an IOP measured by GAT of 6 mm Hg, visual acuity of 20/400, and a cornea that was edematous (Figure 1).A, Slitlamp photograph showing a prominent laser-assisted in situ keratomileusis (LASIK) flap in the patient’s left eye. B, A slit beam focused on the cornea reveals a shallow fluid interface between the stromal bed and the LASIK flap (arrowheads).Increase the frequency of prednisolone administration to 6 times per dayRecheck IOP with a handheld tonometer off the LASIK flapReassure the patient and recheck IOP in 1 week What Would You Do Next?
Increase the frequency of prednisolone administration to 6 times per day
Recheck IOP with a handheld tonometer off the LASIK flap
Inject foscarnet sodium intravitreally
Reassure the patient and recheck IOP in 1 week
Interface fluid syndrome
B
Recheck IOP with a handheld tonometer off the LASIK flap
After the low GAT-determined pressure, the patient’s IOP was rechecked with a TonoPen and was found to be 36 mm Hg (choice B). Anterior segment optical coherence tomography revealed a pocket of intrastromal fluid (Figure 2), consistent with interface fluid syndrome (IFS). Given the highly elevated IOP, reassuring the patient and rechecking IOP in 1 week (choice D) would not be appropriate. Increasing the frequency of prednisolone administration (choice A) would not be recommended because it may contribute to a steroid response and exacerbate IFS. Injecting foscarnet intravitreally (choice C) is not indicated given no evidence of posterior pole involvement.Anterior segment optical coherence tomographic image of the patient’s left eye revealing a fluid interface between the stromal bed and the laser-assisted in situ keratomileusis flap (arrowheads).Interface fluid syndrome, a LASIK flap–associated complication first reported by Lyle and Jin in 1999,1 tends to present within the first weeks to months after LASIK surgery but has been reported as late as 9 years later.1-5 The primary mechanism for IFS is related to a high IOP gradient, which causes transudation of aqueous through the endothelium into the stroma.4,6 In LASIK-naive eyes, the transudation of fluid will permeate the entirety of the cornea, resulting in the appearance of microcystic epithelial edema. However, in eyes that have undergone LASIK, the flap interface becomes a potential space for the collection of fluid.7 Elevated IOP tends to be predominantly attributable to a steroid response given its perioperative use after LASIK surgery.1,2,6-9 Because of this factor, the primary therapeutic goal for these patients should be to decrease IOP through the initiation of ocular hypotensive agents and the rapid discontinuation of steroids. However, there are case reports documenting IFS in contexts other than postoperative steroid use, including in a patient who developed IFS after spontaneous IOP increase years after undergoing LASIK,10 similar to the present patient.A feature that can make IFS a confusing entity is that pressure measurements, especially obtained by GAT, can be erroneously low. The low values are because GAT will measure the pressure within the fluid pocket rather than the actual IOP.8 Early reports of IFS describe patients developing end-stage glaucoma because of these misleading pressures.4,5 Although there is no consensus on the best method to measure IOP in these cases, we believe that TonoPen readings in the peripheral cornea, away from the LASIK flap, can provide a more representative pressure measurement.Despite continuation of maximal medical therapy, the patient’s IOP remained elevated and his visual acuity poor because of IFS. Owing to medical insurance issues, the patient elected to undergo urgent glaucoma surgery in his home country. He returned to our clinic for follow-up 3 months later, after undergoing trabeculectomy with an Ologen implant. When not taking medication, the pressure in his left eye was 10 mm Hg, his visual acuity improved to 20/25, the cornea cleared, and there was an elevated superior bleb. Humphrey visual field analysis of the left eye revealed an inferonasal step corresponding to an area of superotemporal thinning on retinal nerve fiber layer analysis. The patient has continued follow-up at regular intervals.
Ophthalmology
A 24-year-old South Asian man presented with acute-onset pain and decreased vision in his left eye. His ocular history was significant for high myopia, a long-standing traumatic retinal detachment in his right eye, and laser-assisted in situ keratomileusis (LASIK) surgery of the left eye 8 months earlier. Three months earlier, the patient experienced an episode of elevated intraocular pressure (IOP) of the left eye, which was treated with travoprost ophthalmic solution, 0.004%, and oral acetazolamide and resolved. The patient’s presenting visual acuity was counting fingers OD (baseline) and 20/200 OS. His initial IOPs were right eye 16 mm Hg and left eye 46 mm Hg by TonoPen. Examination of the left eye indicated normal gonioscopy results and unremarkable anterior and posterior segment examination findings except for the presence of corneal edema without keratic precipitates. The patient started treatment with dorzolamide hydrochloride, timolol maleate, brimonidine tartrate, latanoprost, and oral acetazolamide.The next day, the left eye IOP improved to 28 mm Hg as measured by Goldmann applanation tonometry (GAT), with a clear cornea and return of visual acuity to 20/20. Despite continuing maximum medical therapy for 1 week, the patient’s examination findings remained unchanged. Because of a concern for possible trabeculitis, he also started treatment with topical prednisolone acetate 4 times daily and oral valacyclovir hydrochloride. Two days later, the patient returned to the clinic with decreased vision in his left eye, which showed an IOP measured by GAT of 6 mm Hg, visual acuity of 20/400, and a cornea that was edematous (Figure 1).A, Slitlamp photograph showing a prominent laser-assisted in situ keratomileusis (LASIK) flap in the patient’s left eye. B, A slit beam focused on the cornea reveals a shallow fluid interface between the stromal bed and the LASIK flap (arrowheads).Increase the frequency of prednisolone administration to 6 times per dayRecheck IOP with a handheld tonometer off the LASIK flapReassure the patient and recheck IOP in 1 week
what would you do next?
What would you do next?
Increase the frequency of prednisolone administration to 6 times per day
Inject foscarnet sodium intravitreally
Reassure the patient and recheck IOP in 1 week
Recheck IOP with a handheld tonometer off the LASIK flap
d
0
1
1
1
male
0
0
24
21-30
South Asian
582
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2720486
A 2-year-old white boy was referred for evaluation of a choroidal mass. He had mild periorbital edema and low-grade eye pain in the left eye over 2 weeks prior to presentation. He had completed antibiotic treatment for presumed sinusitis with no improvement. A recent flulike illness over the past 1.5 weeks responded minimally to ibuprofen. Medical and family history were otherwise unremarkable. Magnetic resonance imaging of the orbits revealed an intraocular mass, suspicious for retinoblastoma. The patient was advised consultation by an ocular oncology service.On examination, visual acuity was fix and follow OU. The right eye showed normal findings anteriorly and posteriorly. The left eye revealed mild conjunctival chemosis with periorbital edema. There was trace eyelid edema and mild leukocoria. Dilated ophthalmoscopic examination of the left eye displayed a solid gray mass in the macular region, deep to the retina and with surrounding subretinal fluid (Figure 1A).A, Dilated ophthalmoscopic examination found a gray mass in the macular region of the left eye measuring 12 mm in diameter and 5.1 mm in thickness, with additional subretinal fluid inferiorly. B, B-scan ultrasonography confirmed the fundus mass, dependent subretinal fluid, and echolucency outside the posterior wall of the eye.Examination under anesthesia confirmed the solid mass, measuring 12 mm in diameter and 5.1 mm in thickness and demonstrating echodensity on ultrasonography (Figure 1B). There were no tumor-feeding vessels, and tumor margins were ill defined. Fluorescein angiography demonstrated slight hyperfluorescence of the disc and faint hyperfluorescence of the mass with overlying scattered pinpoint hyperfluorescent leakage. Optical coherence tomography revealed shallow subretinal fluid extending within 3.2 mm of the foveola. What Would You Do Next?
Treat with intra-arterial chemotherapy
Treat with plaque radiotherapy
Treat with oral corticosteroids
Perform fine-needle aspiration biopsy
Posterior scleritis
C
Treat with oral corticosteroids
A fundus mass with overlying subretinal fluid in a child leads to concern for retinoblastoma, choroidal hemangioma, choroidal melanoma, and even inflammatory scleritis. Detailed clinical history, examination, and diagnostic testing can differentiate between the diagnostic possibilities. In this case, despite the presence of leukocoria and a nonpigmented fundus mass in a child, key features ruled out retinoblastoma, such as the lack of retinal involvement and absence of feeder vessels.Important case features included a history of periocular pain, rarely associated with retinoblastoma or choroidal tumors.1 There are uncommon situations in which spontaneous necrosis of retinoblastoma, related neovascular glaucoma, or choroidal melanoma can lead to ocular pain. However, vital diagnostic features, including absence of intralesional calcification on ultrasonography (characteristic of retinoblastoma), lack of double circulation on fluorescein angiography, and lack of acoustic hollowness on ultrasonography (both characteristic of melanoma),1,2 made initiating treatment with intra-arterial chemotherapy for retinoblastoma (choice A) or plaque radiotherapy for choroidal melanoma (choice B) less preferred options. The most revealing and diagnostically suggestive finding was prominent retrobulbar edema in the Tenon space (T sign) on B-scan ultrasonography, characteristic of posterior scleritis.1-7 This is not pathognomonic of scleritis as secondary inflammatory findings can occur with spontaneous necrosis of retinoblastoma and choroidal melanoma. Based on our suspicion for posterior scleritis, the patient was treated with a 5-week course of oral corticosteroids (choice C). More invasive procedures, such as fine-needle aspiration biopsy (choice D), were held in reserve if conservative treatment were to fail. Repeated ultrasonography 9 weeks after presentation showed resolution of the mass and subretinal fluid as well as reduction of periocular edema with T sign (Figure 2).Repeated ultrasonography following oral corticosteroid taper at 9 weeks. Complete resolution of the intraocular mass and fluid, with reduction of Tenon fascia edema (T sign).Posterior scleritis is a vision-threatening scleral inflammatory disease, most commonly diagnosed in adult women.6 Posterior scleritis can be idiopathic or result from infection, trauma, associated systemic disease, or prior ocular surgery.2 Misdiagnosis can occur owing to its rarity and variable clinical presentation.6 Posterior scleritis can manifest as a diffuse type, with thickening of the entire posterior sclera, or as a nodular type, with localized scleral thickening, simulating a solid intraocular mass.1,2,7In children, posterior scleritis is especially uncommon.4,8 Children with posterior scleritis may present with pain, inflammation, and restricted extraocular motility.8 In contrast with the adult population where up to 50% of cases are associated with systemic disease, rarely are pediatric cases found with underlying systemic abnormalities.4,8 Similarly, this patient had a negative rheumatologic evaluation, including antineutrophil cytoplasmic antibody, rheumatoid factor, and HLA B27.Posterior scleritis can be treated with nonsteroidal antiinflammatory drugs, corticosteroids, or immunosuppressive therapy.1-8 Resolution of mass after corticosteroid treatment can more definitively exclude retinoblastoma and choroidal melanoma, which would not decrease in thickness with oral corticosteroids.Given recent systemic illness and periorbital edema, the patient was treated with oral corticosteroids for 5 weeks, using 2 mg/kg of prednisolone per day with slow taper. The mass resolved rapidly with decreased thickness from 5.1 mm to 2.8 mm at 1 week after presentation. By 9 weeks after presentation, the mass and subretinal fluid were completely resolved, leaving trace retinal pigment epithelial mottling. Tenon fascia edema showed mild reduction.
Ophthalmology
A 2-year-old white boy was referred for evaluation of a choroidal mass. He had mild periorbital edema and low-grade eye pain in the left eye over 2 weeks prior to presentation. He had completed antibiotic treatment for presumed sinusitis with no improvement. A recent flulike illness over the past 1.5 weeks responded minimally to ibuprofen. Medical and family history were otherwise unremarkable. Magnetic resonance imaging of the orbits revealed an intraocular mass, suspicious for retinoblastoma. The patient was advised consultation by an ocular oncology service.On examination, visual acuity was fix and follow OU. The right eye showed normal findings anteriorly and posteriorly. The left eye revealed mild conjunctival chemosis with periorbital edema. There was trace eyelid edema and mild leukocoria. Dilated ophthalmoscopic examination of the left eye displayed a solid gray mass in the macular region, deep to the retina and with surrounding subretinal fluid (Figure 1A).A, Dilated ophthalmoscopic examination found a gray mass in the macular region of the left eye measuring 12 mm in diameter and 5.1 mm in thickness, with additional subretinal fluid inferiorly. B, B-scan ultrasonography confirmed the fundus mass, dependent subretinal fluid, and echolucency outside the posterior wall of the eye.Examination under anesthesia confirmed the solid mass, measuring 12 mm in diameter and 5.1 mm in thickness and demonstrating echodensity on ultrasonography (Figure 1B). There were no tumor-feeding vessels, and tumor margins were ill defined. Fluorescein angiography demonstrated slight hyperfluorescence of the disc and faint hyperfluorescence of the mass with overlying scattered pinpoint hyperfluorescent leakage. Optical coherence tomography revealed shallow subretinal fluid extending within 3.2 mm of the foveola.
what would you do next?
What would you do next?
Treat with oral corticosteroids
Treat with intra-arterial chemotherapy
Treat with plaque radiotherapy
Perform fine-needle aspiration biopsy
a
1
1
1
1
male
0
0
2
0-10
White
583
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2718518
A woman in her mid-60s with a history of breast cancer (status post lumpectomy 7 years earlier [N0, M0]) and benign thyroid nodules presented for evaluation of right-sided proptosis. Bulging of the right eye was first noted 6 months earlier elsewhere when she reported progressive proptosis, worsening vision, and tearing in the right eye. She did not report any pain or diplopia.Visual acuity was 20/150 OD (no improvement with pinhole) and 20/20 OS. Pupillary reaction and confrontational visual fields were normal. External examination was notable for proptosis of 7 mm, hypoglobus, and resistance to retropulsion. A firm, nontender mass was palpable in the right superotemporal orbit. The palpebral lobe of the right lacrimal gland was erythematous and prominent compared with the normal-appearing left lacrimal gland. Ocular motility showed −2 abduction deficit, right eye. Slitlamp examination revealed pseudophakia and posterior capsular opacification (right > left), but was otherwise normal. Sensation was normal in the V1, V2 distribution, and there was no preauricular, submandibular, or anterior cervical lymphadenopathy. Computed tomographic scan of the orbits without contrast from an outside facility revealed a spherical mass of unclear cause in the area of the right lacrimal gland. No lytic bone changes or bone remodeling could be seen (Figure 1).Computed tomographic imaging from outside facility revealed a 1.9 × 2.3 × 1.8-cm mass of an unclear cause within the anterolateral right orbit, exerting mass effect on the globe. What Would You Do Next?
Arrange 6-month follow-up
Obtain additional imaging
Perform an incisional biopsy
Perform an excisional biopsy
Solitary fibrous tumor
B
Obtain additional imaging
Progressive proptosis and decreased visual acuity dictate treatment over observation; hence, delaying further evaluation to a 6-month follow-up, would not be appropriate. The outside computed tomographic imaging showed a mass but was nondiagnostic. Without more accurate identification, performing an incisional or excisional biopsy would not be recommended as the next step, as surgical management of orbital masses varies substantially depending on the cause. For example, if benign mixed tumor is a diagnostic consideration, then incisional biopsy is contraindicated. Similarly, for orbital lymphoproliferative disease, attempts at complete excision are rarely warranted. In this case, cavernous malformation, solitary fibrous tumor (SFT), schwannoma, primary lacrimal gland tumor, metastases, lymphoproliferative disease (less likely), and idiopathic orbital inflammation were considered in the differential diagnosis. Cavernous malformation, schwannoma, and SFT are usually managed with complete excision.1-3 For suspected lymphoproliferative disease or metastases, incisional biopsy is typically performed to establish the diagnosis. Additional imaging was needed to refine the differential diagnosis.Subsequent orbital magnetic resonance imaging and magnetic resonance angiography revealed a well-circumscribed extraconal mass within the right superolateral orbit. Extraconal location and rapid filling made a cavernous malformation unlikely.3 A fat plane separated the mass from the lacrimal gland (Figure 2) ruling out a primary lacrimal tumor. There was no evidence of invasion or infiltration of adjacent structures suggestive of metastatic or lymphoproliferative disease. The mass was T2 hypointense and diffusion nonrestricting. Magnetic resonance angiography displayed brisk enhancement during the early venous phase.T1 orbital magnetic resonance imaging revealed imaging features consistent with solitary fibrous tumor: a fat plane (yellow arrowhead) separating the mass from the lacrimal gland and flow void signal absences (white arrowhead) caused by rapid blood flow.These radiologic features can be seen with other orbital neoplasms, such as SFT, hemangiopericytoma, schwannoma, or meningioma,4-6 but were most consistent with SFT or hemangiopericytoma. The patient agreed to a surgical plan for excision of the entire mass through a lateral orbitotomy approach, preferably without bone window.The mass was excised per plan and without complications. Surgical pathologic examination revealed an encapsulated tumor with microscopic invasion of the capsule. A classic patternless pattern of randomly alternating hypocellular and hypercellular areas with collagen bands and ectatic staghorn vessels were consistent with SFT.7,8 Positive stains for tumor markers CD34, CD99, STAT6, and Bcl-2, and negative stain for S100 were also confirmatory.9 In particular, CD34 immunoreactivity has been shown to be a highly sensitive marker for SFTs.10 By contrast, hemangiopericytomas are classically weakly positive for CD34, and other tumors in the differential diagnosis do not exhibit CD34 activity. No atypical mitotic figures were seen.Solitary fibrous tumors are unusual, spindle-cell mesenchymal tumors with fibroblast-like cells. The tumors usually present with mass effect and progressive proptosis, as in this patient. Most are benign and indolent. However, invasion of adjacent tissue, recurrence after partial resection, and malignant transformation have been documented, making complete excision vital. This case demonstrates the value of magnetic resonance imaging and magnetic resonance angiography for accurate preoperative diagnosis and optimal surgical planning of orbital tumors when clinical and radiologic findings are nondiagnostic.Postoperatively, the patient’s proptosis resolved and her visual acuity returned to baseline. Due to the capsular invasion, close surveillance was recommended. Follow-up examination and standard orbital magnetic resonance imaging with contrast were scheduled for 1 year.
Ophthalmology
A woman in her mid-60s with a history of breast cancer (status post lumpectomy 7 years earlier [N0, M0]) and benign thyroid nodules presented for evaluation of right-sided proptosis. Bulging of the right eye was first noted 6 months earlier elsewhere when she reported progressive proptosis, worsening vision, and tearing in the right eye. She did not report any pain or diplopia.Visual acuity was 20/150 OD (no improvement with pinhole) and 20/20 OS. Pupillary reaction and confrontational visual fields were normal. External examination was notable for proptosis of 7 mm, hypoglobus, and resistance to retropulsion. A firm, nontender mass was palpable in the right superotemporal orbit. The palpebral lobe of the right lacrimal gland was erythematous and prominent compared with the normal-appearing left lacrimal gland. Ocular motility showed −2 abduction deficit, right eye. Slitlamp examination revealed pseudophakia and posterior capsular opacification (right > left), but was otherwise normal. Sensation was normal in the V1, V2 distribution, and there was no preauricular, submandibular, or anterior cervical lymphadenopathy. Computed tomographic scan of the orbits without contrast from an outside facility revealed a spherical mass of unclear cause in the area of the right lacrimal gland. No lytic bone changes or bone remodeling could be seen (Figure 1).Computed tomographic imaging from outside facility revealed a 1.9 × 2.3 × 1.8-cm mass of an unclear cause within the anterolateral right orbit, exerting mass effect on the globe.
what would you do next?
What would you do next?
Obtain additional imaging
Perform an excisional biopsy
Arrange 6-month follow-up
Perform an incisional biopsy
a
0
0
1
1
female
0
1
65
61-70
null
584
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2721040
A 35-year-old woman presented with painless ptosis of the left upper eyelid and intermittent diplopia in all gazes (Figure 1). Her new-onset acquired ptosis began 10 months prior. She had no history of ocular disease or trauma, and her medical history was notable for resection of a benign thyroid mass 3 years prior. Family and social history were unremarkable. Her vision was 20/30 OD and 20/25 OS. Intraocular pressures were normal. She had mild anisocoria with the right and left pupils reacting from 6 to 4 mm and from 5 to 3 mm, respectively. The pupils were equal in light and dark, with no reversal after apraclonidine hydrochloride treatment. No relative afferent pupillary defect was noted. Results of the Hertel testing at a base measurement of 100 mm were 20.5 mm OD and 21.0 mm OS. She had full ductions and versions, mild fullness of the left superior sulcus, and no sensory deficits in her facial cranial nerves. Her vertical palpebral fissure measured 10 mm OD and 7 mm OS. Her margin-to-reflex distance was 4 mm OD and 2 mm OS. With phenylephrine hydrochloride, her margin-to-reflex distance became equal at 4 mm OS. She had no lagophthalmos. Results of anterior slitlamp and dilated ophthalmoscopic examinations were unremarkable.Preoperative photograph of the patient showing ptosis of the left upper eyelid.Check myasthenia laboratory findings and commence pyridostigmine trialMagnetic resonance imaging and/or computed tomographic imaging of the orbits What Would You Do Next?
Check myasthenia laboratory findings and commence pyridostigmine trial
Magnetic resonance imaging and/or computed tomographic imaging of the orbits
Müller muscle conjunctival resection
External levator advancement
Intraosseous hemangioma of the frontal bone
B
Magnetic resonance imaging and/or computed tomographic imaging of the orbits
Ptosis is a commonly encountered chief complaint in the ophthalmologist’s clinic. Types of acquired ptosis include aponeurotic ptosis involving slippage of the aponeurosis seen in elderly patients or after trauma, neurogenic ptosis from damage to the oculomotor or sympathetic nerves in the eyelid, myogenic ptosis from dysfunction of the levator muscle due to diseases such as myasthenia gravis, and mechanical ptosis from eyelid tumors or orbital fat prolapse.Unexplained ptosis in a young patient should alert the ophthalmologist because the cause of acquired ptosis will guide management. Aponeurotic ptosis with normal levator function is often corrected by conjunctival resection of the Müller muscle (choice C) provided there is adequate response of the eyelid to the phenylephrine test.1 Other options include the external levator advancement (choice D) or, in the case of poor levator function, the frontalis suspension. However, these choices are not preferred because this patient was young, and the cause was not confirmed to be aponeurotic ptosis. Other causes, such as myasthenia gravis, should be considered. Levator fatigability can be checked by prolonged upgaze for 1 to 2 minutes, followed by an ice pack test for ptosis correction with muscle cooling.2 Acetylcholine receptor antibody is specific, but caution should be exercised before commencing pyridostigmine bromide treatment (choice A), because this patient did not have systemic symptoms, extraocular muscle restriction, or fatigability. Horner syndrome should be excluded by confirming that no anisocoria is present that responds to apraclonidine or cocaine testing. Third nerve palsy should be excluded by confirming normal extraocular motility and pupils.This patient’s atypical presentation of ptosis warranted imaging. With computed tomography and magnetic resonance imaging (MRI) (choice B), her ptosis was determined to be mechanical and secondary to compression of the superior rectus–levator muscle complex by a superior orbital rim tumor, likely an intraosseous hemangioma. These rare proliferations consist of endothelial-lined vascular channels.3-6 Intraosseous hemangiomas most often occur in the vertebral column, but case reports have reported occurrence in orbital bones.4,6,7 Intraosseous hemangiomas can cause mechanical ptosis, proptosis, diplopia, and vision loss.3,6,8 Radiographically, an appearance of an osteolytic sunburst pattern with superior resolution on MRI shows intralesional vascularity.3,7 The differential diagnosis can include fibrous dysplasia, chondroma, meningioma, osteoma, eosinophilic granuloma, and metastatic disease.3,4 Surgical resection is the definitive treatment for lesions causing secondary complications, although for smaller lesions, resection may not be needed.6Computed tomographic imaging and MRI of the orbit showed an enhancing osseous lesion at the superior orbital rim with extension into the superior orbit and mass effect on the superior rectus–levator muscle complex (Figure 2). A combined neurosurgery and oculoplastic procedure was performed, where the tumor was removed via a left-sided craniotomy and the defect was reconstructed with an orbital rim implant. Pathologic evaluation confirmed the intraosseous hemangioma. After resection, she had resolution of diplopia and ptosis by postoperative month 3.Sagittal T1-weighted magnetic resonance imaging fat saturation sequence showing an intraosseous hemangioma at the left superior orbital rim with mild extension into the superior orbit.
Ophthalmology
A 35-year-old woman presented with painless ptosis of the left upper eyelid and intermittent diplopia in all gazes (Figure 1). Her new-onset acquired ptosis began 10 months prior. She had no history of ocular disease or trauma, and her medical history was notable for resection of a benign thyroid mass 3 years prior. Family and social history were unremarkable. Her vision was 20/30 OD and 20/25 OS. Intraocular pressures were normal. She had mild anisocoria with the right and left pupils reacting from 6 to 4 mm and from 5 to 3 mm, respectively. The pupils were equal in light and dark, with no reversal after apraclonidine hydrochloride treatment. No relative afferent pupillary defect was noted. Results of the Hertel testing at a base measurement of 100 mm were 20.5 mm OD and 21.0 mm OS. She had full ductions and versions, mild fullness of the left superior sulcus, and no sensory deficits in her facial cranial nerves. Her vertical palpebral fissure measured 10 mm OD and 7 mm OS. Her margin-to-reflex distance was 4 mm OD and 2 mm OS. With phenylephrine hydrochloride, her margin-to-reflex distance became equal at 4 mm OS. She had no lagophthalmos. Results of anterior slitlamp and dilated ophthalmoscopic examinations were unremarkable.Preoperative photograph of the patient showing ptosis of the left upper eyelid.Check myasthenia laboratory findings and commence pyridostigmine trialMagnetic resonance imaging and/or computed tomographic imaging of the orbits
what would you do next?
What would you do next?
Check myasthenia laboratory findings and commence pyridostigmine trial
Magnetic resonance imaging and/or computed tomographic imaging of the orbits
Müller muscle conjunctival resection
External levator advancement
b
0
0
0
1
female
0
0
35
31-40
null
585
original
https://jamanetwork.com/journals/jamaoncology/fullarticle/2713851
A middle-aged man with a 25-pack-year history of cigarette smoking was in his usual state of health when he developed excessive coughing with hemoptysis, which spontaneously resolved. Two months later, he presented to a hospital with complaint of shortness of breath, cough, and chest pain. A computed tomographic image of the chest (Figure 1) identified a large, 12-cm left upper lobe mass abutting the anterior chest wall pleura and extending into the mediastinum, with mediastinal lymphadenopathy as well as several liver lesions. A biopsy revealed poorly differentiated carcinoma, consistent with a primary tumor in the lung. On presentation to our multidisciplinary clinic, he reported constant, bothersome sweating of the left forehead. On physical examination, a well-demarcated area of erythematous, perspiring skin on the left forehead was noted.A, Coronal computed tomographic image of the thorax with intravenous contrast at initial presentation, demonstrating a left upper lobe mass; asterisk depicts possible area of sympathetic nerve compression or invasion. B, Radiation planning computed tomographic image taken 2 weeks after initial presentation, demonstrating growth of the left upper lobe tumor. The red contour identifies the tumor; the orange contour, the area of sympathetic trunk compression; the yellow contour, the trachea; the teal contour, the left lung; the purple contour, the esophagus; the peach contour, the first rib. Which Lesion Could Explain This Patient’s Symptoms?
C7 cervical spinal cord compression
Impingement of the left sympathetic trunk
Right cervical ganglion invasion
Transection of the left sympathetic trunk
Apical non–small cell lung cancer with sympathetic nerve involvement
A
C7 cervical spinal cord compression
Apical lung cancers with sympathetic nerve involvement may manifest with Horner’s syndrome, the classic triad of miosis, ptosis, and anhidrosis.1 Eccrine sweat function and thermoregulation of the face is modulated by the sympathetic nervous system via 3-neuron autonomic relays. The axons of first-order neurons within the hypothalamus project through nerve tracts of the brainstem and the lateral gray column of the spinal cord.2 The preganglionic nerve fibers from T1 through T4 spinal cord levels join the sympathetic trunk, traveling in close proximity to the apex of the lung before synapsing in the superior cervical ganglion.2 Postganglionic fibers from third-order neurons then form a nerve plexus and surround the common carotid and downstream vessels to supply sympathetic innervation to the head and neck.2Although absence of function, as seen in Horner’s syndrome, is more commonly reported,1 sympathetic hyperactivity may be a sequela of injury to autonomic nerves as they traverse the thoracic cavity. Hyperhidrosis caused by a comorbid condition, also known as secondary hyperhidrosis, has been the focus of about 3 dozen case reports over the past 70 years.3 Most commonly, these findings have been reported in the context of mesothelioma,4,5 often affecting multiple dermatomes and usually involving the thorax.4,6 Symptoms are typically reported as occurring ipsilateral to the tumor7; however, contralateral dysfunction has also been described.6 In a case report detailed by Walsh et al,8 the pathophysiology of the hyperhidrosis in a patient with an apical lung tumor with ipsilateral hyperhidrosis of the face, arm, and antecubital fossa was postulated to be because of invasion of sympathetic nerves leading to irritation of the nerve fibers and increased signaling. Furthermore, in a patient with a primary central nervous system tumor compressing the thoracic spinal cord and ipsilateral hemifacial hyperhidrosis that resolved after decompression, Kilinçer et al9 attributed the patient’s symptoms to irritation of sympathetic nerves. (A schematic is provided in Figure 2.)Proposed mechanism of hemifacial hyperhidrosis owing to compression of the sympathetic trunk.Clinical workup and management strategies for patients with hyperhidrosis from intrathoracic malignant conditions have been described. Hyperhidrosis may be evaluated though sweat testing using the 1-step iodine-starch method, which demarks perspiring skin using a colorimetric indicator.10 Radiation therapy has been reported to be successful in abating symptoms in some patients.3,8 Interestingly, in mesothelioma, hyperhidrosis has been proposed to be a prognostic indicator, suggesting poor outcomes in patients with this complaint.8The patient presented here was diagnosed with a stage IV, T4N3M1c adenocarcinoma of the left upper lobe of the lung metastatic to the liver. The primary tumor was a rapidly progressing left apical lung cancer with associated flushing and hyperhidrosis limited to the left forehead. On review of the literature, we propose that his symptoms were the result of irritation of the second-order neuron of the left sympathetic trunk prior to synapse in the superior cervical ganglion. We suspect this injury may have occurred through compression by the enlarging tumor or even direct nerve invasion. Because the patient had a limited area of involvement, with seemingly normal sympathetic tone in the lower face and neck, we presume the tumor only affected a portion of the nerve.The patient was started on a palliative course of radiation for his complaint of chest pain with plans for subsequent chemotherapy. He died suddenly 1 week later.
Oncology
A middle-aged man with a 25-pack-year history of cigarette smoking was in his usual state of health when he developed excessive coughing with hemoptysis, which spontaneously resolved. Two months later, he presented to a hospital with complaint of shortness of breath, cough, and chest pain. A computed tomographic image of the chest (Figure 1) identified a large, 12-cm left upper lobe mass abutting the anterior chest wall pleura and extending into the mediastinum, with mediastinal lymphadenopathy as well as several liver lesions. A biopsy revealed poorly differentiated carcinoma, consistent with a primary tumor in the lung. On presentation to our multidisciplinary clinic, he reported constant, bothersome sweating of the left forehead. On physical examination, a well-demarcated area of erythematous, perspiring skin on the left forehead was noted.A, Coronal computed tomographic image of the thorax with intravenous contrast at initial presentation, demonstrating a left upper lobe mass; asterisk depicts possible area of sympathetic nerve compression or invasion. B, Radiation planning computed tomographic image taken 2 weeks after initial presentation, demonstrating growth of the left upper lobe tumor. The red contour identifies the tumor; the orange contour, the area of sympathetic trunk compression; the yellow contour, the trachea; the teal contour, the left lung; the purple contour, the esophagus; the peach contour, the first rib.
which lesion could explain this patient’s symptoms?
Which lesion could explain this patients symptoms?
Impingement of the left sympathetic trunk
Transection of the left sympathetic trunk
Right cervical ganglion invasion
C7 cervical spinal cord compression
d
0
1
1
1
male
0
0
45
41-50
null
586
original
https://jamanetwork.com/journals/jamacardiology/fullarticle/2711637
A woman in her late 40s with a history of hypertension presented to the emergency department after multiple episodes of palpitations with near syncope. While in the emergency department, she developed monomorphic ventricular tachycardia (VT) with hemodynamic instability and was successfully cardioverted. She continued to have nonsustained monomorphic VT, so intravenous amiodarone and oral metoprolol were initiated. She was admitted for further evaluation. Results of tests of electrolyte levels and coronary angiography were normal. Cardiac magnetic resonance imaging with gadolinium contrast revealed normal-sized cardiac chambers and normal biventricular function without delayed enhancement. The presenting electrocardiogram (ECG) is shown in Figure 1.Discharge with oral antiarrhythmic therapy with a class I or III agentDischarge with oral β-blocker or nondihydropyridine calcium channel blocker What Would You Do Next?
Placement of an implantable cardioverter-defibrillator
Electrophysiology study with possible catheter ablation
Discharge with oral antiarrhythmic therapy with a class I or III agent
Discharge with oral β-blocker or nondihydropyridine calcium channel blocker
Outflow tract ventricular tachycardia
B
Electrophysiology study with possible catheter ablation
This patient presented with recurrent symptomatic monomorphic VT suggestive of an outflow tract (OT) origin. An electrophysiology (EP) study to anatomically localize the abnormal rhythm and ablation therapy is the preferred strategy in severely symptomatic patients. Medical therapy may be considered; owing to limited and varying efficacy of pharmacologic therapy and its adverse effects, an EP study with ablation, with its high success rate and low complication rate, is preferred. Outflow tract VT (OTVT) is a monomorphic subset of idiopathic VT diagnosed in patients without underlying structural heart disease, metabolic abnormalities, or cardiac channelopathies. Right ventricular OT (RVOT) VT makes up about 80% of OTVT cases, with most originating just inferior to the pulmonic valve from the anterior and superior septal aspects of the RVOT. The other 20% are left ventricular OT (LVOT) VT, which commonly arises from the region of the aortic cusps in the aortic root.1 A surface ECG with an inferior axis, left bundle branch block (BBB) morphology, and precordial R-wave transition at V4 or after suggests RVOT VT, while one with an inferior axis, right or left BBB morphology, and precordial R-wave transition at V2 or earlier suggests LVOT VT.2 Despite surface ECG findings that may differentiate LVOT from RVOT, definitive localization is via electroanatomic mapping in an EP study.Presenting ECGs (Figure 1) showed runs of a monomorphic wide complex tachycardia interceded by normal sinus rhythm (with a normal axis and narrow QRS complexes). As the wide complex monomorphic tachycardia rhythm developed, a new left BBB morphology with an inferior axis (positive QRS deflections in inferior leads II, III, and aVF) and a precordial R-wave transition (a ratio R-wave to S-wave amplitude of ≥1) at lead V3 were observed (Figure 2).Patient electrocardiogram on development of a wide complex monomorphic tachycardia rhythm. Note positive QRS deflections in the inferior leads II, III, and aVF, indicating a new left branch bundle block morphology with an inferior axis and a precordial R-wave transition at lead V3.Management of OTVT is dictated by symptom frequency and severity and can include medical therapy and/or catheter ablation. Acute termination can be achieved by vagal maneuvers and abortive drugs (eg, adenosine, β-blockers, verapamil). Preventive medications include class IA, IC, and III antiarrhythmic drugs; β-blockers; and verapamil. Radiofrequency catheter ablation can be used early in management (with success rates >90% and recurrence of 5% in the first year in RVOT3,4); indications include severe symptoms or ones refractory to medical therapy, cardiomyopathy, or younger age and desire to avoid long-term medical therapy.5 Idiopathic VT has a favorable prognosis,6 but symptoms can decrease quality of life, and patients may be at risk for cardiomyopathy if premature ventricular contraction (PVC) burden is high.7The patient had sufficient PVCs for activation mapping with a CARTO 3 advanced 3-dimensional mapping system (Biosense-Webster). The origin was localized to the LVOT at the junction between the right and noncoronary cusps of the aortic root. Catheter ablation with radiofrequency energy was performed successfully; VT was noninducible at the end of the EP study, and the patient had no recurrence of VT or PVCs in the next 24 hours per telemetry.
Cardiology
A woman in her late 40s with a history of hypertension presented to the emergency department after multiple episodes of palpitations with near syncope. While in the emergency department, she developed monomorphic ventricular tachycardia (VT) with hemodynamic instability and was successfully cardioverted. She continued to have nonsustained monomorphic VT, so intravenous amiodarone and oral metoprolol were initiated. She was admitted for further evaluation. Results of tests of electrolyte levels and coronary angiography were normal. Cardiac magnetic resonance imaging with gadolinium contrast revealed normal-sized cardiac chambers and normal biventricular function without delayed enhancement. The presenting electrocardiogram (ECG) is shown in Figure 1.Discharge with oral antiarrhythmic therapy with a class I or III agentDischarge with oral β-blocker or nondihydropyridine calcium channel blocker
what would you do next?
What would you do next?
Discharge with oral antiarrhythmic therapy with a class I or III agent
Electrophysiology study with possible catheter ablation
Discharge with oral β-blocker or nondihydropyridine calcium channel blocker
Placement of an implantable cardioverter-defibrillator
b
1
0
1
1
female
0
0
48
41-50
null
587
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2723083
A previously healthy 18-year-old woman presented with a growth in the right oropharynx of 4 months’ duration. At the time of presentation, it had begun to cause changes in her voice, globus sensation, and dysphagia. The patient reported a 20-pound weight loss over the prior 3 months and decreased appetite. She denied any associated difficulty breathing, pain, fever, chills, rash, or night sweats. She was a nonsmoker and nondrinker.On examination, she appeared well. Despite a muffled voice, she was not in respiratory distress. Fiberoptic laryngoscopy revealed a large, partially obstructing mass in the right oropharynx protruding from the right tonsil. It descended inferiorly and was partially compressing the epiglottis. The true and false vocal cords, although partially obstructed, appeared normal and mobile. The lesion was without ulceration, exudate, or erythema. The contralateral tonsil was small and normal in appearance. The remainder of her physical examination findings were unremarkable, including no neck masses.The patient agreed to surgical excision of the mass, including tonsillectomy. Her airway was secured via an awake transoral fiberoptic intubation without difficulty. The planned procedure occurred without difficulty or complication. The specimen excised was a 6 × 3 × 2-cm mass continuous with the right tonsil (Figure 1), without involvement of any surrounding structures.A, Intraoperative photograph of a large, irregularly shaped lesion protruding from the patient’s right tonsil. B, The excised specimen measured 6 × 3 × 2 cm and was continuous with the right tonsil. What Is Your Diagnosis?
Squamous cell carcinoma
Hemangioma
Mixed lymphatic and venous malformation
Non-Hodgkin lymphoma
C. Mixed lymphatic and venous malformation
C
Mixed lymphatic and venous malformation
Surgical pathologic analysis determined that the lesion was a mixed lymphatic and venous malformation. The lesion had large, macrocystic channels mostly devoid of red blood cells (Figure 2A and B). Occasional valvelike structures were noted, consistent with both lymphatic and venous features (Figure 2C and D). It was also noted that many of these vascular structures were associated with smooth muscle, a characteristic more closely associated with veins. Immunohistochemical analysis of the lesional specimen demonstrated strongly positive CD-31 staining (Figure 2E), an endothelial cell marker, and weak to strongly positive D2-40 (podoplanin) staining, an endothelial cell marker specific to lymphatics.1 It was noted that both D2-40–positive and D2-40–negative structures were present in the specimen, suggesting a mixed lymphatic and venous origin of the lesion (Figure 2F). There was no evidence of lymphoma within the specimen.A-D, Hematoxylin-eosin–stained lesional specimens. A and B, Representative low-magnification photomicrographs demonstrate a mixture of vessels and lymphoid follicles. C, Higher magnification demonstrates a vessel with valves. D, Even higher magnification demonstrates vessels. E, Immunohistochemical analysis demonstrates positive staining to CD-31. F, Immunohistochemical analysis demonstrates a mixture of positive and negative staining of vascular structures to D2-40.To our knowledge, this case is unique in that it involves a mixed lymphatic-venous malformation of the palatine tonsil. Lymphatic-venous malformations, as described by the International Society for the Study of Vascular Anomalies (ISSVA),2 resemble both vascular types and exhibit mixed characteristics. The lymphatic vascular system develops as diverticula from the developing venous system around the fifth week embryologically,3 and any aberrant structure created in this process can manifest as either a lymphatic malformation or a mixed lymphatic-venous malformation.Another rare but more commonly reported anomaly of the palatine tonsil is a lymphatic malformation. These occur in only 1 of 2000 to 4000 live births and most commonly involve the head and neck.4,5 While most arise in the skin, they have been reported to develop in the larynx, parotid gland, mouth, and tongue.6 Lymphatic malformation arising from the tonsil is a rare occurrence.In a review of 32 cases of tonsillar lymphatic malformations, Mardekian and Karp7 found that clinical presentation was often associated with dysphagia, dysphonia, sore throat, and foreign body sensation. Patients in this study ranged in age from 3 to 80 years, with a median age of 22.5 years. Lesions grossly appeared pedunculated or polypoid and were described as smooth, firm, and with intact overlying mucosa.7 The lesions evaluated ranged from 0.5 to 5.0 cm, and nearly all cases were unilateral palatine tonsillar lesions. Histologically, 3 common components of these lesions were identified: dilated lymphatic vessels, a fibrous or adipose underlying stroma, and associated lymphoid stromal components. Although we found no descriptions of combined lymphatic-venous malformation arising from the tonsil in the literature, the presentation of the lesion in the present case, except for its large size (6 cm), is markedly similar to the description by Mardekian and Karp7 of tonsillar lymphatic malformations.The presentation of this patient with painless and progressing tonsillar growth and associated weight loss necessitated the ruling out of malignant neoplasm, particularly lymphoma. In the case of tonsillar lymphatic malformation, including lymphatic-venous malformation, surgical excision is curative with the exception of potential for local recurrence. In the present case, complete resection of the lesion was achieved and the patient remained without recurrence at 2-year follow-up.
General
A previously healthy 18-year-old woman presented with a growth in the right oropharynx of 4 months’ duration. At the time of presentation, it had begun to cause changes in her voice, globus sensation, and dysphagia. The patient reported a 20-pound weight loss over the prior 3 months and decreased appetite. She denied any associated difficulty breathing, pain, fever, chills, rash, or night sweats. She was a nonsmoker and nondrinker.On examination, she appeared well. Despite a muffled voice, she was not in respiratory distress. Fiberoptic laryngoscopy revealed a large, partially obstructing mass in the right oropharynx protruding from the right tonsil. It descended inferiorly and was partially compressing the epiglottis. The true and false vocal cords, although partially obstructed, appeared normal and mobile. The lesion was without ulceration, exudate, or erythema. The contralateral tonsil was small and normal in appearance. The remainder of her physical examination findings were unremarkable, including no neck masses.The patient agreed to surgical excision of the mass, including tonsillectomy. Her airway was secured via an awake transoral fiberoptic intubation without difficulty. The planned procedure occurred without difficulty or complication. The specimen excised was a 6 × 3 × 2-cm mass continuous with the right tonsil (Figure 1), without involvement of any surrounding structures.A, Intraoperative photograph of a large, irregularly shaped lesion protruding from the patient’s right tonsil. B, The excised specimen measured 6 × 3 × 2 cm and was continuous with the right tonsil.
what is your diagnosis?
What is your diagnosis?
Mixed lymphatic and venous malformation
Squamous cell carcinoma
Hemangioma
Non-Hodgkin lymphoma
a
0
1
1
1
female
0
0
18
11-20
null
588
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2718634
A healthy man in his mid 30s residing in the northeastern United States was seen by his primary care physician in mid-June with a 2-week history of flulike symptoms. He was empirically started on doxycycline because he reported frequenting wooded areas.One week later, he developed a complete right facial nerve paralysis. He was started on a course of prednisone, with subsequent full recovery of facial nerve function. About 2 weeks later, he reported new-onset right-sided hearing loss, tinnitus, and intermittent otalgia. He was evaluated in the otology clinic approximately 3 weeks after the onset of these otologic symptoms. He had no prior otologic history and no history of noise exposure. His otologic and head and neck examination findings were unremarkable.Comprehensive audiometry was performed, which showed a mild sensorineural hearing loss from 3000 to 8000 Hz in the right ear and a mild sensorineural hearing loss at 8000 Hz only in the left ear. Given the asymmetry in the right ear, magnetic resonance imaging (MRI) of the brain and internal auditory canals (IACs) with contrast was obtained. There was abnormal linear and smooth enhancement of the right facial nerve in the superior distal IAC, labyrinthine segment, and geniculate ganglion. Discrete enhancement was also seen within the inferior aspect of the right IAC in the region of the vestibulocochlear nerve (Figure 1).Initial magnetic resonance imaging. T1-weighted postcontrast axial (A) and coronal (B) magnetic resonance imaging of the brain and internal auditory canals. A, Increased enhancement of the labyrinthine segment of the facial nerve (yellow arrowhead) and geniculate ganglion (pink arrowhead). B, Discrete linear and smooth areas of enhancement in the superior distal portion of the right internal auditory canal and the inferior aspect of the right internal auditory canal, corresponding to the facial nerve (pink arrowhead) and the vestibulocochlear nerve (yellow arrowhead), respectively. What Is Your Diagnosis?
Vestibular schwannoma
Idiopathic sudden sensorineural hearing loss
Neuroborreliosis cranial polyneuritis
Facial nerve schwannoma
C. Neuroborreliosis cranial polyneuritis
C
Neuroborreliosis cranial polyneuritis
Lyme disease, or Lyme borreliosis, caused by the spirochete Borrelia burgdorferi, is the most common vector-borne disease in the United States, seen particularly in the coastal Northeast, upper Midwest, and West.1,2 It is a multisystem disease transmitted to humans by infected ticks of the Ixodes species. Otolaryngologic manifestations, including neck pain and stiffness, sore throat, dizziness, lymphadenopathy, otalgia, tinnitus, cranial nerve neuritis, and hearing loss, affect up to 75% of patients.3 In this patient, the diagnosis of Lyme disease was confirmed in a standard fashion by a positive IgM immunoblot, which showed reactivity to 23-, 39-, and 41-kDa proteins. Lyme disease typically manifests in 3 clinical stages of early localized disease, early disseminated disease, and late disease.Early localized disease (days to weeks) is characterized by erythema migrans at the site of the tick bite and generalized flulike symptoms. If untreated, early disseminated disease (weeks to months) can occur with various clinical manifestations, including early neuroborreliosis (3%-16%).1 Patients may not recall a tick bite or the erythema migrans rash, which can be absent in 20% to 50% of cases,4 and the first recognized symptoms may be those of early disseminated disease. Late disease (months to years) is due to persistent infection and/or irreversible tissue damage from host inflammatory reactions.1The mechanisms thought to be responsible for the manifestations of Lyme disease are (1) direct invasion by the spirochete, (2) host immunological response, and (3) vasculitis.3 Early neuroborreliosis, which typically includes lymphocytic meningitis, cranial neuritis, and painful radiculitis, is thought to be inflammatory in nature involving the subarachnoid space and perineural tissue.5 The facial nerve is the most commonly affected cranial nerve, although vestibulocochlear nerve involvement has been reported, with the cochlear nerve being more affected.3 Hearing loss has been reported in 1.5% to 44% of patients diagnosed as having Lyme disease.3 Conversely, the prevalence of Lyme disease among patients seen with sudden sensorineural hearing loss ranges from 0% to 21.3%.6 In these patients, routine serologic testing for Borrelia should be limited to those residing in endemic regions.6In the presence of an asymmetrical sensorineural hearing loss, imaging studies are indicated to rule out retrocochlear pathology, such as vestibular schwannoma, facial nerve schwannoma, and inflammatory processes. While imaging findings are uncommon in patients with Lyme disease, some authors, including Agarwal and Sze,2 have described the inflammatory response reflected in several different MRI findings, such as gadolinium enhancement of the meninges and cranial nerves.7-9 However, imaging findings do not always correlate with the presence or degree of clinical manifestations, and clinically “silent” enhancement of cranial nerves has been reported.7Abnormal radiographic findings may resolve along with, or after, clinical symptoms.8,9 In this patient, the initial MRI was obtained after the full recovery of facial nerve function, but there was persistent enhancement of the facial nerve. A repeated MRI was obtained 6 months after symptom onset, and the previous increased enhancement of the right facial nerve was diminished, with only normal physiologic enhancement remaining. Comprehensive audiometry was repeated, showing hearing improvement and resolution of the right-sided hearing asymmetry. Also, on repeated MRI, the previous enhancement of the vestibulocochlear nerve in the inferior aspect of the right IAC had resolved (Figure 2).Repeated magnetic resonance imaging (MRI). T1-weighted postcontrast coronal MRI of the brain and internal auditory canals. Previous enhancement of the right facial nerve is significantly reduced (pink arrowhead), with only normal physiologic enhancement remaining. There is no remaining enhancement of the vestibulocochlear nerve in the inferior aspect of the right internal auditory canal (yellow arrowhead).No consensus data are available regarding the expected hearing outcome in patients with hearing loss related to Lyme disease.3 However, reports exist of complete clinical recovery with antibiotic treatment,10 as well as patients with poor recovery of hearing.6 In addition, some authors have found no significant difference in audiologic outcome between seropositive patients treated with antibiotics and seronegative patients with other causes of sudden sensorineural hearing loss.3 Nonetheless, it is generally agreed that treatment with antibiotics, such as doxycycline and ceftriaxone, should be given at an early stage to shorten recovery time and prevent the development of late disease complications.5
General
A healthy man in his mid 30s residing in the northeastern United States was seen by his primary care physician in mid-June with a 2-week history of flulike symptoms. He was empirically started on doxycycline because he reported frequenting wooded areas.One week later, he developed a complete right facial nerve paralysis. He was started on a course of prednisone, with subsequent full recovery of facial nerve function. About 2 weeks later, he reported new-onset right-sided hearing loss, tinnitus, and intermittent otalgia. He was evaluated in the otology clinic approximately 3 weeks after the onset of these otologic symptoms. He had no prior otologic history and no history of noise exposure. His otologic and head and neck examination findings were unremarkable.Comprehensive audiometry was performed, which showed a mild sensorineural hearing loss from 3000 to 8000 Hz in the right ear and a mild sensorineural hearing loss at 8000 Hz only in the left ear. Given the asymmetry in the right ear, magnetic resonance imaging (MRI) of the brain and internal auditory canals (IACs) with contrast was obtained. There was abnormal linear and smooth enhancement of the right facial nerve in the superior distal IAC, labyrinthine segment, and geniculate ganglion. Discrete enhancement was also seen within the inferior aspect of the right IAC in the region of the vestibulocochlear nerve (Figure 1).Initial magnetic resonance imaging. T1-weighted postcontrast axial (A) and coronal (B) magnetic resonance imaging of the brain and internal auditory canals. A, Increased enhancement of the labyrinthine segment of the facial nerve (yellow arrowhead) and geniculate ganglion (pink arrowhead). B, Discrete linear and smooth areas of enhancement in the superior distal portion of the right internal auditory canal and the inferior aspect of the right internal auditory canal, corresponding to the facial nerve (pink arrowhead) and the vestibulocochlear nerve (yellow arrowhead), respectively.
what is your diagnosis?
What is your diagnosis?
Facial nerve schwannoma
Idiopathic sudden sensorineural hearing loss
Vestibular schwannoma
Neuroborreliosis cranial polyneuritis
d
1
1
0
1
male
0
0
30
21-30
null
589
original
https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2719200
A 1-week-old, full-term male newborn presented with increasingly labored breathing, xerostomia, and poor bowel movements. He had been born after an uncomplicated pregnancy, during which routine prenatal ultrasonography had not detected any anatomical abnormalities or lesions. On physical examination, there were no abnormalities other than symmetrical submucosal swelling of the floor of the mouth and a midline external punctum present in the submental area (Figure, A). The tongue was displaced posteriorly and superiorly. Contrast-enhanced magnetic resonance imaging of the neck and face performed shortly after presentation showed a midline sublingual lesion extending inferiorly through the mylohyoid muscle (Figure, B-D). The intrinsic muscles of the tongue appeared minimally displaced without obvious infiltration; however, the genioglossus muscle was difficult to locate relative to the mass. A thin-walled, multiloculated lesion was identified, measuring approximately 2.0 × 2.5 × 3.2 cm. The mass was bright on T2-weighted imaging and dark on T1-weighted imaging and did not enhance with intravenous gadolinium. There were no obvious fluid-fluid levels or calcifications. When in-phase and out-of-phase images were compared, there was no significant signal intensity drop, suggesting an absence of fat within the lesion.A, Appearance of the submental neck. B, Coronal short tau inversion recovery (STIR) T2-weighted magnetic resonance image without contrast enhancement. C, Coronal T1-weighted, fat-saturated magnetic resonance image after contrast enhancement. D, Sagittal T1-weighted non–fat-saturated magnetic resonance image after contrast enhancement. What Is Your Diagnosis?
Macrocystic lymphatic malformation
Cystic teratoma (mature)
Foregut duplication cyst
Dermoid cyst
B. Cystic teratoma (mature)
B
Cystic teratoma (mature)
Teratomas are congenital tumors composed of tissue derived from all 3 embryonic germ cell layers and occur in approximately 1 in 20 000 to 40 000 live births.1,2 Pluripotent cells are believed to deviate during the fourth to fifth gestational week and differentiate to varying degrees, categorizing as immature or mature, with solid to cystic architecture.2,3 Teratomas are commonly located in the sacrococcyx but may present in the gonads, anterior mediastinum, or retroperitoneum.1 Head and neck, or cervical, teratomas account for 3% to 5% of all pediatric teratomas. They typically present in the midline or anterolateral aspect of the neck or the oropharynx and frequently displace or splay the thyroid gland.1,2,4 Most cervical teratomas are mature and contain well-differentiated tissue.4 Clinically, they are generally benign; however, if left untreated, airway obstruction raises mortality to as high as 80% to 100%.1 The urgency of intervention depends on the degree of airway compromise; although intubation may be difficult, tracheostomy is usually not necessary.5 The predominant type of cervical teratoma shifts after the first decade of life, when immature variants, less predictability, and malignant characteristics become more prevalent.1,4,5Surgical excision is considered the criterion standard treatment.2,3 Completely extracranial teratomas are usually treated with surgery alone.1 Adjuvant chemotherapy is used for disseminated, invasive, or residual tumors. In cases of recurrence, chemotherapy is associated with a tumor-free survival rate as high as 98.6%.1 Sclerotherapy, although effective in the more common lymphatic malformations (cystic hygromas), does not show the same effectiveness in cystic teratomas.4-6Because a cystic teratoma and a lymphatic malformation require different interventions, distinguishing them is important, although sometimes virtually impossible. Both lesions may take on the appearance of unilocular or multilocular cystic masses on imaging, with no significant drop in signal intensity.2,5,6 Although comparable in size, gross characteristics, and anatomical origin, a teratoma is more likely to present with calcifications or fatty tissue within the mass.1,3,6 A key characteristic of macrocystic lymphatic malformations is the common finding of fluid-fluid levels, which indicates layering of proteinaceous debris or old blood products within the endothelium-lined macrocysts.6 However, a lack of fluid-fluid levels does not rule out lymphatic malformation. In cases in which it is not possible to differentiate a cystic teratoma from a lymphatic malformation, tissue sampling or excisional biopsy may be needed to confirm the diagnosis.Foregut duplication cysts are simple cysts that contain intramural smooth muscle, attachment to a foregut structure, and foregut-derived epithelium, mucosa, or both. These cysts have a prevalence of 1 in 4500 cases, 0.3% of which are in the head and neck, most commonly the oral tongue.7 On magnetic resonance imaging, the protein content leads to variable signal intensity on T1-weighted images, and cyst wall enhancement by contrast is variable. Foregut duplication cysts are bright on T2-weighted images and typically show a single, thick-walled cyst without septations. Surgical excision is the treatment of choice because serial needle aspiration, marsupialization, and sclerotherapy may not be effective.7Dermoid cysts result from ectoderm and mesoderm sequestration during the midline union of the embryonic first and second branchial arches.8,9 Approximately 7% of dermoid cysts are found in the head and neck, primarily in the supraorbital region, followed by floor of mouth, nose, orbit, and cheek.8 A true dermoid cyst has a stratified, keratinized epithelial lining with skin appendages and may have a “sack-of-marbles” appearance on magnetic resonance imaging. When such a lesion presents, fatty components, often collected in nodules, appear isointense on T1-weighted images and hypointense on T2-weighted images. Non–contrast-enhanced computed tomography shows calcified intracystic free corpuscles.10At 2 weeks of age, this patient underwent resection via a combined approach: externally through a submental incision and transorally via a median glossotomy (tongue split) to remove the sublingual component. The mass was removed en bloc without spillage of contents. Results of pathology studies were consistent with a mature cystic teratoma. The patient spent 5 days in the hospital for postoperative feeding support and was ultimately discharged home and tolerated a normal formula diet. He has been followed up for 5 years, with no evidence of recurrence on serial surveillance imaging.
General
A 1-week-old, full-term male newborn presented with increasingly labored breathing, xerostomia, and poor bowel movements. He had been born after an uncomplicated pregnancy, during which routine prenatal ultrasonography had not detected any anatomical abnormalities or lesions. On physical examination, there were no abnormalities other than symmetrical submucosal swelling of the floor of the mouth and a midline external punctum present in the submental area (Figure, A). The tongue was displaced posteriorly and superiorly. Contrast-enhanced magnetic resonance imaging of the neck and face performed shortly after presentation showed a midline sublingual lesion extending inferiorly through the mylohyoid muscle (Figure, B-D). The intrinsic muscles of the tongue appeared minimally displaced without obvious infiltration; however, the genioglossus muscle was difficult to locate relative to the mass. A thin-walled, multiloculated lesion was identified, measuring approximately 2.0 × 2.5 × 3.2 cm. The mass was bright on T2-weighted imaging and dark on T1-weighted imaging and did not enhance with intravenous gadolinium. There were no obvious fluid-fluid levels or calcifications. When in-phase and out-of-phase images were compared, there was no significant signal intensity drop, suggesting an absence of fat within the lesion.A, Appearance of the submental neck. B, Coronal short tau inversion recovery (STIR) T2-weighted magnetic resonance image without contrast enhancement. C, Coronal T1-weighted, fat-saturated magnetic resonance image after contrast enhancement. D, Sagittal T1-weighted non–fat-saturated magnetic resonance image after contrast enhancement.
what is your diagnosis?
What is your diagnosis?
Macrocystic lymphatic malformation
Dermoid cyst
Cystic teratoma (mature)
Foregut duplication cyst
c
1
0
0
1
male
0
0
0.02
0-10
null
590
original
https://jamanetwork.com/journals/jama/fullarticle/2723632
A 5-year-old girl was referred to a pediatric gastroenterology clinic for chronic constipation and poor weight gain. During her first week of life, she developed diarrhea and vomiting. With initiation of solid food, she developed laxative-dependent constipation. She underwent newborn genetic screening before routine cystic fibrosis (CF) screening. Results were normal. At the time of presentation to the gastroenterology clinic, she had no respiratory symptoms. Results of anorectal manometry, spinal magnetic resonance imaging, and thyroid studies were normal. Family history included constipation in a sister and a great aunt with CF. Her body mass index (BMI) was below the third percentile (eFigureA in the Supplement). Physical examination findings were unremarkable, including normal respiratory examination. Fecal elastase level was within reference range (>500 μg/g); abdominal computed tomographic image revealed a dilated, tortuous sigmoid colon; and full-thickness rectal biopsy was negative for Hirschsprung disease. She was referred for sweat chloride testing to assess for CF. Results of 3 separate sweat chloride tests were indeterminate (Table), prompting pulmonology referral. A 97-mutation CF transmembrane conductance regulator (CFTR) analysis panel was negative. Whole-genome sequence analysis revealed 1 CF-causing mutation (c.2249C>T) and 2 likely benign variants (c.1408A>G and c.2562T>G).No further testing; the patient does not have CF What Would You Do Next?
Additional genetic testing
Alternative CFTR functional testing
Repeat sweat chloride analysis
No further testing; the patient does not have CF
null
B
Alternative CFTR functional testing
Cystic fibrosis is caused by mutations in the CFTR gene on chromosome 7.2,3 The CFTR protein is an anion channel that transports chloride and bicarbonate across the epithelium in many organs. CFTR mutations can cause reduced CFTR protein expression, which subsequently results in a reduction of the number of CFTR anion channels present on the epithelial membrane. CFTR mutations may also result in abnormal channel function, causing impaired ion and fluid homeostasis, hyperviscous secretions, and multisystem disease. CF-related lung disease includes mucus plugging, chronic infection, airway remodeling, and progressive decline in lung function. Gastrointestinal CFTR dysfunction results in chronic constipation and malnutrition due to viscous secretions in the intestinal tract and pancreatic ducts. Additional manifestations of CF include diabetes mellitus, azoospermia, and low bone mineral density. The diagnosis of CF is based on clinical presentation, family history, or a positive newborn screening test in addition to evidence of an abnormal CFTR protein or gene.1 Most patients are now identified through newborn screening; however, there is an increasing number of later diagnoses, especially in adulthood, representing up to 7% of CF cases.In sweat glands, CFTR protein facilitates chloride resorption from the ductal lumen into the epithelium. CFTR protein dysfunction results in defective resorption of chloride and elevated sweat chloride concentrations. Sweat chloride testing should be performed by experienced personnel adhering to international guidelines.4,5 The standard approach uses topical pilocarpine, a muscarinic agonist, and a small electric current to stimulate sweat production for 5 minutes, followed by sweat collection on gauze or with a capillary tube for up to 30 minutes.6 Depending on the method, 75 mg or 15 μL of sweat is required to determine chloride concentration.7The upper limit of normal sweat chloride concentration is 30 mmol/L,1,4 and a concentration greater than 60 mmol/L in a person with clinical features of CF indicates the presence of CF. The test has nearly 100% sensitivity and 96% specificity, and is the criterion standard diagnostic test for CF.1,7,8 Sweat chloride concentrations between 30 mmol/L and 60 mmol/L are considered indeterminate and require additional diagnostic evaluation.4 Federal reimbursement for sweat chloride testing is $82.90.This patient had sweat chloride concentration values consistently in the indeterminate range. In the absence of factors that may falsely elevate sweat chloride concentration (eg, dehydration; malnutrition; ectodermal dysplasia; endocrine disorders, including hypothyroidism, hypoparathyroidism, or cortisol deficiency7), additional diagnostic testing is necessary. Genetic CFTR testing revealed a single CF-causing mutation, which is insufficient to diagnose CF. Additional physiologic CFTR testing is the next diagnostic step. This diagnostic challenge is increasingly frequent, particularly in adults with relatively mild symptoms. Patients who do not meet the diagnostic criteria for CF following extensive workup may be classified as having CFTR-related metabolic syndrome (newborn screen positive for at least 1 known CF-causing CFTR mutation) or CFTR-related disorder (disease limited to 1 organ [eg, pancreatitis]).1Indeterminate sweat chloride concentration results should prompt further testing, starting with genetic CFTR analysis. If the CFTR genotype is undefined, further CFTR functional tests should be performed. Nasal potential difference measurement uses electrodes and sequential perfusion of compounds that affect sodium and chloride transport into the nasal cavity to measure CFTR-dependent voltage across the nasal epithelium. This process quantifies CFTR function in vivo and is highly sensitive and specific for diagnosing CF.9 Intestinal current measurement can be a useful alternative. In this test, epithelium from rectal biopsy samples is stimulated by exposure to compounds affecting sodium and chloride transport, and CFTR-dependent ion transport is quantified as electrical current.10Nasal potential difference testing was attempted but not completed because of poor patient cooperation. Intestinal current measurement demonstrated minimal CFTR function (<5% of normal function; CF diagnostic threshold, <25% of normal function10), and the diagnosis of CF was made. Based on symptoms, including greasy, bulky stools, she was empirically prescribed pancreatic enzyme replacement therapy (PERT). One month after diagnosis, her constipation was improved. PERT and gastrostomy feeding tube improved her BMI to the 70th percentile (eFigureA in the Supplement). Six years after diagnosis she has developed chronic productive cough, sinusitis, mild bronchiectasis (eFigureB in the Supplement), lung function decline, and positive respiratory cultures for classic microorganisms of CF (eFigureC in the Supplement).CF is a multisystem disease with mild forms increasingly recognized in children and adults.Sweat chloride testing remains the diagnostic standard for CF. Sweat chloride testing should be the initial test performed in any patient with clinical suspicion of CF.Genetic testing can help clarify an indeterminate diagnosis, provide prognostic information, and guide disease- and CFTR mutation– specific therapy.Nasal potential difference/intestinal current measurement may be useful to diagnose CF in a patient with indeterminate sweat chloride concentration values and less than 2 well-characterized disease-causing mutations.
Diagnostic
A 5-year-old girl was referred to a pediatric gastroenterology clinic for chronic constipation and poor weight gain. During her first week of life, she developed diarrhea and vomiting. With initiation of solid food, she developed laxative-dependent constipation. She underwent newborn genetic screening before routine cystic fibrosis (CF) screening. Results were normal. At the time of presentation to the gastroenterology clinic, she had no respiratory symptoms. Results of anorectal manometry, spinal magnetic resonance imaging, and thyroid studies were normal. Family history included constipation in a sister and a great aunt with CF. Her body mass index (BMI) was below the third percentile (eFigureA in the Supplement). Physical examination findings were unremarkable, including normal respiratory examination. Fecal elastase level was within reference range (>500 μg/g); abdominal computed tomographic image revealed a dilated, tortuous sigmoid colon; and full-thickness rectal biopsy was negative for Hirschsprung disease. She was referred for sweat chloride testing to assess for CF. Results of 3 separate sweat chloride tests were indeterminate (Table), prompting pulmonology referral. A 97-mutation CF transmembrane conductance regulator (CFTR) analysis panel was negative. Whole-genome sequence analysis revealed 1 CF-causing mutation (c.2249C>T) and 2 likely benign variants (c.1408A>G and c.2562T>G).No further testing; the patient does not have CF
what would you do next?
What would you do next?
Additional genetic testing
Alternative CFTR functional testing
Repeat sweat chloride analysis
No further testing; the patient does not have CF
b
1
1
1
0
female
0
0
5
0-10
null
591
original
https://jamanetwork.com/journals/jama/fullarticle/2723634
A 26-year-old white man presented for evaluation of blurry vision over the past year. He had no back or joint pain or skin rashes. Five years ago he had a black ink tattoo placed on his left arm, and 3 years ago a second tattoo was placed on his right arm. Two years after the right arm tattoo was placed, he noticed tattoo elevation on the left arm and onset of decreased vision. He had recurrent episodes of ocular redness and blurred vision, coincident with inflammation of the skin surrounding both tattoos. Two months before presentation, bilateral peripheral iridotomies (iris laser procedures) were performed because of inflammation-induced adhesions between the iris and the lens (Figure, panel A).A, Slitlamp photograph of left eye. B, Tattooed skin on left arm. C, Hematoxylin-eosin stain of skin biopsy sample (original magnification ×100).At presentation, best corrected visual acuity was 20/20 in the right eye and 20/40 in the left. Intraocular pressure was 34 mm Hg in the right eye (upper limit of normal, 21 mm Hg) and 11 mm Hg in the left. On anterior segment slitlamp examination, there were no visible inflammatory cells in the anterior chamber of the right eye, but there were more than 26 cells per 1-mm field in the left. The left arm tattoo was elevated, with nodular elevation most prominent at the superior aspect of the tattoo (Figure, panel B). Punch biopsy of the tattooed skin revealed epithelioid histiocytes with macrophages containing coarse black granular pigment (Figure, panel C). Results of testing for syphilis, tuberculosis, and sarcoidosis were negative. What Would You Do Next?
Initiate topical steroid eyedrop therapy
Recommend removal of tattoos
Refer immediately to rheumatology
Perform surgical implantation of glaucoma drainage device
Tattoo-associated uveitis
A
Initiate topical steroid eyedrop therapy
The key to the correct diagnosis is the history of recurrent skin tattoo inflammation and concurrent ocular inflammation, with an otherwise negative workup for systemic infectious or inflammatory causes of bilateral uveitis. Results of the skin biopsy suggested that tattoo-associated uveitis was the most likely diagnosis.There is insufficient evidence to suggest that tattoo removal improves outcomes, particularly when large tattooed skin areas preclude easy removal.1 Therapeutic options range from topical and periocular steroids to systemic steroids or steroid-sparing agents such as methotrexate, azathioprine, or adalimumab. Before initiating immunosuppressive therapy, however, common etiologies of uveitis, such as tuberculosis and syphilis, should be ruled out to avoid missing a diagnosis that could be exacerbated by steroid or other immune-compromising therapy. Elevated intraocular pressure due to intraocular inflammation should be treated medically using pressure-lowering eyedrops. Surgical intervention at the time of initial presentation is not recommended before a trial of medical therapy to reduce the underlying inflammation.One of the earliest reports of tattoo-associated uveitis described patients with histologically confirmed tattoo granulomas and concurrent ocular inflammation.2 Tattoo-associated uveitis occurs 6 months to 10 years after tattoo placement. Typical examination findings include bilateral, recurrent anterior or panuveitis with absence of cutaneous inflammation at the time tattoos were placed.3 It has been suggested that the ocular manifestations represent a delayed hypersensitivity reaction to the tattoo ink.2 The inclusion of tattoo pigment in the granulomas, as demonstrated in this patient, suggests an immune response to the heavy metal compounds in tattoo pigment.4 This is consistent with rare case reports of disease resolution after tattoo excision.5 Tattoo pigment has been isolated from regional lymph nodes in animal studies,6 a process likely facilitated by macrophage phagocytosis of sequestered dye. In patients with tattoo-associated uveitis it is likely that T-lymphocyte activation initiates skin granuloma formation, accompanied by an ocular inflammatory response. In animals, tattoo pigment deposits in the liver suggest that the pigment may spread systemically.7 Accumulation of tattoo pigment in the uvea may promote recurrent ocular inflammation. The lack of systemic pulmonary or arthritic findings in these patients suggests that tattoo-associated uveitis is distinct from sarcoidosis, despite the finding of noncaseating granulomas in both conditions.8,9Tattoo-associated uveitis may possibly occur because of an environmental trigger in a genetically predisposed host. However, while several HLA associations, such as HLA-B27 and HLA-A29, have been documented in other uveitides, HLA accounts for only a minor portion of the overall genetic risk in these patients.10 No genetic factors have been implicated in tattoo-associated uveitis.The pathophysiology and clinical course of tattoo-associated uveitis remain poorly understood. To help identify the diagnosis, clinicians should ask patients presenting with uveitis about history of tattoos.Results of a systemic workup for infectious and inflammatory conditions were unremarkable, and the patient was started on topical prednisolone acetate eyedrops for treatment of ocular inflammation. The patient continued to have episodes of uveitis, even after prescription of adalimumab and methotrexate, requiring administration of short-acting intraocular steroids. His vision worsened progressively over the subsequent 9 months until he had difficulty counting fingers at 0.3 m (1 ft) in each eye. The visual decline was attributed to cataract and persistent inflammation. He underwent bilateral fluocinolone acetonide implantation for improved long-term control, with concomitant cataract surgery 12 months after presentation. One month after surgery, best corrected visual acuity had improved to 20/300 in the right eye and 20/30 in the left, with improved intraocular inflammation.
General
A 26-year-old white man presented for evaluation of blurry vision over the past year. He had no back or joint pain or skin rashes. Five years ago he had a black ink tattoo placed on his left arm, and 3 years ago a second tattoo was placed on his right arm. Two years after the right arm tattoo was placed, he noticed tattoo elevation on the left arm and onset of decreased vision. He had recurrent episodes of ocular redness and blurred vision, coincident with inflammation of the skin surrounding both tattoos. Two months before presentation, bilateral peripheral iridotomies (iris laser procedures) were performed because of inflammation-induced adhesions between the iris and the lens (Figure, panel A).A, Slitlamp photograph of left eye. B, Tattooed skin on left arm. C, Hematoxylin-eosin stain of skin biopsy sample (original magnification ×100).At presentation, best corrected visual acuity was 20/20 in the right eye and 20/40 in the left. Intraocular pressure was 34 mm Hg in the right eye (upper limit of normal, 21 mm Hg) and 11 mm Hg in the left. On anterior segment slitlamp examination, there were no visible inflammatory cells in the anterior chamber of the right eye, but there were more than 26 cells per 1-mm field in the left. The left arm tattoo was elevated, with nodular elevation most prominent at the superior aspect of the tattoo (Figure, panel B). Punch biopsy of the tattooed skin revealed epithelioid histiocytes with macrophages containing coarse black granular pigment (Figure, panel C). Results of testing for syphilis, tuberculosis, and sarcoidosis were negative.
what would you do next?
What would you do next?
Recommend removal of tattoos
Initiate topical steroid eyedrop therapy
Refer immediately to rheumatology
Perform surgical implantation of glaucoma drainage device
b
0
1
1
1
male
0
0
26
21-30
White
592
original
https://jamanetwork.com/journals/jama/fullarticle/2721290
A 7-week-old, previously healthy full-term male infant presented with a 4-week history of a worsening, widespread, papulopustulovesicular eruption, which began as scattered papulovesicles over the chest. The infant was born to a 31-year-old mother by normal vaginal delivery. The mother had a benign prenatal course during pregnancy. Since the rash developed, the infant had been fussy with increasing restlessness at night. The parents denied any personal or family history of rash. On further questioning, the parents reported that the infant had had contact with a babysitter who reported pruritus, especially at night. The sitter had provided domestic care for the infant since birth until leaving that position 3 weeks earlier.On examination, the infant was afebrile with normal vital signs. Multiple disseminated, edematous, pin-sized, papulopustulovesicles and 5- to 10-mm coalescing, crusted nodules were found all over the body; some with oval to elongated, serpiginous or J-shaped burrows (Figure 1). No excoriations were visible. A Tzanck smear showed no multinucleated giant cells in the pustular contents. A Gram stain and potassium hydroxide (KOH) preparation of the pustular contents showed no bacterial or fungal elements. Cultures of the pustular contents yielded no bacterial or fungal growth. The differential of a complete blood cell count showed 12% eosinophils. Results of other routine laboratory investigations were unremarkable.Perform skin scraping of burrows from multiple sites What Would You Do Next?
Administer intravenous broad-spectrum antibiotics
Start a topical steroid
Perform skin scraping of burrows from multiple sites
Do nothing and reassure the parents
Neonatal scabies
C
Perform skin scraping of burrows from multiple sites
The key to the correct diagnosis is the widespread inflammatory burrows without excoriations, the history of contact with a babysitter with symptoms consistent with scabies, and the fussiness and restlessness at night in an otherwise healthy neonate. Unlike in adults, the palms, soles, trunk, and skin above the neck are commonly affected by characteristic scabies skin lesions in neonates. Contact with an individual with signs and symptoms of scabies can strongly suggest the diagnosis.Scabies is a global health problem affecting more than 300 million individuals annually. Scabies is an extremely contagious skin infestation caused by female Sarcoptes scabiei mites.1 Scabies often affects children, with varying presentations in different age groups.2 Given that neonates are developmentally ineffective scratchers, fussiness, irritability, and restlessness at night, rather than scratching with its secondary excoriations, are common in neonatal scabies. In neonates, the infestation is insidious and lasts for relatively long periods, frequently leading to multiple lesions in different developmental stages ranging from papulopustulovesicles to nodules. Compared with the intact burrows found predominately in the interdigital web spaces and the extremities of adults, neonates often present with highly inflamed burrows over the whole body, characterized by red, edematous, oval to elongated, serpiginous or J-shaped papules, vesicles, and nodules.Scabies is transmitted through close personal contact with an infected individual or his or her personal items, including clothing, bedding, towels, and gloves.3,4 The symptoms develop within 2 to 6 weeks after infection.3 Considering the various manifestations differing from classic scabies and the rarity of scabies in neonates, neonatal scabies is often diagnosed at a later stage or misdiagnosed altogether. In neonates with a suspected diagnosis of scabies, the presence of 1 or more family members with the typical rash or pruritus increases the likelihood of the diagnosis. Other persons temporarily exposed to the neonate, including domestic workers and health care workers in rehabilitation centers, long-term care facilities, and newborn nurseries, should be evaluated, especially when family members are not infected.4,5The diagnosis of scabies is confirmed by scraping burrows from multiple sites and finding microscopic evidence of mites and ova, which are easily detected with or without smears or stains such as Tzanck smear and KOH preparation.6 In this patient, the characteristic rash, which lasted 4 weeks without constitutional symptoms and systemic involvement, was not consistent with varicella or herpetic infection.7 Compared with neonatal scabies, the vesicles, pustules, or bullae observed in impetigo caused by bacterial infection often present with clear yellow or turbid fluid. The asymptomatic rash of erythema toxicum neonatorum occurs within the first days of life and resolves spontaneously.8 The US Food and Drug Administration has not approved any therapies for infants younger than 2 months. However, a sulfur ointment (5%-10%) and permethrin cream have been recommended as safe options for neonates.2,9 Topical crotamiton or oral ivermectin are potential alternatives; however, data supporting their use in neonates are limited.1 Simultaneous treatment of contacts is needed.The patient and the babysitter underwent skin scraping with microscopic findings of numerous mites (Figure 2). The infant was treated with a sulfur ointment (5%) for 3 consecutive days and treatment was repeated 1 week later, with rapid recovery of normal temperament, good sleep, and complete resolution of the rashes. The babysitter was treated with a permethrin cream (5%) that was reapplied 1 week later, and all family members received prophylaxis with 1 cycle of permethrin therapy. Besides hygienic cleaning of fixtures and appliances, all bedding, clothing, and towels were washed in hot water and dried with high heat to decontaminate.3 Three months later, the infant and all contacts had no evidence of scabies.Sarcoptes scabiei mite visible on light microscopy of skin scraping (original magnification ×40).
General
A 7-week-old, previously healthy full-term male infant presented with a 4-week history of a worsening, widespread, papulopustulovesicular eruption, which began as scattered papulovesicles over the chest. The infant was born to a 31-year-old mother by normal vaginal delivery. The mother had a benign prenatal course during pregnancy. Since the rash developed, the infant had been fussy with increasing restlessness at night. The parents denied any personal or family history of rash. On further questioning, the parents reported that the infant had had contact with a babysitter who reported pruritus, especially at night. The sitter had provided domestic care for the infant since birth until leaving that position 3 weeks earlier.On examination, the infant was afebrile with normal vital signs. Multiple disseminated, edematous, pin-sized, papulopustulovesicles and 5- to 10-mm coalescing, crusted nodules were found all over the body; some with oval to elongated, serpiginous or J-shaped burrows (Figure 1). No excoriations were visible. A Tzanck smear showed no multinucleated giant cells in the pustular contents. A Gram stain and potassium hydroxide (KOH) preparation of the pustular contents showed no bacterial or fungal elements. Cultures of the pustular contents yielded no bacterial or fungal growth. The differential of a complete blood cell count showed 12% eosinophils. Results of other routine laboratory investigations were unremarkable.Perform skin scraping of burrows from multiple sites
what would you do next?
What would you do next?
Perform skin scraping of burrows from multiple sites
Do nothing and reassure the parents
Administer intravenous broad-spectrum antibiotics
Start a topical steroid
a
0
1
1
1
male
0
0
31
31-40
null
593
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2712410
A teenaged male presented with a 2-week history of vegetating, bleeding plaques and pustules on his face, scalp, trunk, and extremities. He reported that lesions began as pustules and developed into painful, itchy plaques. Before evaluation at a referral hospital in Kenya, the patient was treated with prednisolone tablet 5 mg twice daily, clindamycin hydrochloride 300 mg daily, and fluconazole 200 mg daily, but he continued to develop new lesions. A complete review of systems was notable for the patient’s difficulty with walking because of skin lesions. On examination, pustules and hemorrhagic-crusted plaques were seen on the scalp, legs, arms, and trunk along with keloidal plaques on the face (Figure, A and B). Enzyme-linked immunosorbent assay testing for HIV was negative. A punch biopsy specimen from the arm was obtained for histopathologic study (Figure, C and D).Extensive vegetative plaques with bleeding and ulceration on extremities (A), and pustules and ulcerated plaques on extremities and trunk (B). Histopathologic examination of a biopsy specimen taken from the arm demonstrates pseudoepitheliomatous hyperplasia (hematoxylin-eosin stain, original magnification ×100) (C) and dermal neutrophilia and microabscesses with eosinophilia (hematoxylin-eosin stain, original magnification ×400) (D). What Is Your Diagnosis?
Pyoderma gangrenosum
Blastomycosis
Pyoderma vegetans
Halogenoderma
C. Pyoderma vegetans
C
Pyoderma vegetans
Histopathologic study demonstrated epidermal hyperplasia and exocytosis of neutrophils, in addition to dense dermal-mixed inflammatory cell infiltration and microabscesses scattered with eosinophils. No granulomas or multinucleated giant cells were noted. Results of periodic acid–Schiff staining were negative. The findings were consistent with pyoderma vegetans. Acid-fast bacilli stain, tissue culture, and direct immunofluorescence were not available.The patient was treated with dapsone 100 mg daily, prednisolone 50 mg daily, flucloxacillin for impetiginization, and petrolatum jelly–coated gauze. Substantial clinical improvement was noted within a week of hospitalization. After the patient was discharged from the hospital, prednisolone was tapered off over a month and dapsone therapy was continued for 3 months. Nine months after stopping dapsone therapy, the patient continued to show clear skin, with only asymptomatic hypertrophic and keloidal scars in areas of prior involvement.Pyoderma vegetans is a rare inflammatory condition that presents as large vegetative plaques with vesicopustules. Because lesions of the oral mucosa are often present, this condition is also referred to as pyostomatitis vegetans and pyodermatitis-pyostomatitis vegetans. Its origin is unknown and proposed to be related to immunological dysfunction associated with bacterial, fungal, or autoimmune conditions.1 Pyoderma vegetans is associated with primary and acquired immunodeficiencies, including HIV infection, leukemia, alcoholism, diabetes, nutritional deficiency, inflammatory bowel disease, and immunosuppressive therapy.2-4The differential diagnosis for vegetating plaques is broad, including deep fungi, nontuberculous mycobacteria, blastomycosis-like pyoderma, halogenoderma, vegetative pyoderma gangrenosum, pemphigus vegetans, pyoderma vegetans, giant keratoacanthoma, and squamous cell carcinoma. Clinical data may help to distinguish these entities. Halogenoderma occurs in the context of bromide exposure. Associated trauma could support pyoderma gangrenosum in the context of pathergy. In an immunocompetent black African patient, the presence of disseminated lesions suggests that giant keratoacanthoma and squamous cell carcinoma are less likely. Pemphigus vegetans is a rare variant of pemphigus vulgaris that often favors flexural areas and almost always involves oral mucosa.Histopathologic study is most helpful in narrowing the clinical differential diagnosis. In deep fungal infections with blastomycosis-like tissue reaction pattern (eg, blastomycosis, chromoblastomycosis, and sporotrichosis) and blastomyocosis-like pyoderma, granulomas and multinucleated giant cells should be prominent features, in addition to pseudoepitheliomatous hyperplasia and neutrophilic microabscesses. Fungal stains and fungal culture can facilitate diagnosis. Blastomycosis-like pyoderma is considered to be an exaggerated tissue reaction to an underlying bacterial infection, and its diagnosis requires growth of at least 1 pathogenic bacteria from the culture of a tissue biopsy specimen.5 In mycetoma (eumycetoma, actinomycetoma, and botryomycosis), characteristic grains are seen in the center of suppurative zones and suppurative granulomas. Nontuberculous mycobacteria have a variety of histopathologic findings, such as epidermal changes, including ulceration, acanthosis, or pseudoepitheliomatous hyperplasia; tuberculoid granulomas, occasionally with caseous necrosis, poorly formed granulomas, suppurative granulomas, nonspecific chronic inflammation, and rarely lichenoid and granulomatous dermatitis; necrotizing folliculitis; or panniculitis of septal or mixed lobular and septal type. Acid-fast bacilli stain may identify organisms, although culture or polymerase chain reaction testing is often necessary.Vegetating lesions of halogenoderma show pseudoepitheliomatous hyperplasia with intraepidermal and dermal abscess with neutrophils and few eosinophils. Vegetative pyoderma gangrenosum, also known as superficial granulomatous pyoderma, is characterized by focal neutrophilic abscesses in the papillary dermis often associated with peripheral palisading histiocytes and foreign-body giant cells, in addition to pseudoepitheliomatous, vegetative hyperplasia and sinus tract formation.6 Pemphigus vegetans shares histopathologic features with pyoderma vegetans, including acanthosis and intraepidermal abscesses with eosinophils, but pemphigus vegetans demonstrates suprabasal splitting or acantholysis, whereas pyoderma vegetans has focal subepidermal clefting. In addition, direct immunofluorescence of pemphigus vegetans demonstrates intercellular deposition of IgG and C3, whereas results of direct immunofluorescence of pyoderma vegetans are negative; however, cases of pyoderma vegetans associated with inflammatory bowel disease have been reported with direct immunofluorescence showing intercellular IgA deposition.7 Squamous cell carcinoma typically has nests of squamous epithelial cells arising from the epidermis and extending into the dermis with large atypical cells.No clear treatment algorithm exists for pyoderma vegetans. Associated underlying disease should be treated.8 Corticosteroids are widely accepted as first-line treatment; however, lesions can flare after a steroid taper.3 Successful use of systemic antibiotic therapy, based on bacterial susceptibility, in clearing the lesions varies.9 Various second-line therapies have been reported, including cyclosporine, tacrolimus, azathioprine sodium, etanercept, and dapsone.2,3,10 Herein, this case describes a patient with pyoderma vegetans who presented with vegetating plaques and pustules on the head, trunk, and extremities. The clinical differential diagnosis is broad, but histopathologic findings and subsequent response to prednisolone and dapsone helped to determine a diagnosis. Pyoderma vegetans should be considered in the differential diagnosis of vegetating plaques and pustules.
Dermatology
A teenaged male presented with a 2-week history of vegetating, bleeding plaques and pustules on his face, scalp, trunk, and extremities. He reported that lesions began as pustules and developed into painful, itchy plaques. Before evaluation at a referral hospital in Kenya, the patient was treated with prednisolone tablet 5 mg twice daily, clindamycin hydrochloride 300 mg daily, and fluconazole 200 mg daily, but he continued to develop new lesions. A complete review of systems was notable for the patient’s difficulty with walking because of skin lesions. On examination, pustules and hemorrhagic-crusted plaques were seen on the scalp, legs, arms, and trunk along with keloidal plaques on the face (Figure, A and B). Enzyme-linked immunosorbent assay testing for HIV was negative. A punch biopsy specimen from the arm was obtained for histopathologic study (Figure, C and D).Extensive vegetative plaques with bleeding and ulceration on extremities (A), and pustules and ulcerated plaques on extremities and trunk (B). Histopathologic examination of a biopsy specimen taken from the arm demonstrates pseudoepitheliomatous hyperplasia (hematoxylin-eosin stain, original magnification ×100) (C) and dermal neutrophilia and microabscesses with eosinophilia (hematoxylin-eosin stain, original magnification ×400) (D).
what is your diagnosis?
What is your diagnosis?
Blastomycosis
Pyoderma gangrenosum
Pyoderma vegetans
Halogenoderma
c
0
0
1
1
male
0
0
15
11-20
null
594
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2713948
A 26-year-old man presented with a several-year history of persistent round lesions with a scar-like appearance on his face. Before evaluation, his condition had been diagnosed as lupus and lipoatrophy and he tried several treatments with no success.The results of his physical examination showed several round atrophic lesions on his face, symmetric and bilateral, that were located especially on the malar and preauricular areas, with a diameter of 5 to 10 mm. His skin lesions were not indurated and displayed no epidermal changes (Figure 1). The patient denied experiencing previous inflammation. He had no history of chicken pox or acne or traumatic lesions in those areas. New punch biopsy specimens were obtained for further evaluation.Atrophic lesions on the patient’s left cheek (A) and with a cicatricial appearance on the preauricular right area (B). What Is Your Diagnosis?
Primary anetoderma
Atrophoderma vermiculata
Atrophia maculosa varioliformis cutis
Facial morphea
C. Atrophia maculosa varioliformis cutis
C
Atrophia maculosa varioliformis cutis
Atrophia maculosa varioliformis cutis (AMVC) was first described in 1918 by Heidingsdfield and since then approximately 20 cases have been reported.1 It is considered a rare disease that appears spontaneously and is not preceded by other inflammatory or traumatic events. From a clinical point of view, this entity is characterized by the progressive appearance of sharp depressions of various shapes, often round or oval, with regular margins and clear-cut edges, which are all asymptomatic. Its etiology remains unclear, although its family clustering suggests that it could be a genetic disorder.2 The histological findings are variable and nonspecific, including a thinning of the horny layer or the entire epidermis, decreased or fragmented elastic fiber, and a lymphocytic perivascular infiltrate.1-3The differential diagnosis includes scars that are related to acne, chicken pox varicella, dermatitis artifacta, atrophoderma vermiculata, facial morphea, or primary anetoderma. Atrophoderma vermiculata is a rare condition that is considered to belong to the keratosis pilaris group, characterized by smaller lesions on the cheeks, which is presumably a late reaction to inflammation around the horny plugs. The honeycomb-like atrophy on the cheeks of atrophoderma vermiculata helped to exclude this entity in the patient. Facial morphea usually shows an active erythematous or violaceus inflammatory border. Primary anetoderma is a rare idiopathic dermatosis that is characterized by areas of flaccid skin located in extremities and trunk due to the loss of elastic tissue in the absence of a secondary cause.There is no standardized treatment for patients with AMVC, although filler injections, dermabrasion, or laser resurfacing may theoretically be helpful in improving the facial scarring.3,4 Although there are no follow-up data reported in most previous cases, it is important to explain to patients with AMVC that it is a benign and nonprogressive dermatosis.The histopathologic examination results showed a significant epidermal dell and a superficial dermal lymphocytic infiltrate that was associated with dermal fibrosis. An orcein stain demonstrated a reduction of elastic fibers in the dermis beneath the epidermal dell (Figure 2). The patient’s skin lesions remain stable after 1 year of follow-up.A, Panoramic histological view shows a slight depression of the epidermis (hematoxylin-eosin; original magnification, ×2). B, Orcein staining allows a demonstration of the reduction in the elastic fibers beside the lymphocytic perivascular infiltrate compared with the surrounding and healthy area (orcein; original magnification, ×10).
Dermatology
A 26-year-old man presented with a several-year history of persistent round lesions with a scar-like appearance on his face. Before evaluation, his condition had been diagnosed as lupus and lipoatrophy and he tried several treatments with no success.The results of his physical examination showed several round atrophic lesions on his face, symmetric and bilateral, that were located especially on the malar and preauricular areas, with a diameter of 5 to 10 mm. His skin lesions were not indurated and displayed no epidermal changes (Figure 1). The patient denied experiencing previous inflammation. He had no history of chicken pox or acne or traumatic lesions in those areas. New punch biopsy specimens were obtained for further evaluation.Atrophic lesions on the patient’s left cheek (A) and with a cicatricial appearance on the preauricular right area (B).
what is your diagnosis?
What is your diagnosis?
Facial morphea
Atrophia maculosa varioliformis cutis
Atrophoderma vermiculata
Primary anetoderma
b
0
0
1
1
male
0
0
26
21-30
null
595
original
https://jamanetwork.com/journals/jamadermatology/fullarticle/2716289
A 60-year-old man with a history of IgA kappa multiple myeloma was referred to our department. He had been diagnosed with multiple myeloma 2 years earlier after discovery of osteolytic bone lesions of the skull and the findings of a bone marrow biopsy. The patient had been treated with bortezomib and dexamethasone followed by autologous blood progenitor cell transplantation, which resulted in complete response. At the time of the consultation, he was free of disease.The patient was referred for evaluation of an indolent, erythematous papule on his glans penis of 2 weeks’ duration. He denied sexual risk behavior and reported no recent changes in medication. Careful physical examination revealed a shiny, tense, erythematous, hemispherical papule measuring 5 mm in diameter located on the lateral aspect of the glans (Figure 1A). There were no other skin lesions. The lymph nodes were not palpable, and there was no organomegaly. A punch biopsy was performed for histopathological evaluation (Figure 1B and C).A, Shiny, erythematous, hemispherical papule on the lateral aspect of the glans penis. B and C, Hematoxylin-eosin stain was performed for evaluation. What Is Your Diagnosis?
Primary penile B-cell non-Hodgkin lymphoma
Kaposi sarcoma of the penis
Plasmacytoma
Primary nodular amyloidosis of the glans penis
C. Plasmacytoma
C
Plasmacytoma
Histologic examination revealed a nodular, nonencapsulated, dense infiltrate of plasma cells in the superficial submucosa with no involvement of the overlying mucosa (Figure 1B). The proliferating plasma cells had a high nuclear-cytoplasmic ratio and were slightly heterogeneous. Some plasma cells had dispersed chromatin, conspicuous nucleoli, and eosinophilic cytoplasms with ill-defined cell membranes. Scattered mitoses were present.Immunohistochemistry revealed diffuse and intense staining for CD138 and kappa light chains (Figure 2A) and was negative for lambda light chains (Figure 2B).Diffuse and intense staining for kappa light chains (A) and negative staining for lambda light chains (B) (original magnification ×40).There are 4 major types of neoplastic plasma cell proliferations: multiple myeloma, extramedullary plasmacytoma without multiple myeloma, solitary plasmacytoma of bone, and plasma cell leukemia.1Multiple myeloma is a monoclonal plasma cell proliferative disorder that accounts for nearly 10% of all hematological malignancies.2 Cutaneous manifestations occur in an estimated 5% to 10% of cases. The lesions can be nonspecific, such as amyloidosis or purpura, or specific, such as plasma cell neoplasia.3 Neoplastic plasma cell proliferations in the skin secondary to multiple myeloma are uncommon and normally indicate direct spread from a solitary bone plasmacytoma or an underlying osteolytic bone lesion. Although soft tissue involvement of the oral cavity and the upper airway are frequently reported, secondary cutaneous plasmacytomas without bone involvement are rare.1,4Secondary cutaneous plasmacytomas, which may present as solitary or multiple lesions, present as firm, cutaneous or subcutaneous, erythematous-to-reddish, ellipsoidal papules or nodules measuring from a few millimeters to 5 cm, although larger lesions have been described.1,5 Cutaneous lesions show a predilection for the extremities, trunk, head, and neck.1,6 Penile plasmacytoma is an extremely rare entity and scarcely reported in the literature. To date, only 3 cases have been reported: 2 extramedullary cutaneous plasmacytomas involving the penis,7,8 and 1 case of a primary extramedullary plasmacytoma of the penis.9In these patients, the pattern of the dermal plasma cell infiltrate is generally either nodular or interstitial, with the former being more common.6 Plasma cells may display typical morphologic features—basophilic, eccentric nuclei, and clock-face chromatin distribution—but they frequently show atypia with dispersed chromatin and prominent nucleoli.1In immunohistochemical studies, neoplastic plasma cells show positive staining for CD79a, CD138, and epithelial membrane antigen. Immunoreactivity for CD38 and CD43 is less constant.1 The type of immunoglobulin produced by neoplastic plasma cells and light-chain restriction (kappa or lambda) correlates with serum electrophoresis, supporting the clonal origin of the cells and distinguishing them from a reactive plasma cell proliferation.4Treatment targets the monoclonal gammopathy and includes surgical excision plus radiotherapy for a solitary plasmacytoma, and systemic therapy for multiple lesions. However, skin involvement in multiple myeloma is associated with a poor prognosis. In more than 50% of cases, cutaneous manifestations occur late in the course of the disease and are indicative of a high tumor cell burden. Even with aggressive therapy, most patients survive less than 1 year once cutaneous involvement appears.4,6We have documented the first case, to our knowledge, of a secondary plasmacytoma involving the glans penis, which was the first manifestation of relapsed multiple myeloma. The disease progressed, and the patient developed widespread osteolytic lesions of the skull, jaw, and sacrum. Despite combined chemotherapy, he experienced clinical and hematologic progression of the disease and died 3 months after diagnosis of the penile lesion. Cutaneous involvement is a marker for poor survival in this setting.Although infrequent, skin involvement in multiple myeloma is associated with an ominous prognosis. Awareness of this association and close monitoring are therefore important because skin lesions may be the first manifestation of multiple myeloma or indicate a relapse after prior response to therapy.
Dermatology
A 60-year-old man with a history of IgA kappa multiple myeloma was referred to our department. He had been diagnosed with multiple myeloma 2 years earlier after discovery of osteolytic bone lesions of the skull and the findings of a bone marrow biopsy. The patient had been treated with bortezomib and dexamethasone followed by autologous blood progenitor cell transplantation, which resulted in complete response. At the time of the consultation, he was free of disease.The patient was referred for evaluation of an indolent, erythematous papule on his glans penis of 2 weeks’ duration. He denied sexual risk behavior and reported no recent changes in medication. Careful physical examination revealed a shiny, tense, erythematous, hemispherical papule measuring 5 mm in diameter located on the lateral aspect of the glans (Figure 1A). There were no other skin lesions. The lymph nodes were not palpable, and there was no organomegaly. A punch biopsy was performed for histopathological evaluation (Figure 1B and C).A, Shiny, erythematous, hemispherical papule on the lateral aspect of the glans penis. B and C, Hematoxylin-eosin stain was performed for evaluation.
what is your diagnosis?
What is your diagnosis?
Primary penile B-cell non-Hodgkin lymphoma
Kaposi sarcoma of the penis
Primary nodular amyloidosis of the glans penis
Plasmacytoma
d
0
1
1
1
male
0
0
60
51-60
null
596
original
https://jamanetwork.com/journals/jamasurgery/fullarticle/2702085
A 55-year-old man with a history of Childs B cirrhosis presented with a 2.5-year history of right groin pain and increased urinary frequency with difficulty voiding. On examination, he had a 5 cm × 3 cm palpable and partially reducible right groin mass that extended into the scrotum. He had a leukocytosis with a leukocyte count of 16 400/μL (to convert to ×109/L, multiply by 0.001), his creatinine level was 1.14 mg/dL (to convert to micromoles per liter, multiply by 88.4), and urinalysis showed leukocyturia with cultures that had positive results for Streptococcus viridians. Axial and sagittal (Figure) computed tomography scans with contrast are shown. A renography using mercaptoacetyltriglycine showed a 34% reduction of right renal function.Computed tomography cystogram without intravenous contrast, axial (A) and sagittal (B) view.Amyand hernia (appendix included within the inguinal hernia sac) What Is Your Diagnosis?
Sliding hernia
Saphena varix
Amyand hernia (appendix included within the inguinal hernia sac)
Communicating hydrocele
A. Sliding hernia
A
Sliding hernia
Inguinal hernias are one of the most common surgical diseases and account for up to 28% of all elective surgical cases performed in the United States.1 Because other diseases may mimic this condition, it is important to recognize other pathologies, as treatment may be different. Saphena varix may also present as a “groin bulge,” but it is a dilation of the saphenous vein due to valvular incompetence. Amyand hernia is an inguinal hernia containing the appendix. Therefore, an appendectomy may also be indicated at the time of inguinal hernia repair. Communicating hydrocele is when fluid flows between the scrotum and peritoneal cavity because of a patent processus vaginalis. Sliding hernias occur when a retroperitoneal organ constitutes part of the hernia sac.Sliding hernias that contain the bladder occur in about 1% to 4%2 of cases. The incidence of inguinal hernias containing ureter is extremely rare,3 and as of 2013, fewer than 140 cases have been reported.4 Associated bladder herniation is present in 25% of cases with inguinal hernias containing the ureter.5 Severe complications, such as renal failure, have been reported because of ureteral herniation, which is why it is crucial to identify these types of hernias. Comorbidities, such a liver dysfunction, can further complicate hernia repair and postoperative outcomes. In this case, the patient had a sliding hernia with the bladder and the right ureter constituting a portion of the hernia sac, resulting in right hydroureter and hydronephrosis and frequent urinary tract infections.Risk factors for bladder herniation include older age, male sex, obesity, previous herniorrhaphy, and lower urinary tract obstruction, such as in benign prostatic hyperplasia. Most of these are asymptomatic and diagnosed intraoperatively.6 Ureteral hernias most often occur on the right side and can present with abdominal pain, renal colic, or obstructive uropathy.5 Imaging scans to identify bladder and/or ureteral inguinal hernias include computed tomography, magnetic resonance imaging, a pyelogram, and/or a cystogram,2 which are important to obtain so that in addition to operative planning any additional anatomic abnormalities, such as ectopic kidneys, can be detected. Surgical repair is indicated in cases in which the bladder and/or ureter are contained within the hernia sac, because if they are left untreated, it can lead to complications, such as renal failure or urosepsis. It must be noted that when these cases are diagnosed intraoperatively, the operating surgeon must be alert as to not inadvertently injure these structures, which can occur in up to 12% of cases.7Complications due to a fully or partially obstructed bladder and/or ureter in an inguinal hernia can be fully reversed after surgical intervention and can have a dramatic improvement in the quality of life. For example, the resolution of frequent urinary tract infections and hydroureter/hydronephrosis that was noted on postoperative ultrasonography results allowed this patient to return to a normal lifestyle. The involvement of multidisciplinary teams, such as general surgeons and urologists, can aid in workup (ie, cystoscopy) and surgical intervention (ie, bladder or ureteral resection or repair if needed). Additionally, malignancy of the bladder or prostate has been reported in up to 11.2% of patients with bladder herniation. Therefore, these patients must be followed up closely and undergo screening tests.6Comorbidities increase the risk of overall complications for inguinal hernia repair. Cirrhosis in particular is generally a feared health problem, especially in regards to hernia repair, but in recent studies, the presence of cirrhosis (regardless of Child Turcotte Pugh score) did not significantly increase the risk of complications or recurrence.8 Aggressively controlling ascites preoperatively and postoperatively is needed to optimize a patient’s outcome. In this particular case, the patient developed mildly increased liver dysfunction postoperatively that was managed conservatively, and his levels subsequently returned to baseline.The patient had an uncomplicated postoperative course. He had a transient and self-limiting increase in liver function test results but was otherwise asymptomatic. Repeated renal ultrasonography revealed resolution of the hydroureter and the patient reported a greatly improved quality of life.
Surgery
A 55-year-old man with a history of Childs B cirrhosis presented with a 2.5-year history of right groin pain and increased urinary frequency with difficulty voiding. On examination, he had a 5 cm × 3 cm palpable and partially reducible right groin mass that extended into the scrotum. He had a leukocytosis with a leukocyte count of 16 400/μL (to convert to ×109/L, multiply by 0.001), his creatinine level was 1.14 mg/dL (to convert to micromoles per liter, multiply by 88.4), and urinalysis showed leukocyturia with cultures that had positive results for Streptococcus viridians. Axial and sagittal (Figure) computed tomography scans with contrast are shown. A renography using mercaptoacetyltriglycine showed a 34% reduction of right renal function.Computed tomography cystogram without intravenous contrast, axial (A) and sagittal (B) view.Amyand hernia (appendix included within the inguinal hernia sac)
what is your diagnosis?
What is your diagnosis?
Communicating hydrocele
Sliding hernia
Saphena varix
Amyand hernia (appendix included within the inguinal hernia sac)
b
1
1
0
1
male
0
0
55
51-60
null
597
original
https://jamanetwork.com/journals/jamasurgery/fullarticle/2711231
A healthy 29-year-old pregnant woman, with 34 weeks’ gestational age, presented to the emergency department with diffused acute abdominal pain and vomiting. The pain was located in the periumbilical region and intensified within a few hours. The patient felt normal fetal movements during this time. An initial surgical evaluation was done, and the cause for the abdominal pain was assumed to be not of surgical condition. Next, the patient was evaluated by an obstetrician-gynecologist, and although the gynecologic examination, ultrasonography, and monitoring were all normal, the patient was admitted for further supervision in the obstetrics department. The next day, when the abdominal pain and vomiting persisted and an elevated leukocyte blood count of 14 200/mL was noted (to convert to ×109 per liter, multiply by 0.001), a second surgical consultation was done. The physical examination was significant for diffuse abdominal tenderness. Although the patient was pregnant, and owing to nonavailability of magnetic resonance imaging at that time, a contrast-enhanced abdominopelvic computed tomography was performed (Figure 1).Coronal computed tomographic image showing an edematous small intestinal loop on the left abdomen, with uterus and head of fetus in the pelvis. What Is Your Diagnosis?
Crohn ileitis
Foreign body ingestion
Ileo-colic intussusception
Adenocarcinoma of small intestine
C. Ileo-colic intussusception
C
Ileo-colic intussusception
Computed tomography revealed edema of small intestinal loop involving the mucosa, proximal fecalization of intestinal content, and collapse of the distal right colon. The patient was urgently operated on, and resection of necrotic segment of terminal ileum with right colon was performed with primary anastomosis (Figure 2). A decision was made not to deliver the fetus at that time. At postoperative day 13, the patient had a surgical site infection and later wound dehiscence; hence, urgent laparotomy and cesarean section through the previous midline laparotomy incision was performed. Primary closure of the abdomen was done. Pathologic examination of the intestine revealed no leading mass as a cause of intussusception. Two years after the delivery, the patient is healthy and her child is developing normally.The cecum is being cut and necrotic small bowel is revealed.Abdominal pain in pregnant women is a common and bothersome medical concern. In the evaluation of abdominal pain in pregnant patients, one must consider the risks and benefits of the diagnostic methods and therapy to the mother and fetus. The delay in diagnosis and hence the treatment of the surgical abdomen in pregnant patients is caused by attribution of pain to pregnancy and the fear of unnecessary procedures and tests. That contributes to the high complications rate in this population, as seen in this particular case. Attention to detail, heightened suspicion, serial physical examination, clinical awareness, and systematic evaluation can help avert unnecessary maternal complications and fetal loss in emergency surgical conditions. The most common causes of acute abdomen pain in pregnancy are acute appendicitis, gallbladder-related disease, and small-bowel obstruction.1Intestinal obstruction during pregnancy has an incidence of 1 in 1500 pregnancies. Some cases of intestinal obstruction may be falsely diagnosed as hyperemesis gravidarum. Adhesions account for 60% to 70% of cases, and other etiologies include volvulus, intussusception, hernia, neoplasm, and appendicitis. Benign leading point for intussusception of the small intestine may be diverse and include lipoma, Meckel diverticulum, lymph nodes, adhesions, trauma, Peutz Jegher polyps, leiomyomas, neurofibromas, adenomas, and heterotopic pancreatic tissue.2 Malignant causes are often metastases or primary small-bowel tumor.3 About 20% of all cases are idiopathic. With the growing popularity of Roux-en-y gastric bypass for morbid obesity, intussusception in pregnant women after this operation is well documented.4,5 Otherwise, intussusception in pregnancy is rare and mostly spontaneous and without leading point revealed in pathology.6 This case is another reminder that prudent and systemic assessment is required for every pregnant woman with abdominal pain.
Surgery
A healthy 29-year-old pregnant woman, with 34 weeks’ gestational age, presented to the emergency department with diffused acute abdominal pain and vomiting. The pain was located in the periumbilical region and intensified within a few hours. The patient felt normal fetal movements during this time. An initial surgical evaluation was done, and the cause for the abdominal pain was assumed to be not of surgical condition. Next, the patient was evaluated by an obstetrician-gynecologist, and although the gynecologic examination, ultrasonography, and monitoring were all normal, the patient was admitted for further supervision in the obstetrics department. The next day, when the abdominal pain and vomiting persisted and an elevated leukocyte blood count of 14 200/mL was noted (to convert to ×109 per liter, multiply by 0.001), a second surgical consultation was done. The physical examination was significant for diffuse abdominal tenderness. Although the patient was pregnant, and owing to nonavailability of magnetic resonance imaging at that time, a contrast-enhanced abdominopelvic computed tomography was performed (Figure 1).Coronal computed tomographic image showing an edematous small intestinal loop on the left abdomen, with uterus and head of fetus in the pelvis.
what is your diagnosis?
What is your diagnosis?
Foreign body ingestion
Ileo-colic intussusception
Crohn ileitis
Adenocarcinoma of small intestine
b
1
1
0
1
female
1
0
29
21-30
null
598
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2714696
A 54-year-old man with a medical history of poorly controlled diabetes and hepatitis C–associated liver failure presented with a new central scotoma of the right eye 3 days after orthotopic liver transplant. He denied eye pain, floaters, and photopsias. The patient had his first liver transplant 12 years before presentation that failed despite immunosuppressive treatment with tacrolimus. He initiated mycophenolate mofetil treatment 2 months before his second transplant. In addition, the patient was positive for cytomegalovirus (CMV) IgG antibodies and had an undetectable CMV load 1 month before transplantation.On examination, visual acuities were hand motions OD and J1 OS. The patient’s right pupil was sluggishly reactive with a relative afferent pupillary defect. Intraocular pressure was 16 mm Hg OD and 18 mm Hg OS. Extraocular movements were intact, and visual fields were full by confrontation. Anterior segment examination was within normal limits. Fundus photographs of the right eye are shown in the Figure, A. Findings of fundus examination of the left eye were normal.Fundus photograph of the right eye at presentation (A) and at 12 months (B).Perform a vitreous tap and administer intravitreal injection of antifungal agentsPerform a vitreous tap and administer intravenous ganciclovir treatment What Would You Do Next?
Order brain neuroimaging and lumbar puncture
Perform a vitreous tap and administer intravitreal injection of antifungal agents
Perform a vitreous tap and administer intravenous ganciclovir treatment
Administer systemic steroid treatment
Presumed Aspergillus choroiditis
B
Perform a vitreous tap and administer intravitreal injection of antifungal agents
In an immunosuppressed patient after organ transplant, the presence of a large, creamy macular lesion with hemorrhage is highly suggestive of an infectious cause, particularly a fungus. The differential diagnosis includes Aspergillus, Candida, or bacterial endophthalmitis, CMV retinitis, and Toxoplasma retinochoroiditis.1Of the available management options, intravitreal injections of antifungal agents (choice B) are most likely to successfully treat the infection. Neuroimaging and lumbar puncture (choice A) may be useful to assess for central nervous system infection; however, this option does not address the patient’s acute eye disease. Although CMV infection should be considered in an immunosuppressed patient with chorioretinitis, the morphologic features of the lesion were not consistent with CMV infection; thus, intravenous ganciclovir treatment (choice C) would not be helpful. Systemic steroids would be an inappropriate choice for a patient with suspected infection (choice D).Approximately 60% of all cases of endogenous endophthalmitis are fungal in origin, with most cases caused by Candida species; however, Aspergillus endophthalmitis may be more common among orthotopic liver transplant recipients.2,3Aspergillus endophthalmitis classically presents as a confluent, yellowish macular infiltrate in the choroid and subretinal space. Retinal hemorrhages, vascular occlusions, and necrosis with subretinal or subhyaloid hypopyon can develop. An atrophic scar remains after the infection is treated. Compared with Candida lesions, Aspergillus lesions tend to be larger and invade the retinal and choroidal vessels, leading to hemorrhage and large areas of ischemia.3 Conversely, the primary site of infection in Candida endophthalmitis is the vitreous.3 Diagnosis is based on clinical findings and can be supported with results of a vitreous and/or blood culture. Aspergillus species are often difficult to grow from blood culture, with less than 10% of suspected cases having positive culture results, compared with Candida species, with about 50% of cases having positive culture results.4 In cases in which there is a high suspicion for fungal infection, testing for β-D-Glucan, a commonly encountered antigen in fungal infections, may be a useful diagnostic adjunct.4 In this case, although results of fungal cultures were negative, the clinical findings were suggestive of Aspergillus choroiditis; response to targeted therapy helped confirm this diagnosis.The patient underwent a vitreous tap and received amphotericin B, 10 µg/0.1 mL. Results of bacterial and fungal cultures and viral polymerase chain reaction (herpes simplex virus, varicella-zoster virus, and CMV) were negative. Gram staining showed no organisms. Results of serum testing for syphilis, toxoplasmosis, bartonella, and histoplasmosis were negative, whereas the result of a test for β-D-Glucan was positive.The patient was treated with systemic antifungals and weekly injections of intravitreal voriconazole, 50 µg/0.1 mL. His fundus lesions developed into well-demarcated scars without evidence of active inflammation (Figure, B). Approximately 12 months after presentation, the patient’s vision marginally improved to counting fingers at 3 ft OD, and systematically he was doing well; he continued treatment with daily systemic antifungals.
Ophthalmology
A 54-year-old man with a medical history of poorly controlled diabetes and hepatitis C–associated liver failure presented with a new central scotoma of the right eye 3 days after orthotopic liver transplant. He denied eye pain, floaters, and photopsias. The patient had his first liver transplant 12 years before presentation that failed despite immunosuppressive treatment with tacrolimus. He initiated mycophenolate mofetil treatment 2 months before his second transplant. In addition, the patient was positive for cytomegalovirus (CMV) IgG antibodies and had an undetectable CMV load 1 month before transplantation.On examination, visual acuities were hand motions OD and J1 OS. The patient’s right pupil was sluggishly reactive with a relative afferent pupillary defect. Intraocular pressure was 16 mm Hg OD and 18 mm Hg OS. Extraocular movements were intact, and visual fields were full by confrontation. Anterior segment examination was within normal limits. Fundus photographs of the right eye are shown in the Figure, A. Findings of fundus examination of the left eye were normal.Fundus photograph of the right eye at presentation (A) and at 12 months (B).Perform a vitreous tap and administer intravitreal injection of antifungal agentsPerform a vitreous tap and administer intravenous ganciclovir treatment
what would you do next?
What would you do next?
Perform a vitreous tap and administer intravenous ganciclovir treatment
Perform a vitreous tap and administer intravitreal injection of antifungal agents
Administer systemic steroid treatment
Order brain neuroimaging and lumbar puncture
b
0
0
0
1
male
0
0
54
51-60
null
599
original
https://jamanetwork.com/journals/jamaophthalmology/fullarticle/2717487
A woman in her late 50s reported right eye visual field changes. Review of medical history revealed lumpectomy and radiotherapy for breast cancer 6 years prior. Visual acuity was 20/160 OD (her baseline due to amblyopia) and 20/20 OS. Pupillary function was normal. The anterior chamber and vitreous were quiet. Fundus photographs showed optic nerve edema and a peripapillary choroidal infiltrate in the right eye. Swept-source optical coherence tomography (OCT) demonstrated retinal thickening with cystoid abnormalities, subretinal hyperreflective material consistent with fibrosis, and choroidal thickening in the right eye (Figure 1). Optical coherence tomography of the optic nerve head in the right eye demonstrated 360° peripapillary nerve fiber thickening and diffuse nerve head elevation. Fluorescein angiography demonstrated staining with minimal leakage in the temporal peripapillary retina of the right eye.A, Fundus photography of the right eye revealed optic nerve edema and a peripapillary choroidal infiltrate. The line marks the location of the cut through the retina represented by the optical coherence tomography (OCT). B, Swept-source OCT of the right eye demonstrated retinal thickening with cystoid abnormalities, subretinal fibrosis, and choroidal thickening in the area marked on the fundus photo.Testing results for an underlying neoplastic, inflammatory, or infectious process were unrevealing, including negative results of serology tests for tuberculosis, toxoplasmosis, syphilis, Lyme disease, and sarcoidosis. Results of complete blood cell count, serum protein electrophoresis, and bone marrow biopsy were not supportive of lymphoproliferative disease. Results of magnetic resonance imaging of the brain and orbits were normal. Computed tomographic scan of the chest, abdomen, and pelvis demonstrated no lymphadenopathy.Four months prior to presentation, routine electrocardiogram demonstrated a right bundle branch block. Eight months later, complete heart block necessitated urgent pacemaker implantation. What Would You Do Next?
Chorioretinal biopsy
Observation
Referral for cardiac biopsy
Oral corticosteroid therapy
Choroidal infiltrate secondary to giant-cell myocarditis
C
Referral for cardiac biopsy
Giant-cell myocarditis (GCM) is a rare, rapidly progressive, and frequently fatal cardiac disease that typically affects young, healthy patients. It is characterized by a granulomatous inflammation causing infiltrative myocardial disease and manifests as conduction abnormalities, heart failure, and even sudden cardiac death.1 The cause is unknown and likely multifactorial, with implication of infectious, genetic, and autoimmune processes. Giant-cell myocarditis has been associated with underlying autoimmune disorders in 20% of patients.2Ocular manifestations of GCM are rare and limited to case reports of orbital myositis.3-7 In some cases the orbital myositis indicated the underlying disease. Few patients with GCM have had detectable anticardiac antibodies, including anticardiac myosin, which supports the idea that GCM is an immune-mediated process.8 Investigation of extraocular muscle myosin subtypes has revealed the presence of cardiac myosin, providing a plausible molecular mechanism for the concomitant orbital inflammation.9 Furthermore, in the choroid, immunohistochemistry studies have demonstrated the presence of myosin in nonvascular smooth muscle cells.10The most likely disorders associated with choroidal infiltrate include sarcoidosis, tuberculosis, syphilis, toxoplasmosis, or lymphoma. Workup revealed no evidence for these as the underlying cause. The patient underwent a cardiac evaluation for her complete heart block. Depressed systolic function on echocardiography and normal coronary artery perfusion on catheterization raised suspicion for an infiltrative cardiac process. Endomyocardial biopsy was performed and demonstrated lymphocytic infiltrate, granulomatous inflammation, and extensive myocardial destruction in a pattern consistent with GCM (Figure 2). Because ocular involvement was limited to the macula and optic nerve, it was believed that the lesion was not amenable to chorioretinal biopsy. Observation alone was inappropriate given the vision and possibly life-threatening implications of this clinical finding. Initiation of corticosteroids prior to cardiac biopsy could lead to a delay in definitive diagnosis and appropriate treatment.Endomyocardial biopsy demonstrated destruction of the myocardium with a mixed inflammatory infiltrate of macrophages, eosinophils, and lymphocytes consistent with giant-cell myocarditis (hematoxylin-eosin, original magnification ×600).The patient eventually received treatment with prednisone and cyclosporine and had resolution of the intraretinal cystoid abnormalities, decreased optic nerve head swelling, and return of the choroid to normal thickness. This improvement correlated well with the stabilization of cardiac function. While we could not confirm histologically that the ocular inflammation and myocardial inflammation shared a common cause, it seems likely given the concurrence of symptom onset and parallel response to therapy.This instance of choroidal infiltration may represent a rare manifestation of GCM and a similarly rare underlying cause of a choroidal infiltrative process. It provides an example of potential benefits of systemic evaluation in patients presenting with ocular inflammatory disease. Prior to presentation of visual symptoms, the newly detected right bundle branch block on routine electrocardiogram and need for urgent pacemaker implantation for complete heart block were the clues to the underlying disease.The patient has remained clinically stable with more than 1.5 years of follow-up during immunosuppressive therapy with cyclosporine and low-dose oral corticosteroid therapy.
Ophthalmology
A woman in her late 50s reported right eye visual field changes. Review of medical history revealed lumpectomy and radiotherapy for breast cancer 6 years prior. Visual acuity was 20/160 OD (her baseline due to amblyopia) and 20/20 OS. Pupillary function was normal. The anterior chamber and vitreous were quiet. Fundus photographs showed optic nerve edema and a peripapillary choroidal infiltrate in the right eye. Swept-source optical coherence tomography (OCT) demonstrated retinal thickening with cystoid abnormalities, subretinal hyperreflective material consistent with fibrosis, and choroidal thickening in the right eye (Figure 1). Optical coherence tomography of the optic nerve head in the right eye demonstrated 360° peripapillary nerve fiber thickening and diffuse nerve head elevation. Fluorescein angiography demonstrated staining with minimal leakage in the temporal peripapillary retina of the right eye.A, Fundus photography of the right eye revealed optic nerve edema and a peripapillary choroidal infiltrate. The line marks the location of the cut through the retina represented by the optical coherence tomography (OCT). B, Swept-source OCT of the right eye demonstrated retinal thickening with cystoid abnormalities, subretinal fibrosis, and choroidal thickening in the area marked on the fundus photo.Testing results for an underlying neoplastic, inflammatory, or infectious process were unrevealing, including negative results of serology tests for tuberculosis, toxoplasmosis, syphilis, Lyme disease, and sarcoidosis. Results of complete blood cell count, serum protein electrophoresis, and bone marrow biopsy were not supportive of lymphoproliferative disease. Results of magnetic resonance imaging of the brain and orbits were normal. Computed tomographic scan of the chest, abdomen, and pelvis demonstrated no lymphadenopathy.Four months prior to presentation, routine electrocardiogram demonstrated a right bundle branch block. Eight months later, complete heart block necessitated urgent pacemaker implantation.
what would you do next?
What would you do next?
Chorioretinal biopsy
Observation
Referral for cardiac biopsy
Oral corticosteroid therapy
c
1
1
1
1
female
0
1
58
51-60
null
600
original