Tài liệu Infectious Diseases of the Head and Neck pptx

33 652 0
Tài liệu Infectious Diseases of the Head and Neck pptx

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

Microbiology and Infectious Disease / HEAD AND NECK INFECTIONS Infectious Diseases of the Head and Neck A Review Kathleen T Montone, MD Key Words: Infectious diseases; Head and neck; Bacteria; Fungus; Protozoa; Virus DOI: 10.1309/6BBT12WGNK77N4EH Abstract A variety of infectious diseases may involve head and neck structures These include bacterial, viral, fungal, and protozoal infections This article describes the pathologic features of a variety of infectious diseases that surgical pathologists may encounter in analysis of tissue specimens from the head and neck area Bacterial Infections Bacterial Sinusitis and Tonsillitis Acute and chronic sinus infections are most commonly caused by bacterial organisms, including Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, Staphylococcus aureus, other streptococcal strains, and anaerobic bacteria.1-6 These infections may manifest acutely and are most often treated with antibiotics without need for surgery Immunosuppressed patients are at risk for other bacterial infections, such as Pseudomonas species ❚Image 1❚ Patients with repeated infections may develop chronic sinusitis (infections >12 weeks in duration), which may require surgical intervention to open the sinonasal passageways Histologically, the sinonasal mucosa in chronic sinusitis shows chronic inflammation, fibrosis, vascular congestion, stromal edema, and cystic dilatation of the submucosal glands.4-6 Sinusitis with an allergic cause may show numerous eosinophils In addition, there may be basement membrane thickening, goblet cell hyperplasia, and dystrophic calcification.6 True sinonasal inflammatory polyp formation may be observed The bony tissue may show reactive changes Histologic changes similar to those seen in chronic osteomyelitis have been reported.7-9 Although bacteria are considered the cause of chronic sinusitis, the exact mechanism of infection has yet to be determined One study suggested that direct invasion of the mucosa by the bacterial pathogens may not be the mechanism of infection.10 In addition, a recent study implicated the nasopharyngeal reflux of gastric acid as a major contributing factor in the development of chronic sinusitis refractory to therapy.11 The presence of bacterial biofilms on sinonasal Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 35 DOI: 10.1309/6BBT12WGNK77N4EH 35 35 Montone / HEAD AND NECK INFECTIONS A B ❚Image 1❚ Acute bacterial sinusitis A, Acute necrotizing sinusitis in an immunosuppressed patient with acute lymphocytic leukemia (H&E, ×25) B, Numerous gram-negative bacteria are demonstrated (Gram, ×100) Cultures grew Pseudomonas aeruginosa mucosa may help explain disease that is refractory to antibiotic therapy in many patients.12,13 Acute tonsillitis is usually due to infection by Streptococcus species.14 The disease is characterized by throat pain and fever Most cases with a bacterial cause are treated with antibiotics, and surgery is unwarranted Complications include abscess formation, cellulitis, scarlet fever, acute rheumatic fever, and poststreptococcal glomerulonephritis.14 When tonsillitis becomes chronic, there is often hypertrophy of the tonsillar tissue, which is histologically characterized by follicular hyperplasia There are controversies about the benefit of examination of routine tonsillar specimens in surgical pathology.15,16 Cat-Scratch Disease Cat-scratch disease is a self-limited infection caused by the gram-negative bacteria Bartonella henselae, the same organism that causes bacillary angiomatosis (BA).17-21 Cervical lymph nodes are most commonly affected Symptoms vary from mild to severe and include malaise, anorexia, and localized lymphadenopathy Most cases are associated with a cat scratch or bite, but the disease has also been linked to scratches caused by other animals and foreign objects.17,18,22 Males are more commonly affected than females, and disease is almost always identified in patients younger than 21 years About to 10 days after exposure, a raised skin lesion develops in the area of inoculation and is often followed by painful, regional lymphadenopathy, fever, malaise, anorexia, weight loss, headache, splenomegaly, pharyngitis, and/or a maculopapular rash.17,18,22 Atypical features include conjunctivitis, encephalitis, cranial and 36 36 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH peripheral nerve palsies, thrombotic thrombocytopenic purpura, osteomyelitis, hepatosplenomegaly, erythematous skin disease, and pneumonia.17,18,23 The clinical differential diagnosis includes most causes of infectious lymphadenitis such as mycobacterial infections, lymphogranuloma venereum, tularemia, brucellosis, infectious mononucleosis, syphilis, toxoplasmosis, and fungal infections, as well as sarcoidosis, lymphoma, drug reactions, and collagen vascular diseases If the clinical picture fits and serologic data are consistent, lymph node biopsy is usually not performed, and the patient is treated for presumed cat-scratch disease If a biopsy is performed, the lymph node often shows focal areas of necrosis with neutrophils surrounding germinal centers ❚Image 2❚ Organisms may be seen within histiocytes and in necrotic areas and thrombosed blood vessels Necrotizing granulomas and multinucleated giant cells may be seen The gram-negative organisms are often seen with Warthin-Starry stain or Brown-Hopps modified Gram stain.17 Organisms may be difficult to find because of the background and nonspecific staining seen with histochemical stains, and additional ancillary techniques such as serum titers, immunohistochemical analysis, and polymerase chain reaction (PCR) may be used for diagnosis.21,24-28 Treatment is usually supportive; however, if the symptoms are severe, antibiotics may be used The disease usually lasts to months but may persist for as long as years The disease usually resolves without sequelae Although immunosuppressed patients may be at risk for the development of disseminated disease,23,29 antibiotic therapy can eradicate the organism without sequelae © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE Bacillary Angiomatosis BA is a systemic disease caused by B henselae and Bartonella quintana.22 The disease, which was first described in 1983, is usually seen in immunosuppressed people, especially those infected with HIV, although cases have been reported in immunocompetent people.22,30-33 BA is characterized by the presence of solitary or multiple, tender, red papular or nodular skin lesions usually involving the face and extremities Postmortem examination may show similar lesions involving the brain, bone, lung, lymph nodes, liver (peliosis hepatis), spleen, and mucosal and peritoneal surfaces.30,34 There are skin lesions characteristically seen with BA22,31: (1) nodules resembling lobular capillary hemangioma (pyogenic granuloma); (2) nodules similar to those seen in Kaposi sarcoma; (3) dry, scaly, hyperkeratotic plaque lesions; and (4) subcutaneous nodules More recent studies have also described the presence of pseudoangiomatous hyperplasia in patients with BA.35 Although BA and cat-scratch disease are caused by the same organism,36 the histologic features are quite different Histologic examination of BA lesions shows ectatic vessels lined by plump, cuboidal endothelial cells that may show marked pleomorphism and mitotic activity and, therefore, may be confused with malignancy22 ❚Image 3A❚ The presence of neutrophils and fibrin adjacent to blood vessels may aid in the diagnosis ❚Image 3B❚ The organisms are best observed with Warthin-Starry stain or Grocott-methenamine silver stain The diagnosis can be confirmed by immunohistochemical analysis or PCR-based studies.36-38 Similar lesions may be A ❚Image 2❚ Cat-scratch disease Necrotizing lymphadenitis due to cat-scratch disease (H&E, ×50) Serology studies confirmed the diagnosis of Bartonella henselae encountered in the oral mucosa, lymph nodes, liver, spleen, bone marrow, larynx, gastrointestinal tract, peritoneum, diaphragm, lung, and central nervous system.31 Mycobacterial Lymphadenitis Mycobacterial cervical lymphadenitis is most often seen in immunocompromised people In adults, more than 90% of cases of mycobacteria-induced lymphadenitis are due to B ❚Image 3❚ Bacillary angiomatosis (BA) A, Skin biopsy specimen from an HIV+ male with BA Note the dermal vascular proliferation with surrounding inflammatory response (H&E, ×25) B, Higher power view of BA in an HIV+ male The vascular proliferation is characterized by the presence of plump, cuboidal endothelial cells and is surrounded by inflammatory cells, especially neutrophils (H&E, ×100) This finding aids in making the diagnosis of BA Glass slides for photographing provided by Rosalie Elenitsas, MD, Dermatopathology, University of Pennsylvania Medical Center, Philadelphia Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 37 DOI: 10.1309/6BBT12WGNK77N4EH 37 37 Montone / HEAD AND NECK INFECTIONS Mycobacteria tuberculosis (MTB; scrofula), whereas in children, more than 90% of cases are due to atypical mycobacteria.39-41 Although the incidence of MTB in the United States is lower than in other countries, it is increasing owing to immigration from endemic countries and urban crowding Lymphadenitis is seen in about 5% of immunocompetent patients infected with MTB, with cervical lymph node involvement being seen most commonly.39-41 Clinically, patients with scrofula usually seek care because of a painless enlarging neck mass, fever, chills, weight loss, and malaise Lymphadenitis usually involves anterior cervical nodes.40 Nontuberculous mycobacterial infections usually manifest with a long-standing neck mass without systemic signs and symptoms.40 The clinical differential diagnosis is quite broad and includes congenital disorders such as branchial cleft cyst, thyroglossal duct cyst, cystic hygroma, a variety of infectious diseases, and malignancies The diagnosis can be made by fine-needle aspiration (FNA) biopsy with cytologic examination of the biopsy material.42-44 In addition, the FNA can supply material for culture and PCR, which may be used to rapidly identify the species of the mycobacteria.42-45 PCR can also be performed on formalin-fixed, paraffin-embedded tissue samples.46,47 Histopathologically, the resected lymph nodes characteristically show necrotizing granulomas, often associated with giant cells; however, a variety of histopathologic changes have been described, including the presence of well-formed granulomas with little necrosis; well-formed granulomas with central necrosis; poorly formed granulomas with a necrotic background; and a mixed histiocytic, lymphocytic, and neutrophilic infiltrate with necrosis and without granuloma formation48,49 ❚Image 4❚ Mycobacterial cervical lymphadenitis Cervical lymph node showing necrotizing granulomatous inflammation (H&E, ×50) 38 38 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH ❚Image 4❚ Organisms may be seen by acid-fast stains such as Ziehl-Neelsen, Kinyoun, and auramine O, as well as Dieterle stain.50 However, histopathologic examination cannot effectively type mycobacteria, and ancillary studies, such as culture or PCR, are necessary for accurate species identification Features that have been used to distinguish nontuberculous mycobacterial lymphadenitis from tuberculous lymphadenitis include the presence of microabscesses, ill-defined granulomas, noncaseating granulomas, and fewer giant cells in the former,48,49 although culture or molecular testing is still considered the best means for diagnosis Rhinoscleroma Rhinoscleroma (“hard nose”) is a chronic, self-limited, granulomatous disease that involves the nasal cavity and other structures of the upper respiratory tract The disease is caused by Klebsiella rhinoscleromatis, a gram-negative organism that is endemic to Central America, Egypt, Africa, India, and Indonesia51,52 and rarely seen in the United States The organism is contracted by direct inhalation of contaminated material during a prolonged period.51,52 The pathogenesis of the disease is basically unknown, but infection is associated with crowded conditions and poor hygiene Rhinoscleroma most commonly involves the nasal cavity but may also affect the larynx, nasopharynx, oral cavity, paranasal sinuses, soft tissues surrounding head and neck structures, and, rarely, the orbit.51-57 Young females are affected more commonly than are males Clinically, patients have inflammatory polyps that involve the nasal septum and spare the sinuses.51,52,58 Other features include rhinorrhea, epistaxis, dysphagia, nasal deformities, oral anesthesia, stridor, dysphonia, and anosmia.51,52 Patients rarely have a Rosai-Dorfman type of reaction in regional lymph nodes.59 There are stages of rhinoscleroma51,52: (1) the catarrhal or atrophic stage in which patients have a nonspecific rhinitis that develops into a purulent nasal discharge with mucosal crusting; can last weeks to months; (2) the granulomatous or hypertrophic stage in which there is development of nasal polyps, bleeding, nasal enlargement, and nasal cartilage destruction; and (3) the sclerotic or fibrotic stage in which the inflammatory changes undergo fibrosis; can result in significant facial deformities The diagnosis can be made by culturing the organism; however, only 50% of lesions will be positive.52 The diagnosis can also be made by cytologic examination of smears of nasal brushing material, which may demonstrate the characteristic large, vacuolated Mikulicz cells that are histiocytes containing the organisms Tissue biopsies also reveal distinctive findings In the catarrhal stage, there may be squamous metaplasia and a nonspecific neutrophil infiltrate along with granulation tissue In the granulomatous stage, a lymphoplasmacytic infiltrate with Russell bodies, pseudoepitheliomatous © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE hyperplasia of the mucosa, and groups of large vacuolated histiocytes (Mikulicz cells) containing the bacterial organisms are seen ❚Image 5❚ If numerous, the bacteria can be seen by H&E staining, but Gram, periodic acid–Schiff (PAS), silver, or immunohistochemical staining may be required to confirm the diagnosis.60,61 In the sclerotic stage, extensive fibrosis may lead to stenosis and disfigurement Diagnostic cells may be difficult to identify in this stage The differential diagnosis includes other infectious granulomatous diseases caused by bacteria (eg, tuberculosis, actinomycosis, syphilis, and leprosy), fungi (eg, histoplasmosis, blastomycosis, paracoccidioidomycosis, and sporotrichosis), and parasites (eg, mucocutaneous leishmaniasis) Noninfectious processes to be considered include inflammatory conditions, such as Rosai-Dorfman disease and Wegener granulomatosis, and neoplastic processes, particularly lymphoma The disease is treated with long-term antibiotic therapy Leprosy Leprosy is a chronic granulomatous disease involving skin and peripheral nerves that is caused by Mycobacterium leprae.62,63 The disease was described in biblical times, and the incidence peaked in Europe in the 13th century The infection usually affects superficial peripheral nerves, skin, mucous membranes, urinary tract, the eyes, and the testes, and disease manifestations are a result of the inflammatory reaction to the organism leading to scarring and deformities Involvement of the nasal cavity and paranasal sinuses may manifest before the clinical skin lesions.64 A Leprosy is a spectrum of disease ranging from a localized, deforming, self-limited process (tuberculoid leprosy [TL]) to systemic disease that, left untreated, may be fatal (lepromatous leprosy [LL]).65-67 Between these stages, there are several borderline forms of the disease (borderline tuberculoid, midborderline, and borderline lepromatous) The disease may change from one stage to another with treatment Treatment often moves the disease process toward TL, whereas decreased immunosuppression can move borderline or TL toward LL Most cases of leprosy are concentrated in Brazil, India, Madagascar, Mozambique, and Nepal.61,62,66,67 In the United States, leprosy is rare but is found in parts of Florida, Louisiana, and Texas, areas of New York City, and Asian and Mexican communities in California.61,62,66,67 In TL, there is usually a large, ill-defined, numb skin plaque The plaque may be seen anywhere on the skin but often spares the scalp Spontaneous resolution may take years, resulting in scars and depigmentation Progression can also occur, leading to borderline-type leprosy, and, in rare cases, untreated patients can develop LL TL commonly involves nerves, and patients develop tender, thickened nerves with loss of sensation and motor function In LL, there is often no sensation loss and there is absence of nerve thickening LL can result in nodules or plaques on the skin Eye involvement causes pain, photophobias, decreased visual acuity, glaucoma, and blindness Laryngeal involvement results in stridor and hoarseness Nasal disease can cause a saddle-nose deformity, and nasal endoscopy has become a way in which patients can be followed up.64 LL cannot revert to less severe forms of the disease B ❚Image 5❚ Rhinoscleroma A, Florid phase of rhinoscleroma in a 25-year-old woman Note the presence of a mixed inflammatory cell infiltrate consisting of lymphocytes, plasma cells, and neutrophils, as well as the presence of histiocytes with clear cytoplasm (Mikulicz cells) (H&E, ×50) B, Higher power view of Mikulicz cells in the florid phase of rhinoscleroma (H&E, ×100) The infection is most easily diagnosed in this phase of the disease Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 39 DOI: 10.1309/6BBT12WGNK77N4EH 39 39 Montone / HEAD AND NECK INFECTIONS A B ❚Image 6❚ Leprosy A, Low-power view of tuberculoid leprosy (TL) showing a superficial and deep dermal inflammatory infiltrate consisting of granulomas (H&E, ×5) B, Higher power view of TL showing multiple granulomas (H&E, ×50) In this form of leprosy, there is nerve destruction by the granulomatous disease Only rare organisms are seen in tissue sections C, Lowpower view of lepromatous leprosy (LL) showing a superficial and deep dermal inflammatory infiltrate (H&E, ×12.5) D and E, Higher power view of LL showing numerous histiocytes with clear cytoplasm (Virchow, or lepra, cells) (D, H&E, ×100; E, H&E, ×100) This form of leprosy spares nerves and rarely shows granulomas F, Tissue stain showing acid-fast bacillus (AFB)-positive organisms in a patient with LL confirmed by culture (AFB, ×315) Glass slides for photographing provided by Rosalie Elenitsas, MD, Dermatopathology, University of Pennsylvania Medical Center, Philadelphia The diagnosis may require skin or nerve biopsy Histologic findings vary according to the type of leprosy.62 In TL, noncaseating granulomas that destroy dermal nerves are present Skin appendages are lost, and organisms are difficult to identify ❚Image 6A❚ and ❚Image 6B❚ In LL, the epidermis is normal, and a Grenz zone separates the epidermis from a diffuse dermal inflammatory reaction consisting of macrophages, foamy histiocytes (Virchow, or lepra, cells), and many intracellular organisms ❚Image 6C❚, ❚Image 6D❚, ❚Image 6E❚, and ❚Image 6F❚ Epithelioid cells and giant cells are not found Inflammation is most prominent around blood vessels, nerves, and skin appendages, but dermal nerve structure is preserved Assays have been developed for demonstration of M leprae–specific DNA and ribosomal RNA (rRNA) sequences in various specimens such as nasal smears, skin, and blood PCR for bacterial DNA is most sensitive and can detect fewer than 10 organisms.68,69 The majority of patients with suspected leprosy who have negative screening smears have positive PCR results In situ hybridization can be used to establish the diagnosis if necessary.69,70 With early diagnosis and treatment, progression of disease can be limited, but recovery of neuronal function is variable.66,67 Much of the chronic changes result from repeated trauma to numb extremities Even when disease is controlled, the stigma and social isolation often persist 40 40 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH Gonorrhea Gonorrhea is a sexually transmitted disease caused by Neisseria gonorrhoeae Infection may affect almost any mucous membrane In the head and neck, the most commonly involved areas include the pharynx and the conjunctiva.71,72 A significant number of patients have pharyngitis, which usually is asymptomatic but may cause discomfort that can be severe Conjunctivitis can occur in adults and in children following direct inoculation and can lead to blindness In neonates, bilateral conjunctivitis often follows vaginal delivery by an infected mother Blindness from neonatal gonococcal infection is a serious problem in developing countries but is uncommon in the United States where prophylaxis during the neonatal period is common Prognosis is excellent if therapy is initiated promptly Syphilis Syphilis is a sexually transmitted infectious disease caused by the spirochete Treponema pallidum It is almost always transmitted by sexual contact with infectious lesions, but it also can be transmitted in utero and through blood transfusion.73 T pallidum penetrates abraded skin and intact mucous membranes and rapidly disseminates through the lymphoreticular system The initial lesion of syphilis develops at the transmission site about 10 to 90 days after exposure and then heals © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE C D E F spontaneously in to weeks Secondary syphilis develops about to 10 weeks after the appearance of the primary lesion.73 Patients often complain of malaise, fever, myalgias, and arthralgias Physical examination may reveal lymphadenopathy and a maculopapular rash Secondary syphilis usually resolves without treatment and becomes latent In about 30% of patients in the latent stage, tertiary syphilis will develop, which may affect the heart and nervous system.73 All stages of syphilis may manifest with head and neck findings.74-78 The chancre sore of primary syphilis can involve oral mucosa.74-78 Patients with secondary syphilis may have mucosal erosions on the tongue, lips, and oral mucosa.74-78 Patients may also have hairy leukoplakia, oral lichen planus, oral erythema multiforme, alopecia, sore throat, headache, stiff neck pain, and optic neuritis Gummatous lesions of tertiary syphilis may involve mucous membranes, and in patients with neurosyphilis, meningitis and dementia may develop Oral lesions in primary and secondary syphilis are nonspecific and characterized by squamous hyperplasia and a plasma cell infiltrate that extends deep into the submucosa.74-78 Although these findings are nonspecific, the presence of a marked, deeply infiltrating plasma cell infiltrate should alert pathologists to the possibility of syphilis Neuritis has also been described in primary and secondary disease Mucosal ischemic changes due to obliterative endarteritis may also be seen Tertiary syphilis involving the oral cavity may also show a chronic inflammatory infiltrate with few plasma cells.74-78 Actinomycosis Actinomycosis is a subacute to chronic bacterial infection caused by filamentous, gram-positive, anaerobic to microaerophilic bacteria The bacteria include Actinomyces israelii, Actinomyces gerensceriae, Actinomyces naeslundii, Actinomyces odontolyticus, Actinomyces viscosus, Actinomyces meyeri, and Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 41 DOI: 10.1309/6BBT12WGNK77N4EH 41 41 Montone / HEAD AND NECK INFECTIONS Propionibacterium propionicum Actinomycosis is characterized by a suppurative and granulomatous inflammatory reaction and formation of multiple abscesses and sinus tracts Cervicofacial actinomycosis is the most common manifestation of infection.79-84 Patients with cervicofacial disease usually report a history of dental surgery, oral trauma, poor hygiene, or periodontal disease and painless soft tissue swelling involving the submandibular area Long-standing infection may be associated with multiple sinus tracts that have a tendency to heal and recur Patients have a nodular mass at the angle of the jaw The masses gradually increase in size and number and form sinuses that open onto the face and neck Although infection is often seen in the jaw area, Actinomyces infections have been described in other sites, including the sinuses, mastoid, major salivary glands, and thyroid.85-92 Histologically, infection is characterized by a mixed suppurative and granulomatous inflammatory reaction A Anthrax Bacillus anthracis is an aerobic, spore-forming, grampositive bacillus The organism has received worldwide attention because of its potential for use as a bioterrorism B ❚Image 7❚ Actinomyces infection A, Mandibular bone in a patient after radiation therapy for parotid gland mucoepidermoid carcinoma in whom a necrotic mandible showed Actinomyces osteomyelitis (H&E, ×100) B, Gram stain highlights the filamentous Actinomyces organisms (×100) C, Grocott silver stain also highlights the Actinomyces organisms (×100) C 42 42 “Sulfur granules” are for the most part pathognomonic for infection and can be found in the lesions and in the sinus drainage ❚Image 7A❚ and ❚Image 7B❚ The granules are approximately mm in diameter and can be seen by the naked eye as yellowish particles Microscopically, the particles are shown to consist of filamentous bacteria surrounded by neutrophils Gram stain reveals gram-positive, branching filaments, with radially arranged hyphae Treatment with antibiotics can eradicate the infection; however, surgery may be necessary for proper drainage Death may occur with infections that cannot be controlled with antibiotics or surgery Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE weapon There are major forms of infection with the organism: cutaneous, inhalation, and gastrointestinal.93 The details of these forms of anthrax will not be discussed with the exception of gastrointestinal anthrax, which may manifest with a localized oropharyngeal lesion that most likely is a result of colonization of the oropharyngeal mucosa by the organisms It is believed that the organism gains entrance into the mucosa through abrasions or possibly by spore-contaminated food products Localized ulcers, cervical lymphadenopathy, and localized edema may develop Histologically, the lesions show hemorrhage, edema, and necrosis with a neutrophilic infiltrate.93 Lymph nodes can show extensive necrosis and hemorrhage with the presence of gram-positive, boxcar-shaped, and encapsulated bacteria93 ❚Image 8❚ The organism can be cultured, and immunostains can be used to identify the organism in smears and tissue sections.94 A C Fungal Diseases A variety of fungal infections can result in head and neck manifestations Head and neck fungal infections have been associated with mucosal ulceration and erosion, lymphadenopathy, and sinonasal disease This section will not detail all types of fungal infections but will concentrate on sinonasal fungal disease Sinonasal Fungal Disease Sinonasal fungal disease was initially described in the 1600s; however, only recently has this topic become of great interest There are types of sinonasal fungal disease: (1) chronic indolent invasive sinusitis, (2) fulminant fungal sinusitis, (3) fungus ball, and (4) allergic fungal sinusitis (AFS).95-99 Chronic indolent invasive fungal sinusitis is of types: chronic invasive granulomatous sinusitis and chronic invasive fungal sinusitis.95-99 Chronic granulomatous sinusitis is seen in B ❚Image 8❚ Anthrax A, Lymph node in a patient with disseminated Bacillus anthracis at autopsy (H&E, ×50) Note the extensive hemorrhagic necrosis of the node This picture is typical for B anthracis infection B, B anthracis in a lymph node in the case mentioned in A (Giemsa, ×100) C, B anthracis The organisms are gram-positive, long slender rods (Gram, ×250) Photographs courtesy of Centers for Disease Control and Prevention/Marshall Fox, MD, via the Public Health Image Library Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 43 DOI: 10.1309/6BBT12WGNK77N4EH 43 43 Montone / HEAD AND NECK INFECTIONS immunocompetent patients, usually in northern India, Sudan, and North Africa The most common causative fungal organism is Aspergillus flavus Patients have proptosis caused by a granulomatous reaction to the fungus Treatment includes surgical debridement followed by antifungal therapy Chronic invasive fungal sinusitis is a slowly progressive, low-grade, invasive fungal infection that usually occurs in patients with diabetes Patients have symptoms of long-standing sinusitis and are usually not in acute distress Chronic invasive fungal sinusitis is most commonly associated with Aspergillus fumigatus Treatment consists of surgery followed by systemic antifungal therapy Fulminant (angioinvasive) fungal sinusitis is often seen in immunosuppressed patients, particularly patients with hematologic malignancies and diabetes, although occasionally the disease may develop in immunocompetent patients.95-99 The patients get infected with the fungus, and subsequently tissue and vascular invasion with massive necrosis develop, necessitating surgery and antifungal therapy ❚Image 9❚ Patients have fever, cough, nasal discharge, and mental status changes The organisms usually implicated are Aspergillus species and organisms in the Mucorales order (Rhizopus, Rhizomucor, Absidia, Mucor, Cunninghamella, Mortierella, Saksenaea, and Apophysomyces).95-99 Treatment includes emergency surgery with sinus debridement and intravenous antifungal therapy Despite therapy, the prognosis is poor A fungus ball is a tangled mass of fungus (most commonly A flavus or A fumigatus) in the sinonasal tract (most commonly the maxillary sinus) associated with minimal if any inflammatory reaction.95-102 Patients are usually healthy and nonatopic and have unilateral sinonasal blockage and pain and are treated with surgical debridement alone The fungus may be A related to a history of trauma or surgery Histologically, the specimen consists of a mass of fungal organisms Little inflammatory reaction is present ❚Image 10❚ Although the disease is usually indolent, reports have demonstrated that these lesions may develop into acute fulminant sinusitis in previously healthy patients who become immunosuppressed.103 AFS occurs in immunocompetent patients with nasal polyps, atopy, and asthma in whom an allergic immune response develops to extramucosal fungal antigens.95-99,104-111 The disease was first described in 1983 by Katzenstein et al,104 who presented a series of patients who had allergic symptoms, sinonasal contents with a histologic picture similar to that seen in allergic bronchopulmonary aspergillosis, and the presence of fungi histologically compatible with Aspergillus Since that time, several fungi have been implicated in the pathogenesis of this disease entity.112-114 AFS represents approximately 10% of all chronic sinusitis cases requiring surgery Studies have indicated that AFS may be more common than previously thought A group from the Mayo Clinic grew fungal organisms in washing specimens from 96% of patients with chronic sinusitis.115 However, this study has been criticized because the patients may not have fulfilled the diagnostic criteria for AFS.116 AFS can be seen in patients of any age or sex, and the diagnosis is suspected based on clinical manifestations and characteristic computed tomography scan or magnetic resonance imaging findings.95-99,104-111,117 Patients may have facial deformities and visual loss The diagnosis is confirmed by histologic examination of the sinus contents for the presence of “allergic mucin.” Fungal cultures and allergy testing may support the diagnosis Although the pathogenesis of AFS is not entirely understood, AFS is considered to represent a hypersensitivity reaction B ❚Image 9❚ Angioinvasive fungal sinusitis A, Sinonasal mucosa biopsy specimen in an immunosuppressed patient after treatment for acute lymphocytic leukemia There is extensive necrosis due to angioinvasive fungal infection histologically consistent with Aspergillus (H&E, ×25) B, Grocott silver stain confirms the presence of angioinvasive fungus (×50) 44 44 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE whereas others show systemic symptoms including fever, pharyngitis, and multisystem organ failure, findings similar to those seen in severe acute infectious mononucleosis In the head and neck area, PTLD can manifest almost anywhere and has been described in lymph nodes, tonsils, adenoids, oropharynx, thyroid, parathyroid, hard palate, parotid gland, nasopharynx, trachea, submandibular gland, lacrimal gland, and conjunctiva.179 Head and neck involvement has been reported as a cause of sudden respiratory compromise in children.184,185 Histopathologically, PTLDs can be classified in a variety of ways Nalesnik et al179 proposed classification based on the morphologic features of the infiltrate as polymorphous or monomorphous PTLD Polymorphous PTLD contains an infiltrate composed of a variety of cell types, including small and large lymphocytes, plasma cells, immunoblasts, and transformed lymphocytes ❚Image 20❚ These lesions, while polymorphous, can be polyclonal, oligoclonal, or monoclonal by immunohistochemical analysis or by analysis for immunoglobulin heavy or light chain gene rearrangements Monomorphous lesions are almost consistently monoclonal Patient outcome based on this pathologic classification is difficult to predict Some monoclonal lesions regress, whereas others progress to disseminated disease and death in a short time A newer classification scheme was reported by Harris et al.186 This system includes main categories: (1) early lesions: a proliferation of plasma cells and plasmacytic-type cells with minimal atypia; tendency to show scattered EBV+ cells by in situ hybridization; lesion often regresses with or A even without immunosuppression reduction, often polyclonal, no genetic abnormalities, and often not fatal; (2) polymorphous PTLD: a proliferation of lymphoid cells consisting of a mixture of small and large lymphocytes, plasma cells, and immunoblasts with cytologic atypia; individual cell and zonal necrosis often seen; frequent diffuse positivity for EBV by in situ hybridization; lesions show a spectrum from reactive lymphoid hyperplasia to polymorphous B-cell lymphoma, often monoclonal by gene rearrangement studies, not associated with genetic abnormalities, and almost always regresses; (3) monomorphous PTLD: a proliferation of atypical lymphoid cells of a single cell type, usually immunoblasts or plasmablasts, and histologically resemble immunoblastic lymphoma or plasmablastic myeloma; lesions clonal, possibly associated with genetic alterations (c-myc and p53 mutations), and have uniformly fatal outcome EBV can be detected in patients with PTLD by several methods.187-190 Serologic examination may reveal recent EBV infection or reactivation of past EBV infection Southern blot analysis using DNA probes specific for the EBV long terminal repeats can determine if the lymphoid proliferation is secondary to a clonal expansion of EBV-infected cells Rapid, accurate in situ localization techniques for EBV DNA, RNA, and proteins are easily performed in many pathology laboratories The treatment for PTLD is variable Most lymphoproliferations regress with reduction of immunosuppression or with no treatment Some studies have tried treatment with anti–B cell antibodies or with donor mononuclear cell infusions with mixed results.190,191 Studies have shown that reconstitution of B ❚Image 20❚ Posttransplantation lymphoproliferative disorder (PTLD) A, A tonsillar mass in a young male after kidney and pancreas transplantation Note the polymorphous atypical lymphoid infiltrate (periodic acid–Schiff, ×100) This lesion was clonal by immunoglobulin heavy chain gene rearrangements Following reduction of immunosuppression medications, the PTLD resolved B, In situ hybridization for Epstein-Barr virus small RNA is positive in many of the atypical lymphocytes (diaminobenzidine/hematoxylin counterstain, ×100) Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 53 DOI: 10.1309/6BBT12WGNK77N4EH 53 53 Montone / HEAD AND NECK INFECTIONS EBV-specific T-cell immunity using EBV-specific autologous cytotoxic T cells obtained before transplantation and infused after transplantation remained cytotoxic against EBV and may be able to reduce the incidence of PTLD.192 X-Linked Lymphoproliferative Disease (Duncan Disease).—X-linked lymphoproliferative disease is a disease caused by an inherited immunodeficiency state in which patients (mainly males) develop infectious mononucleosis, acquired hypogammaglobulinemia, or malignant lymphoma following EBV infection.164,180-182,193-197 Patients have a defect in the ability to regulate EBV infections; however, response to other infectious agents does not seem to be impaired The pathology is similar to that seen in EBV-associated PTLD and fatal infectious mononucleosis.164,180-182 About two thirds of patients die of fatal infectious mononucleosis An additional 20% will develop lymphoma if they survive the initial EBV infection About 85% of patients will die in childhood There is 100% mortality by 40 years of age Burkitt Lymphoma.—Burkitt lymphoma, a high-grade Bcell lymphoma, is another EBV-associated malignancy There are forms of the disease, the endemic (African) form and the nonendemic (sporadic) form.198 The endemic form is associated with a combination of EBV infection and malaria Recent studies have also implicated a third agent, mosquito-transmitted arbovirus, and arbovirally related illnesses have been described in patients before the onset of the lymphoma.199 The nonendemic form is often not EBV-related unless it is seen in immunosuppressed patients Burkitt lymphoma is one of the fastest growing malignancies The endemic form most commonly infects the jaw (maxilla or mandible) of children.198 Histologically, Burkitt lymphoma is characterized by a proliferation of small noncleaved B cells that are uniform in appearance and show a “starry sky” appearance, which is secondary to the presence of scattered tingible body macrophages due to the tumor’s high proliferative rate.200 In situ hybridization for EBV small encoded RNA, a marker of latent EBV infection, often highlights many cells in the endemic Burkitt lesions A study showed evidence of lytic EBV infection in the cells adjacent to the tingible body macrophages that induce the starry sky appearance.201 Most Burkitt lymphomas carry a t(8;14) that results in a fusion of the c-myc gene on chromosome with the immunoglobulin heavy chain gene on chromosome 14 Other gene rearrangements include t(8;2) (fusion with κ light chain gene) and t(8;22) (fusion with λ light chain gene).198 Although Burkitt lymphoma is considered a small cell lymphoma, it is often difficult to distinguish Burkitt lymphoma from diffuse large B-cell lymphoma based on pure histologic features This distinction is important because the treatment protocols are different Recent studies have used gene expression profiling to aid in this distinction Burkitt lymphoma specimens are more likely to have high expression of c-myc 54 54 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH target genes and germinal center B-cell genes compared with large cell lymphomas.202,203 Sinonasal NK/T-Cell Lymphoma.—Sinonasal NK/T-cell lymphomas (angiocentric lymphoma/CD56 lymphoma) are a group of high-grade, aggressive lymphomas that have a propensity to occur in the nose and paranasal sinuses.204,205 Other names for this tumor include lethal midline granuloma, polymorphic reticulosis, and midline malignant reticulosis, although the preferred terminology is sinonasal NK/T-cell lymphoma.204,205 The tumors are more commonly seen in men older than 40 years They peak during the fifth decade These tumors are uncommon in the United States, constituting fewer than 1% of lymphomas, but are common in Asia and Central and South America.204,205 Histologically, these tumors are composed of large atypical lymphocytes with enlarged, hyperchromatic, and convoluted nuclei admixed with smaller lymphocytes, plasma cells, and histiocytes The tumor cells often infiltrate large blood vessels, resulting in zones of tissue necrosis; however, this feature is evident in only 70% of tumors204 ❚Image 21A❚ and ❚Image 21B❚ Immunophenotypically, the lesional cells are positive for CD56, a marker of NK cell differentiation, CD2, CD7, and CD8 and are negative for CD3 and CD5.204,205 The tumors often express T-cell intracellular antigen-1 (TIA-1), perforin, and FAS (CD95/Apo1) ligand.206 T-cell receptor gene rearrangement studies are negative.204,205 The Epstein-Barr viral genome is almost always identified in lesional cells, indicating a primary role for this virus in disease pathogenesis207 ❚Image 21C❚ Mutations of p53 and c-kit have been described.208 In addition, alterations in chromosomes 1, 2, 12, 10, 13, and 17 have been reported.209 Localized disease may be treated with radiation, with the majority of patients showing a complete remission Disseminated disease may involve the skin, regional lymph nodes, lungs, and brain The prognosis for disseminated disease is poor Recent studies have found that adoptive transfer of EBV-specific cytotoxic T cells may be useful in treating patients with disseminated disease.210 Nasopharyngeal Carcinoma.—Nasopharyngeal carcinomas are another EBV-associated malignancy The World Health Organization (WHO) has classified nasopharyngeal carcinoma into categories: WHO-1, keratinizing squamous cell carcinoma; WHO-2, nonkeratinizing squamous cell carcinoma; and WHO-3, undifferentiated carcinoma This latter entity is almost always EBV-associated.198,211,212 Nasopharyngeal carcinoma has a bimodal age distribution A small peak is observed in late childhood, and a second peak occurs in people 50 to 60 years old People of Asian and North African descent are also more commonly affected In the United States, WHO-3 nasopharyngeal carcinoma is more commonly seen in black teenagers.211,212 The pathogenesis of the tumor is largely unknown, but EBV © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE A C infection coupled with overexpression of p53 and loss of p16 and p27 protein expression has been suggested.213 Nasopharyngeal carcinoma rarely comes to medical attention before it has spread to regional lymph nodes, and, in fact, diagnosis is often made initially on cervical lymph node biopsy Enlargement and extension of the tumor can result in nasal obstruction, bleeding, hearing loss, and cranial nerve palsies Endoscopic examination reveals a mass arising in the nasopharynx, most frequently in the fossa of Rosenmüller A paraneoplastic osteoarthropathy has been described in patients with widespread disease.214 Most cases of WHO-3 disease contain EBV nucleic acids within the malignant epithelial cells In fact, the presence of EBV-infected cells in a carcinoma metastatic to cervical lymph nodes is highly suggestive of a nasopharyngeal primary site.215 EBV expression has been shown to be an independent prognostic factor associated with better prognosis in some studies.216 B ❚Image 21❚ Nasal T-cell/natural killer (NK)-cell lymphoma A, Nasal NK-cell lymphoma showing a markedly atypical lymphoid reaction surrounded by zones of necrosis (H&E, ×25) B, Higher power view showing the lymphoid atypia of sinonasal NK-cell lymphoma (H&E, ×100) C, In situ hybridization for Epstein-Barr virus small RNA is positive in the lymphoma cells (diaminobenzidine/hematoxylin counterstain, ×100) Histologically, WHO-3 lesions are characterized by the presence of large atypical nucleolated epithelial cells that grow singly or in a nested pattern and are often surrounded by a lymphoplasmacytic infiltrate ❚Image 22❚ Recent studies have identified in situ nasopharyngeal carcinoma that seems to precede an invasive diagnosis by about to years EBV nucleic acids are identified in these noninvasive lesions.217,218 With a diagnosis of nasopharyngeal carcinoma, EBV titers, including IgA and IgG antibodies to the viral capsid antigen, should be performed These titers correlate with tumor burden and decrease with treatment They are an excellent means for monitoring response of the neoplastic process to therapy and for detection of tumor recurrence.219 Human Herpesvirus HHV-6 is a virus that most commonly infects the oropharynx in early childhood Disease is characterized Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 55 DOI: 10.1309/6BBT12WGNK77N4EH 55 55 Montone / HEAD AND NECK INFECTIONS A B ❚Image 22❚ Nasopharyngeal carcinoma A, Undifferentiated nasopharyngeal carcinoma in a 55-year-old man (H&E, ×50) B, In situ hybridization for Epstein-Barr virus small RNA is positive in the carcinoma cells The lymphocytes are negative (diaminobenzidine/hematoxylin counterstain, ×25) by a fever and rash (roseola).147,220-223 Latent virus seems to be in salivary gland and lymphoid tissues.224 Adults infected with HHV-6 may have a mononucleosis-like illness.221-223 The virus has been implicated in some cases of Hodgkin disease and angioimmunoblastic lymphadenopathy.220-223 Human Herpesvirus Infection with HHV-7 usually occurs during childhood.147,221-223 The most common manifestation of the infection is fever The virus has been identified in pharyngeal tissues in adults.224 There are no known associations with systemic disease processes Human Herpesvirus (KS Herpesvirus) HHV-8 is a γ herpesvirus that has been associated with a variety of neoplastic states, including KS, the plasma cell variant of Castleman disease, and effusion-based lymphomas.225-227 Primary infection with the virus is usually characterized by a maculopapular rash and long-standing fever Like other herpesviruses, the virus becomes latent following primary infection Recent studies have suggested lymph node as the site of latency.224 Reactivation of the virus can lead to the development of the aforementioned malignancies, the most common of which is KS Almost all cases of KS contain the HHV-8 genome KS frequently occurs with head and neck manifestations, including involvement of skin, mucosal surfaces, and lymph nodes There are variants of KS143,228: (1) classic, usually seen on the lower extremities of older men; (2) endemic, which usually occurs in Africa and involves skin and lymph nodes; (3) 56 56 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH transplant-related, in which the tumor is seen in solid organ recipients; and (4) AIDS-related, seen in patients with AIDS and highly aggressive and often disseminated Before treatment with antiviral therapy for HIV, many patients with AIDS died of KS KS manifests clinically and pathologically in a variety of ways143,228 ❚Image 23❚: The patch stage is characterized by the presence of a flat lesion that shows a proliferation of numerous vascular spaces within the dermis The slit-like vessels are present around preexisting blood vessels, skin adnexa, and dermal collagen bundles The neoplastic vessels are lined by plump, atypical endothelial cells Glomerular-like vascular proliferations may be evident Extravasated RBCs and hemosiderin may be present The differential diagnosis includes granulation tissue The plaque stage shows more prominent spindle cells that blend together with the slit-like vascular spaces The proliferation involves the superficial and deep dermis and subcutaneous tissue There are often hemosiderin deposition and PAS-positive hyaline globules The nodular stage shows a defined lesion characterized by prominent spindle cells surrounding slit-like vascular spaces RBC extravasation is prominent The features are more prominent than those in the plaque stage PAS-positive hyaline globules are evident Other appearances include lymphadenopathic, exophytic, infiltrative, ecchymotic, telangiectatic, keloidal, and cavernous-like.228 The first line of treatment in AIDS-related KS is the control of HIV replication With improvement of antiretroviral therapy, the incidence of AIDS-related KS has declined significantly.229 © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE A B C D E ❚Image 23❚ Human herpesvirus (Kaposi sarcoma [KS] virus) A, Gross appearance of a KS lesion on the gingiva of an HIV+ male Photograph courtesy of Centers for Disease Control and Prevention (CDC)/Sol Silverman, Jr, DDS, via the Public Health Image Library B and C, Skin biopsy specimen in an HIV+ male showing a dermal vascular proliferation invading among the dermal collagen bundle fibers consistent with KS (B, H&E, ×25; C, H&E, ×50) D, Numerous hyaline globules in KS (H&E, ×100) Photograph courtesy of CDC/Edwin P Ewing, MD, via the Public Health Image Library E, Immunohistochemical stain for human herpesvirus in KS showing nuclear staining in the vascular endothelial cells (diaminobenzidine/hematoxylin counterstain, ×50) Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 57 DOI: 10.1309/6BBT12WGNK77N4EH 57 57 Montone / HEAD AND NECK INFECTIONS Human Papillomavirus Infection Human papillomavirus has been implicated in a variety of papillomatous and malignant squamous proliferations, including those seen in the head and neck area such as sinonasal papillomas, laryngeal papillomas, and squamous cell carcinoma.230-234 The details of these lesions will not be discussed Protozoal Diseases Rhinosporidiosis Rhinosporidiosis is a chronic infection of the mucous membranes that typically involves the nose, nasopharynx, conjunctiva, and lacrimal ducts.235,236 The disease is caused by Rhinosporidium seeberi and usually manifests grossly as friable, hemorrhagic polyps.237 The infection is endemic to India, Sri Lanka, South America, and Africa, with the overwhelming majority of cases originating in India and Sri Lanka.235 The disease is rare in the United States, but infection has been reported in Texas and the Southeast.235 R seeberi has not been grown in culture, but with rRNA analysis, the organism has been established as a protistan parasite in the class Mesomycetozoea, which contains organisms that produce similar diseases in fish and amphibians.238,239 Rhinosporidiosis is typically limited to the mucosal epithelium Infection usually results from traumatic inoculation of the organism The organism locally replicates, and the immune response to the organism induces hyperplastic growth of the infected tissue ❚Image 24❚ Rhinosporidiosis Nasal polyp lesion demonstrating submucosal organisms consistent with Rhinosporidium seeberi (H&E, ×50) Photograph courtesy of Centers for Disease Control and Prevention/Martin Hicklin, MD, via the Public Health Image Library 58 58 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH Most patients have unilateral nasal obstruction or bleeding Gross examination reveals soft, friable “strawberry-like” polyps in the infected sites.237 Histologically, the sporangia of R seeberi are observed beneath the normal epithelium ❚Image 24❚ They are associated with inflammatory cells, including neutrophils, lymphocytes, plasma cells, multinucleated giant cells, and scattered granulomas The overlying epithelium is hyperplastic and shows hypervascularity At times, rhinosporidiosis may be confused with the changes seen in cylindrical cell papilloma (oncocytic schneiderian papilloma); however, the cysts seen in cylindrical cell papillomas are intramucosal, not submucosal as in rhinosporidiosis Diagnosis can also be made by FNA cytology of mass lesions.240 Treatment involves surgical excision of the lesions The prognosis is excellent, except in patients with disseminated disease.241,242 Leishmaniasis Leishmaniasis is a disease caused by the protozoa Leishmania, transmitted by the female sandfly There are many species, and the spectrum of disease is broad.243-245 There are forms of disease: (1) cutaneous, which is most common; (2) visceral; and (3) mucocutaneous, which is the rarest form In addition, infection may be subclinical In the United States, the disease is uncommon but is endemic in parts of southwest Texas.246 Most other cases in the United States are identified in immigrants from countries where the infection is endemic Infection in soldiers returning from the Middle East is being increasingly seen.247,248 More than million cases of cutaneous leishmaniasis are diagnosed annually in countries where sandflies are common, including Afghanistan, Brazil, Iran, Iraq, Peru, and Saudi Arabia.249 Visceral leishmaniasis is most common in Bangladesh, Brazil, India, Nepal, and Sudan.249 The organism’s life cycle begins in the gut of the sandfly, but the organism completes its life cycle in the host following the fly bite The sandfly inoculates the host with the organisms in the promastigote stage Once the organisms are inoculated, they are phagocytized by host macrophages The promastigotes then replicate as amastigotes and infect additional macrophages locally or, if the organisms have disseminated, systemically Cutaneous leishmaniasis is characterized by the presence of skin lesions on exposed areas, similar to leprosy The initial lesions are seen several weeks after inoculation Some cases of cutaneous disease may spontaneously resolve, or the disease can become debilitating, resulting in significant scarring and disfigurement Visceral leishmaniasis incubates for weeks to months before clinical manifestation The disease onset can be acute, subacute, or chronic It can be indolent and self-limited, but may also be disseminated and fulminant Symptoms of visceral leishmaniasis include weight loss, fever, leukopenia, hepatosplenomegaly, diarrhea, and lymphadenopathy.249,250 © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE The least common form of leishmaniasis is mucocutaneous The ulcerated mucosal lesions seen in this form of the disease are due to the disseminated spread of the organism from the initial site of infection The ulcers often become secondarily infected with bacteria The mucosal lesions can involve the oral mucosa, pharynx, lacrimal glands, nasal mucosa, larynx, and other sites.251-255 If untreated, there may be erosion of the nasal septum and the palate, resulting in facial disfigurement Cytologic evaluation of nasal smears can be used to evaluate treatment efficacy in patients with nasal involvement.256 In endemic countries where tests are not usually available, the diagnosis is made on a clinical basis If tissue specimens are obtained, the organism may be seen by H&E staining, especially in cases of cutaneous and visceral leishmaniasis Skin tests may reveal positive results months after the initial lesions are observed Special immunohistochemical assays and molecular genetic testing can be used to identify the species of the organism In cutaneous leishmaniasis, serum anti–leishmanial antibody can be detected; however, diagnosis is usually made by identification of the organisms in tissue or cytologic specimens.249 Material from the ulcer base usually has the highest yield Detection of the organism’s nucleic acids in lesional material by PCR is usually the most sensitive method for the diagnosis of cutaneous and mucocutaneous disease For visceral leishmaniasis, diagnosis is best made by direct visualization of organisms in clinical specimens.249 Serum anti–leishmanial IgG in high titer can also be diagnostic Tests for leishmanial antigen or antibody in the urine are newer techniques used for diagnosis of visceral disease.249 Immunohistochemical analysis and in situ hybridization have been used to identify the organisms in tissue samples.257 A Microscopically, the parasites appear as 1.5- to 3-µm ovoid to round organisms within histiocytes ❚Image 25❚ In human tissue, Leishmania organisms appear as amastigotes The organisms are identified on H&E and Giemsa stains on smears In tissue sections, Giemsa stains are not usually useful, but the organisms may be highlighted with Brown-Hopps modified Gram stain Differential diagnosis includes small fungi such as Histoplasma and Penicillium The complications of leishmaniasis include secondary bacterial infection of ulcerated lesions, bleeding, disfigurement, and, rarely, splenic rupture With early treatment, the majority of patients are cured, but untreated cases can be fatal Death is usually secondary to bacterial infection of ulcerated lesions Toxoplasmosis Toxoplasma gondii is an obligate intracellular parasite and one of the most common human parasites in the world.258 Cats are the definitive host for the organism There are forms of the parasite: oocysts, trophozoites (tachyzoites, rapidly proliferating; or bradyzoites, slowly proliferating), and tissue cysts The noninfectious oocysts are excreted by the cat, and, in the soil, if they are exposed to the right conditions, will grow into infectious oocysts The tachyzoites are the proliferating form of the organism, and, when present in tissue, they incite an acute inflammatory response and tissue necrosis They cause disease predominantly in immunocompromised hosts.258 The slowly growing bradyzoites form into cysts These cysts are present late in infection and are associated with little, if any, inflammatory response Humans can become infected with T gondii by ingesting material contaminated with infectious oocysts or tissue cysts contained in raw or undercooked meat B ❚Image 25❚ Leishmaniasis A, Cutaneous leishmaniasis showing a diffuse superficial and deep dermal lymphocytic infiltrate (H&E, ×5) B, High-power view shows the characteristic leishmanial organisms in histiocytes (H&E, ×100) Glass slides for photographing provided by Rosalie Elenitsas, MD, Dermatopathology, University of Pennsylvania Medical Center, Philadelphia Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 59 DOI: 10.1309/6BBT12WGNK77N4EH 59 59 Montone / HEAD AND NECK INFECTIONS from an intermediate host Human to human transmission can only occur from mother to fetus through a transplacental route In immunocompetent hosts, most cases are benign The most severe symptoms occur in congenital disease and in immunocompromised hosts The most common head and neck manifestation of T gondii infection is cervical lymphadenopathy.259 Histologically, lymph nodes show follicular hyperplasia associated with monocytoid B-cell hyperplasia and clusters of epithelioid histiocytes and loosely formed nonnecrotizing granulomas ❚Image 26A❚ Individual organisms are almost never found, but cysts containing organisms may rarely be observed If organisms are prominent, the patient is most likely immunosuppressed The differential diagnosis includes Hodgkin disease, mononucleosis, and Whipple disease Recent reports have shown the diagnosis of T gondii lymphadenitis using FNA biopsy of infected lymph nodes.260 Another important head and neck manifestation of T gondii is ocular involvement Ocular toxoplasmosis occurs from activation of cysts deposited in the retina and is manifested by retinal A B C D ❚Image 26❚ Toxoplasma gondii A, Cervical lymph node biopsy specimen from a patient with confirmed T gondii by serologic testing Follicular hyperplasia, monocytoid B-cell hyperplasia, and collections of epithelioid histiocytes are shown (H&E, ×25) Organisms are rarely identified in toxoplasmosis lymphadenitis, especially in immunocompetent patients B, T gondii encephalitis in an HIV+ patient There are extensive necrosis and cysts containing the organisms (H&E, ×50) C, Higher power view of T gondii encephalitis showing individual tachyzoites in the areas of necrosis (H&E, ×100) D, In situ hybridization for T gondii ribosomal RNA highlights the tachyzoites in the tissue (diaminobenzidine/hematoxylin counterstain, ×100) The presence of tachyzoites is an indication of active infection The cysts may persist for years after infection, and their presence in tissue does not confirm active disease 60 60 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE necrosis Most cases of ocular toxoplasmosis are the result of congenital infection, so clinical manifestation in adults usually represents reactivation of latent infection Reactivation of infection was recently reported following laser eye surgery.261 The diagnosis of toxoplasmosis is often made by identifying the organisms within tissue samples ❚Image 26B❚ In addition, diagnosis of disseminated disease may be made by examination of sputum.262 Tachyzoites and cysts are often seen on H&E stain ❚Image 26C❚ PAS stain can also highlight the organisms Disseminated disease has been diagnosed by examination of sputum smears The presence of rare cysts should be correlated with the clinical findings because cysts may just represent latent infection Immunohistochemical analysis, in situ hybridization, and PCR have all been used to diagnose infection ❚Image 26D❚.263-265 The prognosis is good except in immunocompromised people Mimics of Infections Myospherulosis Myospherulosis is a rare entity resulting from phagocytosis of parts of RBCs by macrophages.266 The entity is usually seen following surgeries that use petroleum-based antibiotic ointment The macrophages containing the portions of cells histologically resemble the endospores of Coccidioides or macrophages containing fungal forms such as Histoplasma or Cryptococcus The histiocytic particles are negative for silver and PAS stains Vasculitis Vasculitides in the head and neck area may histologically mimic infections.267 Necrotizing vasculitis may overlap with features of angioinvasive fungal infections Special stains highlight the organisms, and the “vasculitis” seen is secondary to the fungal infection Another vasculitis that mimics infection is Wegener granulomatosis, which is characterized by granulomatous vasculitis with zones of necrosis Infectious agents such as acid-fast organisms, some other bacterial infections, and fungal infections can manifest with similar histologic findings Elastic stains to confirm the granulomatous infiltrate centered around blood vessels should aid in the distinction between Wegener granulomatosis and infection Special stains for organisms should be performed to rule out infection when one is dealing with histologic features suspected to represent Wegener granulomatosis Although vasculitis syndromes often mimic infections, there is a strong possibility that many of these diseases result from infectious processes, particularly unknown viruses.268 From the Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia Address reprint requests to Dr Montone: Dept of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, 3400 Spruce St, Founders, Philadelphia, PA 19104 References Brook I Chronic sinusitis in children and adults: role of bacteria and antimicrobial management Curr Allergy Asthma Rep 2005;5:482-490 Biel MA, Brown CA, Levinson RM Evaluation of the microbiology of chronic maxillary sinusitis Ann Otol Rhinol Laryngol 1998;107(11 pt 1): 942-945 Brook I Microbiology and management of sinusitis J Otolaryngol 1996;25:249-256 Johnson JT, Ferguson BJ Infection/paranasal sinus Otolaryngol Head Neck Surg 1998;2:1107-1134 Wright ED, Frenkiel S Infectious adult rhinosinusitis: etiology, diagnosis and management principles J Otolaryngol 2005;34(suppl):S7-S13 Coleman H, Meer S, Altini M, et al Maxillary sinus pathology in 119 patients: a histopathologic study SADJ 2005;60:140, 142-145 Kennedy DW, Senior BA, Gannon FH, et al Histology and histomorphometry of ethmoid bone in chronic rhinosinusitis Laryngoscope 1998;108(4 pt 1):502-507 Khalid AN, Hunt JL, Perloff JR, et al The role of bone in chronic rhinosinusitis Laryngoscope 2002;112:1951-1957 Perloff JR, Gannon FH, Bolger WE, et al Bone involvement in sinusitis: an apparent pathway for the spread of disease Laryngoscope 2000;110:2095-2099 10 Ebenfelt A Bacterial location in chronic sinusitis Am J Rhinol 2005;19:458-461 11 DelGaudio JM Direct nasopharyngeal reflux of gastric acid is a contributing factor in refractory chronic rhinosinusitis Laryngoscope 2005;115:946-957 12 Perloff JR, Palmer JN Evidence of bacterial biofilms in a rabbit model of sinusitis Am J Rhinol 2005;19:1-6 13 Sanderson AR, Leid JG, Hunsaker D Bacterial biofilms on the sinus mucosa of human subjects with chronic rhinosinusitis Laryngoscope 2006;116:1121-1126 14 Tewfik TL, Al Garni M Tonsillopharyngitis: clinical highlights J Otolaryngol 2005;34(suppl 1):S45-S49 15 Dell’Aringa AR, Juares AJ, Melo C, et al Histological analysis of tonsillectomy and adenoidectomy specimens: January 2001 to May 2003 Rev Bras Otorrinolaringol (Engl Ed) 2005;71:18-22 16 Dost P Histological examination following adenoidectomy and tonsillectomy in children: surprising results are very rare HNO 2006;54:16-19 17 Cockerell CJ, Connor DH Cat scratch disease In: Connor DH, Chandler FW, Schwartz DA, et al, eds Pathology of Infectious Diseases Stamford, CT: Appleton and Lange; 1997:461-468 18 Murakami K, Tsukahara M, Tsuneoka H, et al Cat scratch disease: analysis of 130 seropositive cases J Infect Chemother 2002;8:349-352 19 Adal KA, Cockerell CJ, Petri WA Jr Cat scratch disease, bacillary angiomatosis, and other infections due to Rochalimaea N Engl J Med 1994;330:1509-1515 20 English CK, Wear DJ, Margileth AM Cat-scratch disease: isolation and culture of the bacterial agent JAMA 1988;259:1347-1352 Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 61 DOI: 10.1309/6BBT12WGNK77N4EH 61 61 Montone / HEAD AND NECK INFECTIONS 21 Min KW, Reed JA, Welch DF, et al Morphologically variable bacilli of cat scratch disease are identified by immunocytochemical labeling with antibodies to Rochalimaea henselae Am J Clin Pathol 1994;101:607-610 22 Chian CA, Arrese JE, Pierard GE Skin manifestations of Bartonella infections Int J Dermatol 2002;41:461-466 23 de Kort JG, Robben SG, Schrander JJ, et al Multifocal osteomyelitis in a child: a rare manifestation of cat scratch disease: a case report and systematic review of the literature J Pediatr Orthop B 2006;15:285-288 24 Rolain JM, Gouriet F, Enea M, et al Detection by immunofluorescence assay of Bartonella henselae in lymph nodes from patients with cat scratch disease Clin Diagn Lab Immunol 2003;10:686-691 25 Qian X, Jin L, Hayden RT, et al Diagnosis of cat scratch disease with Bartonella henselae infection in formalin-fixed paraffin-embedded tissues by two different PCR assays Diagn Mol Pathol 2005;14:146-151 26 Hansmann Y, DeMartino S, Piemont Y, et al Diagnosis of cat scratch disease with detection of Bartonella henselae by PCR: a study of patients with lymph node enlargement J Clin Microbiol 2005;43:3800-3806 27 Maruyama S, Kabeya H, Nogami S, et al Three cases of cat scratch disease diagnosed by indirect immunofluorescence antibody assay and/or polymerase chain reaction of 16S rRNA gene of Bartonella henselae J Vet Med Sci 2000;62:1321-1324 28 Cheuk W, Chan AK, Wong MC, et al Confirmation of diagnosis of cat scratch disease by immunohistochemistry Am J Surg Pathol 2006;30:274-275 29 Bonatti H, Mendez J, Guerrero I, et al Disseminated Bartonella infection following liver transplantation Transpl Int 2006;19:683-687 30 Rigopoulos D, Paparizos V, Katsambas A Cutaneous markers of HIV infection Clin Dermatol 2004;22:487-498 31 Ahsan N, Holman MJ, Riley TR, et al Peliosis hepatis due to Bartonella henselae in transplantation: a hemato-hepato-renal syndrome Transplantation 1998;65:1000-1003 32 Kreitzer T, Saoud A Bacillary angiomatosis following the use of long-term methotrexate therapy: a case report W V Med J 2006;102:317-318 33 Turgut M, Alabaz D, Karakas M, et al Bacillary angiomatosis in an immunocompetent child with a grafted traumatic wound J Dermatol 2004;31:844-847 34 LeBoit PE Bacillary angiomatosis In: Connor DH, Chandler FW, Schwartz DA, et al, eds Pathology of Infectious Diseases Stamford, CT: Appleton and Lange; 1997:407-415 35 Amsbaugh S, Huiras E, Wang NS, et al Bacillary angiomatosis associated with pseudoepitheliomatous hyperplasia Am J Dermatopathol 2006;28:32-35 36 Dauga C, Miras I, Grimont PA Identification of Bartonella henselae and B quintana 16s rDNA sequences by branch-, genus- and species-specific amplification J Med Microbiol 1996;45:192-199 37 Reed JA, Brigati DJ, Flynn SD, et al Immunocytochemical identification of Rochalimaea henselae in bacillary (epithelioid) angiomatosis, parenchymal bacillary peliosis, and persistent fever with bacteremia Am J Surg Pathol 1992;16:650-657 38 Sander A, Penno S Semiquantitative species-specific detection of Bartonella henselae and Bartonella quintana by PCR-enzyme immunoassay J Clin Microbiol 1999;37:3097-3101 39 Manolidis S, Frenkiel S, Yoskovitch A, et al Mycobacterial infections of the head and neck Otolaryngol Head Neck Surg 1993;109(3 pt 1):427-433 62 62 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH 40 Bayazit YA, Bayazit N, Namiduru M Mycobacterial cervical lymphadenitis ORL J Otorhinolaryngol Relat Spec 2004;66:275-280 41 Albright JT, Pransky SM Non-tuberculous mycobacterial infections of the head and neck Pediatr Clin North Am 2003;50:503-514 42 Aljafari AS, Khalil EA, Elsiddig KE, et al Diagnosis of tuberculous lymphadenitis by FNAC, microbiological methods and PCR: a comparative study Cytopathology 2004;15:44-48 43 Goel MM, Ranjan V, Dhole TN, et al Polymerase chain reaction vs conventional diagnosis in fine needle aspirates of tuberculous lymph nodes Acta Cytol 2001;45:333-340 44 Baek CH, Kim SI, Ko YH, et al Polymerase chain reaction detection of Mycobacterium tuberculosis from fine-needle aspirate for the diagnosis of cervical tuberculous lymphadenitis Laryngoscope 2000;110:30-34 45 Yokoyama J, Shiga K, Saijo S, et al Rapid diagnosis of cervical tuberculous lymphadenitis by application of DNA probe Otolaryngol Head Neck Surg 1999;121:501-504 46 Pahwa R, Hedau S, Jain S, et al Assessment of possible tuberculous lymphadenopathy by PCR compared to nonmolecular methods J Med Microbiol 2005;54:873-878 47 Kwon KS, Oh CK, Jang HS, et al Detection of mycobacterial DNA in cervical granulomatous lymphadenopathy from formalin-fixed, paraffin-embedded tissue by PCR J Dermatol 2000;27:355-360 48 Kraus M, Benharroch D, Kaplan D, et al Mycobacterial cervical lymphadenitis: the histological features of nontuberculous mycobacterial infection Histopathology 1999;35:534-538 49 Ramanathan VD, Jawahar MS, Paramasivan CN, et al A histological spectrum of host responses in tuberculous lymphadenitis Indian J Med Res 1999;109:212-220 50 Brady JG, Schutze GE, Seibert R, et al Detection of mycobacterial infections using the Dieterle stain Pediatr Dev Pathol 1998;1:309-313 51 Thompson LD Rhinoscleroma Ear Nose Throat J 2002;81:506 52 Schwartz DA, Geyer SJ Klebsiella and rhinoscleroma In: Connor DH, Chandler FW, Schwartz DA, et al, eds Pathology of Infectious Diseases Stamford, CT: Appleton and Lange; 1997:589-595 53 Bhargava D, Date A Palatal presentation of scleroma J Laryngol Otol 2001;115:679-680 54 Alfar-Monge JM, Fernandez-Espinosa J, Soda-Merhy A Scleroma of the lower respiratory tract: case report and review of the literature J Laryngol Otol 1994;108:161-163 55 Iyengar P, Laughlin S, Keshavjee S, et al Rhinoscleroma of the larynx [letter] Histopathology 2005;47:224-225 56 De Champs C, Vellin JF, Diancourt L, et al Laryngeal scleroma associated with Klebsiella pneumoniae subsp ozaenae J Clin Microbiol 2005;43:5811-5813 57 Verma G, Kanawaty D, Hyland R Rhinoscleroma causing upper airway obstruction Can Respir J 2005;12:43-45 58 Ammar ME, Rosen A Rhinoscleroma mimicking nasal polyposis Ann Otol Rhinol Laryngol 2001;110:290-292 59 Kasper HU, Hegenbarth V, Buhtz P Rhinoscleroma associated with Rosai-Dorfman reaction of regional lymph nodes Pathol Int 2004;54:101-104 60 Gumprecht TF, Nichols PW, Meyer PR Identification of rhinoscleroma by immunoperoxidase technique Laryngoscope 1983;93:627-629 © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE 61 Meyer PR, Shum TK, Becker TS, et al Scleroma (rhinoscleroma): a histologic immunohistochemical study with bacteriologic correlates Arch Pathol Lab Med 1983;107:377-383 62 Cockerell CJ Leprosy In: Connor DH, Chandler FW, Schwartz DA, et al, eds Pathology of Infectious Diseases Stamford, CT: Appleton and Lange; 1997:605-614 63 Jacobson RR, Krahenbuhl JL Leprosy Lancet 1999;353:655-656 64 Martins AC, Castro Jde C, Moreira JS A ten-year historic study of paranasal cavity endoscopy in patients with leprosy Rev Bras Otorhinolaryngol (Engl Ed) 2005;71:609-615 65 Ridley DS, Jopling WH Classification of leprosy according to immunity: a five-group system Int J Lepr Other Mycobact Dis 1966;34:255-273 66 Moschella SL An update on the diagnosis and treatment of leprosy J Am Acad Dermatol 2004;51:417-426 67 Katoch VM Advances in the diagnosis and treatment of leprosy Expert Rev Mol Med 2002;2002:1-14 68 Scollard DM, Gillis TP, Williams DL Polymerase chain reaction assay for the detection and identification of Mycobacterium leprae in patients in the United States Am J Clin Pathol 1998;109:642-646 69 Natrajan M, Katoch K, Katoch VM, et al In situ hybridization in the histological diagnosis of early and clinically suspect leprosy Int J Lepr Other Mycobact Dis 2004;72:296-305 70 Arnoldi J, Schluter C, Duchrow M, et al Species-specific assessment of Mycobacterium leprae in skin biopsies by in situ hybridization and polymerase chain reaction Lab Invest 1992;66:618-623 71 Balmelli C, Gunthard HF Gonococcal tonsillar infection: a case report and literature review Infection 2003;31:362-365 72 Tikjob G, Petersen CS, Ousted M, et al Localization of gonococci in the anterior oral cavity: a possible reservoir of the gonococcal infection? Ann Clin Res 1985;17:73-75 73 Koelker SE, Manz HJ, Schwartz DA Syphilis In: Connor DH, Chandler FW, Schwartz DA, et al, eds Pathology of Infectious Diseases Stamford, CT: Appleton and Lange; 1997:605-614 74 Little JW Syphilis: an update Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2005;100:3-9 75 Bruce AJ, Rogers RS III Oral manifestations of sexually transmitted diseases Clin Dermatol 2004;22:520-527 76 Zawar V, Chuh A, Gugle A Oral lesions of syphilis: an isolated, rare manifestation [serial online] Dermatol Online J 2005;11:46 Available at http://dermatology.cdib.org/113/ unknown/Zawar.html Accessed May 13, 2007 77 Barrett AW, Dorrego MV, Todgson TA, et al The histopathology of syphilis of the oral mucosa J Oral Pathol Med 2004;3:286-291 78 Lu SY, Eng HL Secondary syphilis-related oral ulcers: report of four cases Chang Gung Med J 2002;25:683-688 79 Miller M, Haddad AJ Cervicofacial actinomycosis Oral Surg Oral Med Oral Pathol Oral Radiol Endod 1998;85:496-508 80 Pulverer G, Schutt-Gerowitt H, Schaal KP Human cervicofacial actinomycoses: microbiological data for 1997 cases Clin Infect Dis 2003;37:490-497 81 Smego RA Jr, Foglia G Actinomycosis Clin Infect Dis 1998;26:1255-1261 82 Volante M, Contucci AM, Fantoni M, et al Cervicofacial actinomycosis: still a difficult differential diagnosis Acta Otorhinolaryngol Ital 2005;25:116-119 83 Stewart AE, Palma JR, Amsberry JK Cervicofacial actinomycosis Otolaryngol Head Neck Surg 2005;132:957-959 84 Oostman O, Smego RA Cervicofacial actinomycosis: diagnosis and management Curr Infect Dis Rep 2005;7:170-174 85 Belmont MJ, Behar PM, Wax MK Atypical presentations of actinomycosis Head Neck 1999;21:264-268 86 Park YH, Baik JH, Yoo J Acute thyroiditis of actinomycosis Thyroid 2005;15:1395-1396 87 Subha ST, Raman R, Cheah PL, et al Actinomyces infection of the mastoid: a rare entity Med J Malaysia 2004;59:680-681 88 Ulvi BM Actinomycosis of the parotid gland Plast Reconstr Surg 2005;115:659-661 89 Ozcan C, Talas D, Gorur K, et al Actinomycosis of the middle turbinate: an unusual cause of nasal obstruction Eur Arch Otorhinolaryngol 2005;262:412-415 90 Bubbico L, Caratozzolo M, Nardi F, et al Actinomycosis of submandibular gland: an unusual presentation Acta Otorhinolaryngol Ital 2004;24:37-39 91 Cevera JJ, Butehorn HF III, Shapiro J, et al Actinomycosis abscess of the thyroid gland Laryngoscope 2003;113:2108-2111 92 Roth M, Montone KT Actinomycosis of the paranasal sinuses: a case report and review Otolaryngol Head Neck Surg 1996;114:818-821 93 Jaax NK, Fritz DL Anthrax In: Connor DH, Chandler FW, Schwartz DA, et al, eds Pathology of Infectious Diseases, Stamford, CT: Appleton and Lange; 1997:397-406 94 Ezzell JW, Abshire TG, Little SF, et al Identification of Bacillus anthracis by using monoclonal antibody to cell wall galactose-N-acetylglucosamine polysaccharide J Clin Microbiol 1990;28:223-231 95 Brandwein M Histopathology of sinonasal fungal disease Otolaryngol Clin North Am 1993;26:949-978 96 Corey JP, Romberger CF, Shaw GY Fungal disease of the sinuses Otolaryngol Head Neck Surg 1990;103:1012-1015 97 Adelson RT, Marple BF Fungal rhinosinusitis: state of the art diagnosis and treatment J Otolaryngol 2005;34(suppl 1):S18-S23 98 Granville L, Chirala M, Cernoch P, et al Fungal sinusitis: histologic spectrum and correlation with culture Hum Pathol 2004;35:474-481 99 Taxy JB Paranasal fungal sinusitis: contributions of histopathology to diagnosis: a report of 60 cases and literature review Am J Surg Pathol 2006;30:713-720 100 Ferreiro JA, Carlson BA, Cody DT III Paranasal sinus fungus balls Head Neck 1997;19:481-486 101 Dufour X, Kauffmann-Lacroix C, Ferrie JC, et al Paranasal sinus fungus ball: epidemiology, clinical features and diagnosis: a retrospective analysis of 173 cases from a single medical center in France, 1989-2002 Med Mycol 2006;44:61-67 102 Dufour X, Kauffmann-Lacroix C, Ferrie JC, et al Paranasal sinus fungus ball and surgery: a review of 175 cases Rhinology 2005;43:34-39 103 Gungor A, Adusumilli V, Corey JP Fungal sinusitis: progression of disease in immunosuppression: a case report Ear Nose Throat J 1998;77:207-210 104 Katzenstein AL, Sale SR, Greenberger PA Pathologic findings in allergic Aspergillus sinusitis: a newly recognized form of sinusitis Am J Surg Pathol 1983;7:439-443 105 Allphin AL, Strauss M, Abdul-Karim FW Allergic fungal sinusitis: problems in diagnosis and treatment Laryngoscope 1991;101:815-820 106 Bent JP, Kuhn FA Diagnosis of allergic fungal sinusitis Otolaryngol Head Neck Surg 1994;111:580-587 107 Cody DT, Neel HB, Ferreiro JA, et al Allergic fungal sinusitis: the Mayo Clinic experience Laryngoscope 1994;104:1074-1079 Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 63 DOI: 10.1309/6BBT12WGNK77N4EH 63 63 Montone / HEAD AND NECK INFECTIONS 108 Corey JP Allergic fungal sinusitis Otolaryngol Clin North Am 1992;25:225-229 109 Ence BK, Gourley DS, Jorgensen NL, et al Allergic fungal sinusitis Am J Rhinol 1990;4:169-178 110 Goldstein MF Allergic fungal sinusitis: an under diagnosed problem Hosp Pract (Off Ed) 1992:73-74, 79-84, 87-88, passim 111 Schubert MS Allergic fungal sinusitis Clin Rev Allergy Immunol 2006;30:205-216 112 Bartynski JM, McCaffrey TV, Frigas E Allergic fungal sinusitis secondary to dematiaceous fungi: Curvularia lunata and Alternaria Otolaryngol Head Neck Surg 1990;103:32-39 113 Friedman GC, Hartwick RW, Ro JY, et al Allergic fungal sinusitis report of three cases associated with dematiaceous fungi Am J Clin Pathol 1991;96:368-372 114 Manning SC, Schaefer SD, Close LG, et al Culture-positive allergic fungal sinusitis Arch Otolaryngol Head Neck Surg 1991;117:174-178 115 Ponikau JU, Sherris DA, Kern EB, et al The diagnosis and incidence of allergic fungal sinusitis Mayo Clin Proc 1999;74:877-884 116 Heffner DK Allergic fungal sinusitis is a histopathologic diagnosis; paranasal mucocele is not Ann Diagn Pathol 2004;8:316-323 117 Campbell JM, Graham M, Gray HC, et al Allergic fungal sinusitis in children Ann Allergy Asthma Immunol 2006;96:286-290 118 Singh N, Bhalodiya NH Allergic fungal sinusitis (AFS): earlier diagnosis and management J Laryngol Otol 2005;119:875-881 119 Manning SC, Mabry RL, Schaefer SD, et al Evidence of IgEmediated hypersensitivity in allergic fungal sinusitis Laryngoscope 1993;103:717-721 120 Ramadan HH, Quraishi HA Allergic mucin without fungus Am J Rhinol 1997;11:145-147 121 Ferguson BJ Eosinophilic mucin rhinosinusitis: a distinct clinicopathological entity Laryngoscope 2000;110(5 pt 1):799-813 122 Shatkin JS, Delsupehe KG, Thisted RA, et al Mucosal allergy in the absence of systemic allergy in nasal polyposis and rhinitis: a meta-analysis Otolaryngol Head Neck Surg 1994;111:553-556 123 Perez LA, Lanza DC, Loevner LA, et al In situ hybridization for Aspergillus and Penicillium in allergic fungal sinusitis: a rapid means of speciating fungal pathogens in tissues Laryngoscope 1997;107:233-240 124 Smith KJ, Yeager J, Skelton H Molluscum contagiosum: its clinical, histopathologic, and immunohistochemical spectrum Int J Dermatol 1999;38:664-672 125 Dohil MA, Lin P, Lee J, et al The epidemiology of molluscum contagiosum in children J Am Acad Dermatol 2006;54:47-54 126 Redmond RM Molluscum contagiosum is not always benign [letter] BMJ 2004;329:403 127 Pitini V, Arrigo C, Barresi G Disseminated molluscum contagiosum in a patient with chronic lymphocytic leukaemia after alemtuzumab Br J Haematol 2003;123:565 128 Dave S, Thappa DM, Karthikeyan K Disseminated and disfiguring molluscum contagiosum in a child Pediatr Dermatol 2003;20:436-439 129 Forghani B, Oshiro LS, Chan CS, et al Direct detection of molluscum contagiosum virus in clinical specimens by in situ hybridization using biotinylated probe Mol Cell Probes 1992;6:67-77 64 64 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH 130 Chidzonga MM HIV/AIDS orofacial lesions in 156 Zimbabwean patients at referral oral and maxillofacial clinics Oral Dis 2003;9:317-322 131 Campanini A, Marani M, Mastroianni A, et al Human immunodeficiency virus infection: personal experience in changes in head and neck manifestations due to recent antiretroviral therapies Acta Otorhinolaryngol Ital 2005;25:30-35 132 Hillel A, O’Mara W, Nemechek A, et al Head and neck manifestations in HIV infection J La State Med Soc 2004;156:245-253 133 Ndjolo A, Njock R, Ngowe NM, et al Early ENT manifestations of HIV infection/AIDS: an analysis of 76 cases observed in Africa [in French] Rev Laryngol Otol Rhinol (Bord) 2004;125:39-43 134 Marsot-Dupuch K, Quillard J, Meyohas MC Head and neck lesions in the immunocompromised host Eur Radiol 2004;14(suppl 3):E155-E167 135 Gurney TA, Murr AH Otolaryngologic manifestations of human immunodeficiency virus infection Otolaryngol Clin North Am 2003;36:607-624 136 Reznik DA Oral manifestations of HIV disease Top HIV Med 2005-2006;13:143-148 137 Baroni CD, Uccini S The lymphadenopathy of HIV infection Am J Clin Pathol 1993;99:397-401 138 Burke AP, Anderson D, Mannan P, et al Systemic lymphadenopathic histology in human immunodeficiency virus-1–seropositive drug addicts without apparent acquired immunodeficiency syndrome Hum Pathol 1994;25:248-256 139 Ost A, Baroni CD, Biberfeld P, et al Lymphadenopathy in HIV infection: histological classification and staging Acta Pathol Microbiol Immunol Scand Suppl 1989;8:7-15 140 Favia G, Capodiferro S, Scivetti M, et al Multiple parotid lymphoepithelial cysts in patients with HIV-infection: report of two cases Oral Dis 2004;10:151-154 141 Ihrler S, Zietz C, Riederer A, et al HIV-related parotid lymphoepithelial cysts: immunohistochemistry and 3-D reconstruction of surgical and autopsy material with special reference to formal pathogenesis Virchows Arch 1996;429:139-147 142 Mandel L, Reich R HIV parotid gland lymphoepithelial cysts: review and case reports Oral Surg Oral Med Oral Pathol 1992;74:273-278 143 Wilkins K, Turner R, Doley JC, et al Cutaneous malignancy and human immunodeficiency virus disease J Am Acad Dermatol 2006;54:189-206 144 Raju PV, Rao GR, Ramani TV, et al Skin disease: clinical indicator of immune status in human immunodeficiency virus (HIV) infection Int J Dermatol 2005;44:646-649 145 Arduino PG, Porter SR Oral and perioral herpes simplex virus type (HSV-1) infection: review of its management Oral Dis 2006;12:254-270 146 Kolokotronis A, Doumas S Herpes simplex virus infection, with particular reference to the progression and complications of primary herpetic gingivostomatitis Clin Microbiol Infect 2006;12:202-211 147 Stoopler ET Oral herpetic infections (HSV 1-8) Dent Clin North Am 2005;49:15-29 148 Huber MA Herpes simplex type-1 virus infection Quintessence Int 2003;34:453-467 149 Simmons A Clinical manifestations and treatment considerations of herpes simplex virus infection J Infect Dis 2002;186(suppl 1):S71-S77 © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE 150 Pepose JS, Keadle TL, Morrison LA Ocular herpes simplex: changing epidemiology, emerging disease patterns, and the potential of vaccine prevention and therapy Am J Ophthalmol 2006;141:547-557 151 Bonfioli AA, Eller AW Acute retinal necrosis Semin Ophthalmol 2005;20:155-160 152 Syrjänen S, Leimola-Virtanen R, Schmidt-Westhausen A, et al Oral ulcers in AIDS patients frequently associated with cytomegalovirus (CMV) and Epstein-Barr virus (EBV) infections J Oral Pathol Med 1999;28:204-209 153 Leimola-Virtanen R, Happonen RP, Syrjänen S Cytomegalovirus (CMV) and Helicobacter pylori (HP) found in oral mucosal ulcers J Oral Pathol Med 1995;24:14-17 154 Chan BW, Woo JK, Liew CT Cytomegalovirus infection of the nasopharynx J Clin Pathol 2002;55:970-972 155 Wiegand TW, Young LH Cytomegalovirus retinitis Int Ophthalmol Clin 2006;46:91-110 156 Scholz M, Doerr HW, Cinatl J Human cytomegalovirus retinitis: pathogenicity, immune evasion and persistence Trends Microbiol 2003;11:171-178 157 See RF, Rao NA Cytomegalovirus retinitis in the era of combined highly active antiretroviral therapy Ophthalmol Clin North Am 2002;15:529-536 158 Lopez-Amado M, Yebra-Pimentel MT, Garcia-Sarandeses A Cytomegalovirus causing necrotizing laryngitis in a renal and cardiac transplant recipient Head Neck 1996;18:455-457 159 Wax TD, Layfield LJ, Zaleski S, et al Cytomegalovirus sialadenitis in patients with the acquired immunodeficiency syndrome: a potential diagnostic pitfall with fine-needle aspiration cytology Diagn Cytopathol 1994;10:169-172 160 Frank TS, LiVolsi VA, Connor AM Cytomegalovirus infection of the thyroid in immunocompromised adults Yale J Biol Med 1987;60:1-8 161 Liesegang TJ Herpes zoster virus infection Curr Opin Ophthalmol 2004;15:531-536 162 Gurwood AS, Savochka J, Sirgany BJ Herpes zoster ophthalmicus Optometry 2002;73:295-302 163 Ebell MH Epstein-Barr virus infectious mononucleosis Am Fam Physician 2004;70:1279-1287 164 Cohen JI Benign and malignant Epstein-Barr virus–associated B-cell lymphoproliferative diseases Semin Hematol 2003;40:116-123 165 Godshall SE, Kirchner JT Infectious mononucleosis: complexities of a common syndrome Postgrad Med 2000;107:175-179, 183-184 166 Gowing NF Infectious mononucleosis: histopathologic aspects Pathol Annu 1975;10:1-20 167 Sengor A, Willke A, Aydin O, et al Isolated necrotizing epiglottitis: report of a case in a neutropenic patient and review of the literature Ann Otol Rhinol Laryngol 2004;113(3 pt 1):225-228 168 Iijima T, Sumazaki R, Mori N, et al A pathological and immunohistological case report of fatal infectious mononucleosis, Epstein-Barr virus infection, demonstrated by in situ and Southern blot hybridization Virchows Arch A Pathol Anat Histopathol 1992;421:73-78 169 Moura MD, Grossmann Sde M, Fonseca LM, et al Risk factors for oral hairy leukoplakia in HIV-infected adults of Brazil J Oral Pathol Med 2006;35:321-326 170 Syrjänen S, Laine P, Happonen R-P, et al Oral hairy leukoplakia is not a specific sign of HIV-infection but related to immunosuppression in general J Oral Pathol Med 1989;18:28-31 171 Eisenberg E, Krutchkoff D, Yamase H Incidental oral hairy leukoplakia in immunocompetent persons: a report of two cases Oral Surg Oral Med Oral Pathol 1992;74:332-333 172 Felix DH, Watret K, Wray D, et al Hairy leukoplakia in anHIV-negative, nonimmunosuppressed patient Oral Surg Oral Med Oral Pathol 1992;74:563-566 173 Triantos D, Porter SR, Scully C, et al Oral hairy leukoplakia: clinicopathologic features, pathogenesis, diagnosis, and clinical significance Clin Infect Dis 1997;25:1392-1396 174 Greenspan JS, Greenspan D, Lennette ET, et al Replication of Epstein-Barr virus within the epithelial cells of oral “hairy” leukoplakia: an AIDS-associated lesion New Engl J Med 1985;313:1564-1571 175 Dias EP, Rocha ML, Silva A Jr, et al Oral hairy leukoplakia: histopathologic and cytopathologic features of a subclinical phase Am J Clin Pathol 2000;114:395-401 176 Frizzera G, Hanto DW, Gajl-Peczalska K, et al Polymorphic diffuse B cell hyperplasias and lymphomas in renal transplant recipients Cancer Res 1981;41:4262-4279 177 Hanto DW, Frizzera G, Purtilo DT, et al Clinical spectrum of lymphoproliferative disorders in renal transplant recipients and evidence for the role of Epstein-Barr virus Cancer Res 1981;41:4253-4261 178 Hanto DW, Gajl-Peczalska K, Frizzera G, et al Epstein-Barr virus (EBV) induced polyclonal and monoclonal B-cell lymphoproliferative disease occurring after renal transplantation: clinical, pathologic, and virologic findings and implications for therapy Ann Surg 1983;198:356-369 179 Nalesnik M, Jaffe R, Starzl TE, et al The pathology of posttransplant lymphoproliferative disorders occurring in the setting of cyclosporine A–prednisone immunosuppression Am J Pathol 1988;133:173-193 180 Purtilo DT, Strobach RS, Okanao M, et al Epstein-Barr virus–associated lymphoproliferative disorders Lab Invest 1992;67:5-23 181 Purtilo DT Opportunistic cancers in patients with immunodeficiency syndromes Arch Pathol Lab Med 1987;111:1123-1129 182 Cohen JI Epstein-Barr virus lymphoproliferative disease associated with acquired immune deficiency Medicine 1991;70:137-160 183 Starzl TE, Porter KA, Iwatsuki S, et al Reversibility of lymphomas and lymphoproliferative lesions developing under cyclosporin-steroid therapy Lancet 1988;1:583-587 184 Hague K, Catalano P, Rothschild M, et al Posttransplant lymphoproliferative disease presenting as sudden respiratory arrest in a three-year-old child Ann Otol Rhinol Laryngol 1997;106:244-247 185 Lones MA, Mishalani S, Shintaku IP, et al Changes in tonsils and adenoids in children with posttransplant lymphoproliferative disorder: report of three cases with early involvement of Waldeyer’s ring Hum Pathol 1995;26:525-530 186 Harris NL, Swerdlow SH, Frizerra G, et al Post-transplant lymphoproliferative disorders In: Jaffe ES, Harris NL, Stein H, et al, eds Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues Lyon, France: IARC Press; 2001:264269 World Health Organization Classification of Tumours 187 Ho M, Miller G, Atchison RW, et al Epstein-Barr virus infections and DNA hybridization studies in posttransplantation lymphoma and lymphoproliferative lesions: the role of primary infection J Infect Dis 1985;152:876-886 188 Randhawa PS, Jaffe R, Demetris AJ, et al Expression of Epstein-Barr virus small RNA (by EBER-1 gene) in liver specimens from transplant recipients with post-transplantation disease N Engl J Med 1992;137:1710-1714 Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 65 DOI: 10.1309/6BBT12WGNK77N4EH 65 65 Montone / HEAD AND NECK INFECTIONS 189 Montone KT, Hodinka R, Salhany KE, et al Identification of lytic Epstein-Barr virus activity in posttransplant lymphoproliferative disease Mod Pathol 1996;9:621-630 190 Fischer A, Blanche S, Le Bidois J, et al Anti–B cell monoclonal antibodies in the treatment of severe B-cell lymphoproliferative syndrome following bone marrow and organ transplantation N Engl J Med 1991;324:1451-1456 191 Porter DL, Orloff GJ, Antin JH Donor mononuclear cell infusions as therapy for B-cell lymphoproliferative disorder following allogeneic bone marrow transplant Transplant Sci 1994;4:12-16 192 Haque T, Amlot PL, Helling N, et al Reconstitution of EBVspecific T-cell immunity in solid organ transplant recipients J Immunol 1998;160:6204-6209 193 Gilmour KC, Gaspar HB Pathogenesis and diagnosis of Xlinked lymphoproliferative disease Expert Rev Mol Diagn 2003;3:549-561 194 Gaspar HB, Sharifi R, Gilmour KC, et al X-linked lymphoproliferative disease: clinical, diagnostic and molecular perspective Br J Haematol 2002;119:585-595 195 Morra M, Howie D, Grande MS, et al X-linked lymphoproliferative disease: a progressive immunodeficiency Annu Rev Immunol 2001;19:657-682 196 Nelson DL, Terhorst C X-linked lymphoproliferative syndrome Clin Exp Immunol 2000;122:291-295 197 Maia DM, Garwacki CP X-linked lymphoproliferative disease: pathology and diagnosis Pediatr Dev Pathol 1999;2:72-77 198 Thompson MP, Kurzrock R Epstein-Barr virus and cancer Clin Cancer Res 2004;10:803-821 199 van den Bosch CA Is endemic Burkitt’s lymphoma an alliance between three infections and a tumour promoter? Lancet Oncol 2004;5:738-746 200 Fujita F, Buziba N, Kumatori A, et al Early stage of EpsteinBarr virus lytic infection leading to the “starry sky” pattern formation in endemic Burkitt lymphoma Arch Pathol Lab Med 2004;128:549-552 201 Labrecque LG, Xue SA, Kazembe P, et al Expression of Epstein-Barr virus lytically related genes in African Burkitt’s lymphoma: correlation with patient response to therapy Int J Cancer 1999;81:6-11 202 Hummel M, Bentink S, Berger H, et al, for the Molecular Mechanisms in Malignant Lymphomas Network Project of the Deutsche Krebshilfe A biologic definition of Burkitt’s lymphoma from transcriptional and genomic profiling N Engl J Med 2006;354:2419-2430 203 Dave SS, Fu K, Wright GW, et al, for the Lymphoma/Leukemia Molecular Profiling Project Molecular diagnosis of Burkitt’s lymphoma N Engl J Med 2006;354:2431-2442 204 Vega F, Lin P, Medeiros LJ Extranodal lymphomas of the head and neck Ann Diagn Pathol 2005;9:340-350 205 Sheahan P, Donnelly M, O’Reilly S, et al T/NK cell non–Hodgkin’s lymphoma of the sinonasal tract J Laryngol Otol 2001;115:1032-1035 206 Ohshima K, Suzumiya J, Shimazaki K, et al Nasal T/NK cell lymphomas commonly express perforin and Fas ligand: important mediators of tissue damage Histopathology 1997;31:444-450 207 Medeiros LJ, Jaffe ES, Chen YY, et al Localization of EpsteinBarr viral genomes in angiocentric immunoproliferative lesions Am J Surg Pathol 1992;16:439-447 208 Hongyo T, Li T, Syaifudin M Specific c-kit mutations in sinonasal natural killer/T-cell lymphoma in China and Japan Cancer Res 2000;60:2345-2347 66 66 Am J Clin Pathol 2007;128:35-67 DOI: 10.1309/6BBT12WGNK77N4EH 209 Ko YH, Choi KE, Han JH, et al Comparative genomic hybridization study of nasal-type NK/T-cell lymphoma Cytometry 2001;46:85-91 210 Cho HI, Hong YS, Lee MA, et al Adoptive transfer of Epstein-Barr virus–specific cytotoxic T-lymphocytes for the treatment of angiocentric lymphomas Int J Hematol 2006;83:66-73 211 Chan AT, Teo PM, Huang DP Pathogenesis and treatment of nasopharyngeal carcinoma Semin Oncol 2004;6:794-801 212 Dolcetti R, Menezes J Epstein-Barr virus and undifferentiated nasopharyngeal carcinoma: new immunobiological and molecular insights on a long-standing etiopathogenic association Adv Cancer Res 2003;87:127-157 213 Fan SQ, Ma J, Zhou J, et al Differential expression of Epstein-Barr virus–encoded RNA and several tumor-related genes in various types of nasopharyngeal epithelial lesions and nasopharyngeal carcinoma using tissue microarray analysis Hum Pathol 2006;37:593-605 214 Biswal BM, Kareem A, Ahmed NM Hypertrophic osteoarthropathy: an unusual manifestation in nasopharyngeal cancer Australas Radiol 2001;45:71-73 215 Nakao K, Yuge T, Mochiki M, et al Detection of Epstein-Barr virus in metastatic lymph nodes of patients with nasopharyngeal carcinoma and a primary unknown carcinoma Arch Otolaryngol Head Neck Surg 2003;129:338340 216 Nakao K, Mochiki M, Nibu K, et al Analysis of prognostic factors of nasopharyngeal carcinoma: impact of in situ hybridization for Epstein-Barr virus encoded small RNA Otolaryngol Head Neck Surg 2006;134:639-645 217 Zhong BL, Zong YS, Lin SX, et al Epstein-Barr virus infection in precursor lesions of nasopharyngeal carcinoma Ai Zheng 2006;25:136-142 218 Pak MW, To KF, Lo YM, et al Nasopharyngeal carcinoma in situ (NPCIS): pathologic and clinical perspectives Head Neck 2002;24:989-995 219 Wong MM, Lye MS, Cheng HM, et al Epstein-Barr virus serology in the diagnosis of nasopharyngeal carcinoma Asian Pac J Allergy Immunol 2005;23:65-67 220 Zerr DM Human herpesvirus 6: a clinical update Herpes 2006;13:20-24 221 Ward KN Human herpesviruses-6 and -7 infections Curr Opin Infect Dis 2005;18:247-252 222 Clark DA Human herpesvirus and human herpesvirus 7: emerging pathogens in transplant patients Int J Hematol 2002;76(suppl 2):246-252 223 Jackson MA, Sommerauer JF Human herpesviruses and Pediatr Infect Dis J 2002;21:565-566 224 Chen T, Hudnall SD Anatomical mapping of human herpesvirus reservoirs of infection Mod Pathol 2006;19:726737 225 Ablashi DV, Chatlynne LG, Whitman JE Jr, et al Spectrum of Kaposi’s sarcoma–associated herpesvirus, or human herpesvirus 8, diseases Clin Microbiol Rev 2002;15:439-464 226 Malnati MS, Dagna L, Ponzoni M, et al Human herpesvirus (HHV-8/KSHV) and hematologic malignancies Rev Clin Exp Hematol 2003;7:375-405 227 Aoki Y, Tosato G Pathogenesis and manifestations of human herpesvirus-8–associated disorders Semin Hematol 2003;40:143-153 228 Schwartz RA Kaposi’s sarcoma: an update J Surg Oncol 2004;87:146-151 © American Society for Clinical Pathology Microbiology and Infectious Disease / REVIEW ARTICLE 229 Vanni T, Sprinz E, Machado MW, et al Systemic treatment of AIDS-related Kaposi sarcoma: current status and perspectives Cancer Treat Rev 2006;32:445-455 230 Almadori G, Galli J, Cadoni G, et al Human papillomavirus infection and cyclin D1 gene amplification in laryngeal squamous cell carcinoma: biologic function and clinical significance Head Neck 2002;24:597-604 231 Sisk EA, Soltys SG, Zhu S, et al Human papillomavirus and p53 mutational status as prognostic factors in head and neck carcinoma Head Neck 2002;24:841-849 232 Baez A, Almodovar JI, Cantor A, et al High frequency of HPV16-associated head and neck squamous cell carcinoma in the Puerto Rican population Head Neck 2004;26:778-784 233 Weinberger PM, Yu Z, Haffty BG, et al Molecular classification identifies a subset of human papillomavirus– associated oropharyngeal cancers with favorable prognosis J Clin Oncol 2006;24:736-747 234 Torrente MC, Ampuero S, Abud M, et al Molecular detection and typing of human papillomavirus in laryngeal carcinoma specimens Acta Otolaryngol 2005;125:888-893 235 Makannavar JH, Chavan SS Rhinosporidiosis: a clinicopathological study of 34 cases Indian J Pathol Microbiol 2001;44:17-21 236 Parthasarathi R, Sen S, Khan M Conjunctival rhinosporidiosis: J Indian Med Assoc 2003;101:667-670 237 Khoo JJ, Kumar KS Rhinosporidiosis presenting as recurrent nasal polyps Med J Malaysia 2003;58:282-285 238 Fredricks DN, Jolley JA, Lepp PW, et al Rhinosporidium seeberi: a human pathogen from a novel group of aquatic protistan parasites Emerg Infect Dis 2000;6:273-282 239 Herr RA, Ajello L, Taylor JW, et al Phylogenetic analysis of Rhinosporidium seeberi’s 18S small-subunit ribosomal DNA groups this pathogen among members of the protoctistan Mesomycetozoa clade J Clin Microbiol 1999;37:2750-2754 240 Mohanty RC, Khalkho J, Sahu A Diagnosis of disseminated rhinosporidiosis by fine needle aspiration cytology [letter] Acta Cytol 2006;50:234 241 Angunawela P, De Tissera A, Dissanaike AS Rhinosporidiosis presenting with two soft tissue tumors followed by dissemination Pathology 1999;31:57-58 242 Thappa DM, Venkatesan S, Sirka CS, et al Disseminated cutaneous rhinosporidiosis J Dermatol 1998;25:527-532 243 Kafetzis DA An overview of paediatric leishmaniasis J Postgrad Med 2003;49:31-38 244 Berman JD Human leishmaniasis: clinical, diagnostic, and chemotherapeutic developments in the last 10 years Clin Infect Dis 1997;24:684-703 245 Murray HW, Berman JD, Davies CR, et al Advances in leishmaniasis Lancet 2005;366:1561-1577 246 Markle WH, Makhoul K Cutaneous leishmaniasis: recognition and treatment Am Fam Physician 2004;69:1455-1460 247 Willard RJ, Jeffcoat AM, Benson PM, et al Cutaneous leishmaniasis in soldiers from Fort Campbell, Kentucky returning from Operation Iraqi Freedom highlights diagnostic and therapeutic options J Am Acad Dermatol 2005;52:977-987 248 Weina PJ, Neafie RC, Wortmann G, et al Old world leishmaniasis: an emerging infection among deployed US military and civilian workers Clin Infect Dis 2004;39:16741680 249 Piscopo TV, Mallia AC Leishmaniasis Postgrad Med J 2006;82:649-657 250 Guerin PJ, Olliaro P, Sundar S, et al Visceral leishmaniasis: current status of control, diagnosis, and treatment, and a proposed research and development agenda Lancet Infect Dis 2002;2:494-501 251 Di Lella F, Vincenti V, Zennaro D, et al Mucocutaneous leishmaniasis: report of a case with massive involvement of nasal, pharyngeal and laryngeal mucosa Int J Oral Maxillofac Surg 2006;35:870-872 252 Aliaga L, Cobo F, Mediavilla JD, et al Localized mucosal leishmaniasis due to Leishmania (Leishmania) infantum: clinical and microbiologic findings in 31 patients Medicine (Baltimore) 2003;82:147-158 253 Delbrouck C, Choufani G, Nagy N, et al Systemic leishmaniasis involving the nose Acta Otorhinolaryngol Belg 2003;57:83-85 254 Shareef MM, Trotter MI, Cullen RJ Leishmaniasis of the nasal cavity: a case report J Laryngol Otol 2005;119:10151017 255 Lambertucci JR, Coulaud R, Silva LC Mucosal leishmaniasis Rev Soc Bras Med Trop 2003;36:307-308 256 Guerreiro JB, Cruz AA, Barral A, et al Mucosal leishmaniasis: quantitative nasal cytology as a marker of disease activity and indicator of healing Ann Otol Rhinol Laryngol 2000;109:8994 257 van Eys GJ, Schoone GJ, Ligthart GS, et al Detection of Leishmania parasites by DNA in situ hybridization with nonradioactive probes Parasitol Res 1987;73:199-202 258 Frenkel JK Toxoplasmosis In: Connor DH, Chandler FW, Schwartz DA, et al, ed Pathology of Infectious Diseases Stamford, CT: Appleton and Lange; 1997:1261-1278 259 McCabe RE, Brooks RG, Dorfman RF, et al Clinical spectrum in 107 cases of toxoplasmic lymphadenopathy Rev Infect Dis 1987;9:754-774 260 Viguer JM, Jimenez-Heffernan JA, Lopez-Ferrer P, et al Fine needle aspiration of toxoplasmic (Piringer-Kuchinka) lymphadenitis: a cytohistologic correlation study Acta Cytol 2005;49:139-143 261 Barbara A, Shehadeh-Masha’our R, Sartani G, et al Reactivation of ocular toxoplasmosis after LASIK J Refract Surg 2005;21:759-761 262 Laibe S, Ranque S, Curtillet C, et al Timely diagnosis of disseminated toxoplasmosis by sputum examination J Clin Microbiol 2006;44:646-648 263 Cermakova Z, Ryskova O, Pliskova L Polymerase chain reaction for detection of Toxoplasma gondii in human biological samples Folia Microbiol (Praha) 2005;50:341-344 264 Switaj K, Master A, Skrzypczak M, et al Recent trends in molecular diagnostics for Toxoplasma gondii infections Clin Microbiol Infect 2005;11:170-176 265 Montone KT In situ hybridization for ribosomal RNA sequences: a rapid, sensitive method for diagnosis of infectious pathogens in anatomic pathology substrates Acta Histochem Cytochem 1994;27:601-606 266 Rosai J The nature of myospherulosis of the upper respiratory tract Am J Clin Pathol 1978;69:475-481 267 Suresh E Diagnostic approach to patients with suspected vasculitis Postgrad Med J 2006;82:483-488 268 Mandell BF, Calabrese LH Infections and systemic vasculitis Curr Opin Rheumatol 1998;10:51-57 Am J Clin Pathol 2007;128:35-67 © American Society for Clinical Pathology 67 DOI: 10.1309/6BBT12WGNK77N4EH 67 67 ... a nodular mass at the angle of the jaw The masses gradually increase in size and number and form sinuses that open onto the face and neck Although infection is often seen in the jaw area, Actinomyces... several ways in the head and neck area Many of these lesions have been (or will be) discussed separately Almost 70% of HIV-infected patients will have lesions involving the head and neck. 130-136... HHV-7, and HHV-8 can have manifestations in the head and neck HSV-1 and HSV-2 Infection with HSVs may result in significant head and neck manifestations The severity seems to depend on the immune

Ngày đăng: 19/02/2014, 08:20

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan