iaet08i1p49

18
7/21/2019 iaet08i1p49 http://slidepdf.com/reader/full/iaet08i1p49 1/18 Review Article Correspondence and reprint requests:  Dr V.K. Vijayan, Director, V.P. Chest Institute, University of Delhi, Delhi-110 007, India; Phone: (Off.) 91-11-27666180, (Resi.) 91-11-27667027; Fax: 91-11-27667420; E-mail: [email protected]. Tropical Parasitic Lung Diseases V.K. Vijayan Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India ABSTRACT Though parasitic lung diseases are frequently seen in tropical countries, these are being increasingly reported from many parts of the world due to globalisation and travel across the continents. In addition, the emergence of human immunodeficiency virus (HIV) infection/acquired immunodeficiency syndrome (AIDS), the frequent use of immunosuppressive drugs in many diseases and the increasing numbers of organ transplantations have resulted in a renewed interest in many tropical parasitic lung diseases. This review outlines the recent developments in the pathogenesis, diagnosis and management of common and rare parasitic lung diseases. [Indian J Chest Dis Allied Sci 2008; 50: 49-66] Key words: Parasitic pneumonias, Pulmonary amoebiasis, Pulmonary malaria, Pulmonary paragonimiasis, Pulmonary ascariasis, Pulmonary strongyloidiasis, Tropical pulmonary eosinophilia. INTRODUCTION Protozoal and helminthic parasitic diseases are common in tropical regions of the world. Helminthic parasites induce blood and tissue eosinophilia. However, protozoal infestations usually do not cause eosinophilia. Pulmonary disease affecting the major airways and/or parenchyma associated with either blood and/or tissue eosinophilia is classified as eosinophilic lung disease. The first four cases with minimal respiratory symptoms, fleeting pulmonary infiltrates in the chest radiographs and peripheral blood eosinophilia were described by Loffler in 1932. 1  Although all types of eosinophilic lung diseases are encountered in the tropics as in other parts of the world, the diseases that are particularly prevalent in the tropics are due to parasitic infestations. 2  The important diseases that can cause tropical parasitic lung diseases are listed in the table. PROTOZOAL PARASITES Protozoal parasites that cause pulmonary diseases are Entamoeba histolytica  , Leishmania donovani  , malarial parasites (Plasmodium vivax, P. falciparum, P. malariae and, P. ovale), Toxoplasma gondii  , Babesia microti and Babesia divergens. Pulmonary Amoebiasis The causative agent of amoebiasis is the protozoan parasite, Entamoeba, histolytica  and pulmonary amoebiasis occurs mainly by the extension from the amoebic liver abscess. 3,4  Fever, right upper quadrant Table. Tropical parasitic lung diseases Diseases Parasites Protozoa Pulmonary amoebiasis Entamoeba histolytica Pulmonary leishmaniasis Leishmania donovani Pulmonary malaria Plasmodium vivax, Plasmodium falciparum Plasmodium malariae Plasmodium ovale Pulmonary toxoplasmosis Toxoplasma gondii Pulmonary babesiosis Babesia microti, Babesia divergens Helminths Cestodes Pulmonary hydatid cyst Echinococcus granulosus Echinococcus multilocularis Trematodes Pulmonary schistosomiasis Schistosoma haematobium Schistosoma mansoni Schistosoma japonicum Pulmonary paragonimiasis Paragonimus westermani  Nematodes Pulmonary ascariasis Ascaris lumbricoides Pulmonary ancylostomiasis Ancylostoma duodenale Necator americanus Pulmonary strongyloidiasis Strongyloides stercoralis Tropical pulmonary eosinophilia Wuchereria bancrofti Brugia malayi Pulmonary dirofilariasis Dirofilaria immitis Dirofilaria repens Visceral larva migrans Toxocara canis Toxocara cati Pulmonary trichinellosis Trichinella spiralis pain, chest pain and cough are the features of pleuro- pulmonary amoebiasis. Some patients may present with respiratory distress and shock. Haemoptysis and expectoration of “anchovy souce-like” pus indicate

description

jurnal paru

Transcript of iaet08i1p49

Page 1: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 1/18

Review Article

Correspondence and reprint requests: Dr V.K. Vijayan, Director, V.P. Chest Institute, University of Delhi, Delhi-110 007,India; Phone: (Off.) 91-11-27666180, (Resi.) 91-11-27667027; Fax: 91-11-27667420; E-mail: [email protected].

Tropical Parasitic Lung Diseases

V.K. Vijayan

Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India

ABSTRACT

Though parasitic lung diseases are frequently seen in tropical countries, these are being increasingly reported from manyparts of the world due to globalisation and travel across the continents. In addition, the emergence of humanimmunodeficiency virus (HIV) infection/acquired immunodeficiency syndrome (AIDS), the frequent use of immunosuppressive drugs in many diseases and the increasing numbers of organ transplantations have resulted in arenewed interest in many tropical parasitic lung diseases. This review outlines the recent developments in the pathogenesis,diagnosis and management of common and rare parasitic lung diseases. [Indian J Chest Dis Allied Sci 2008; 50: 49-66]

Key words: Parasitic pneumonias, Pulmonary amoebiasis, Pulmonary malaria, Pulmonary paragonimiasis, Pulmonary ascariasis,Pulmonary strongyloidiasis, Tropical pulmonary eosinophilia.

INTRODUCTION

Protozoal and helminthic parasitic diseases are commonin tropical regions of the world. Helminthic parasitesinduce blood and tissue eosinophilia. However,protozoal infestations usually do not cause eosinophilia.Pulmonary disease affecting the major airways and/orparenchyma associated with either blood and/or tissueeosinophilia is classified as eosinophilic lung disease.The first four cases with minimal respiratory symptoms,fleeting pulmonary infiltrates in the chest radiographsand peripheral blood eosinophilia were described by

Loffler in 1932.1

 Although all types of eosinophilic lungdiseases are encountered in the tropics as in other partsof the world, the diseases that are particularly prevalentin the tropics are due to parasitic infestations.2 Theimportant diseases that can cause tropical parasitic lungdiseases are listed in the table.

PROTOZOAL PARASITES

Protozoal parasites that cause pulmonary diseases areEntamoeba histolytica , Leishmania donovani , malarialparasites (Plasmodium vivax, P. falciparum, P. malariae and,P. ovale), Toxoplasma gondii , Babesia microti and Babesia

divergens.

Pulmonary Amoebiasis

The causative agent of amoebiasis is the protozoanparasite, Entamoeba, histolytica   and pulmonaryamoebiasis occurs mainly by the extension from theamoebic liver abscess.3,4 Fever, right upper quadrant

Table. Tropical parasitic lung diseases

Diseases Parasites

ProtozoaPulmonary amoebiasis Entamoeba histolyticaPulmonary leishmaniasis Leishmania donovaniPulmonary malaria Plasmodium vivax,

Plasmodium falciparumPlasmodium malariaePlasmodium ovale

Pulmonary toxoplasmosis Toxoplasma gondiiPulmonary babesiosis Babesia microti,

Babesia divergensHelminths

Cestodes

Pulmonary hydatid cyst Echinococcus granulosusEchinococcus multilocularis

Trematodes

Pulmonary schistosomiasis Schistosoma haematobiumSchistosoma mansoniSchistosoma japonicum

Pulmonary paragonimiasis Paragonimus westermani

 Nematodes

Pulmonary ascariasis Ascaris lumbricoidesPulmonary ancylostomiasis Ancylostoma duodenale

Necator americanusPulmonary strongyloidiasis Strongyloides stercoralisTropical pulmonary eosinophilia Wuchereria bancrofti

Brugia malayiPulmonary dirofilariasis Dirofilaria immitis

Dirofilaria repensVisceral larva migrans Toxocara canis

Toxocara catiPulmonary trichinellosis Trichinella spiralis

pain, chest pain and cough are the features of pleuro-pulmonary amoebiasis. Some patients may present withrespiratory distress and shock. Haemoptysis andexpectoration of “anchovy souce-like” pus indicate

Page 2: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 2/18

50 Parasitic Lung Diseases V.K. Vijayan

amoebiasis.4 Pulmonary amoebic abscess without liverinvolvement has been reported to cause superior venacava syndrome.5

Diagnosis of thoracic amoebiasis is suggested by thefindings of elevated hemidiaphragm, tenderhepatomegaly, pleural effusion and basal pulmonaryinvolvement. Active trophozoites of E. histolytica can be

demonstrated in sputum or pleural pus. Microscopicexamination of stool samples may reveal cysts ortrophozoites of amoebae. The presence of amoeba in thestool does not signify that the disease is due to E.histolytica as other two non-pathogenic species found inhumans (E. dispar and E. moshkovskii) areindistinguishable morphologically. A single-roundpolymerase chain reaction (PCR) assay has beendeveloped as an accurate, rapid and effective diagnosticmethod for the detection and discrimination of thesethree Entamoeba species.6,7 Other diagnostic tests includeculture of E. histolytica and serological tests [indirecthaemagglutination test (IHA), enzyme linked

immunosorbent assay (ELISA) and indirect fluorescentantibody test (IFAT)]. A combination of serological testswith detection of the parasite by antigen detection orPCR is the best approach for diagnosis.8 Metronidazoleis the treatment of choice. Adverse reactions tometronidazole include hypersensitivity reactions.9 Drugresistance in amoebiasis can result from indiscriminateuse of metronidazole and the factors involved in drugresistance have been described.10 A luminal amoebicidaldrug, diloxanide furoate can eliminate intestinalEntamoeba cysts. Lactoferrin and lactoferricins have beenfound to kill E. histolytica and, it has been suggested thatlactoferrin and lactoferricin can be co-administered witha low dose of metronidazole to reduce toxicity of 

metronidazole.11 Studies are in progress to identifypossible vaccine candidates against amoebiasis.12

Pulmonary Leishmaniasis

Visceral leishmaniasis is caused by Leishmania donovaniand the infection is transmitted by various species of Phlebotomus, the sand fly.13 Pneumonitis, pleural effusionand mediastinal adenopathy are reported in patients co-infected with human immunodeficiency virus.Leishmania  amastigotes can be found in the alveoli,pulmonary septa and bronchoalveolar lavage (BAL)fluid.14,15 Visceral leishmaniasis has also been reported inlung transplant patients, and it has been suggested that

serological testing for latent infection due to Leishmaniaspp may be included in the pre-transplantationscreening from endemic regions.16 The drugs for thetreatment of leishmaniasis include pentavalentantimonials, amphotericin B especially the liposomeformulations and pentamidine. An oral drug,miltefosine is now available.17

Pulmonary Malaria

Malaria is caused by the obligate intraerythrocytic

protozoa of the genus Plasmodium  and is primarilytransmitted by the bite of an infected female Anophelesmosquito. Four types of malarial parasites (Plasmodiumvivax, P. falciparum, P. malaria and, P. ovale) infect man.Falciparum malariae is the most deadly type of malariainfection. Sequestration of erythrocytes containingmature forms of P. falciparum in the microvasculature of 

organs is the main feature in falciparum malaria.Quantification of the sequestrated parasite is possible bythe measurement of Plasmodium falciparum specifichistidine-rich protein 2 (PfHRP2) using a quantitativeantigen-capture enzyme-linked immunosorbent assay.18

The pulmonary manifestations range from cough tosevere and rapidly fatal non-cardiogenic pulmonaryoedema and acute respiratory distress syndrome(ARDS). It has also been reported that ARDS can occurin vivax malaria. 19 There has been no convincingevidence for the existence of true malarial pneumonitisand if it occurs, it may be due to viral and secondary

 bacterial infect ions. Gas transfer was signif icantly

impaired in patients with severe malaria.20

Light microscopy of thick and thin stained bloodsmears are the Gold standards for the diagnosis of malaria. Thin smears allow identification of malariaspecies. Radiological findings in severe falciparummalaria include lobar consolidation, diffuse interstitialoedema, pulmonary oedema and pleural effusion.Polymerase chain reaction detection of Plasmodium falciparum in human urine and saliva samples has beendescribed.21 The drugs used for treatment of severemalaria are quinine dihydrochloride, quinidinegluconate and injectable Artemisinin derivatives.Parenteral chloroquine is the drug of choice forchloroquine-susceptible P. falciparum infections and for

those rare cases of life-threatening malaria caused by P.ovale, P. malariae, and P. vivax. A point mutation in the P. falciparum  chloroquine-resistance transporter gene(PfCRT) is responsible for chloroquine-resistantfalciparum malaria.22  A study from Malawi haddemonstrated that the disappearance of the K76Tmutation in PfCRT was associated with chloroquinesusceptibility.23   Artemisinin-based combinationtherapies (artemether + lumefantrin, artesunate +amodioquine, artesunate + mefloquine or artesunate +sulfadoxine-pyrimethamine) are the best anti-malarialdrugs.24,25 The patients with respiratory failure requiresmechanical ventilation.26 One vaccine (RTS, S/ASO2)

has shown promising results in endemic areas.27

 Itappears that the best way to prevent malaria is the useof insecticide-treated bed-nets in which insecticide isincorporated into the net fibres.27

Pulmonary Toxoplasmosis

Toxoplasmosis is caused by one celled protozoanparasite, Toxoplasma gondii. Cats are the primary carriersof the organism.28 Humans get the infection by eatingparasitic cyst-contaminated raw or undercooked meat,

Page 3: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 3/18

2008; Vol. 50 The Indian Journal of Chest Diseases & Allied Sciences 51

vegetables or milk products. The symptoms of toxoplasmosis are flue-like syndrome, enlarged lymphnodes or myalgia. Infection in early pregnancy cancause foetal death, and chorioretinitis and neurologicsymptoms in the newborn. Chronic toxoplasmosis cancause chorioretinitis, jaundice, encephalitis andconvulsions. Pulmonary toxoplasmosis has been

reported with increasing frequency in patients with HIVinfection. Toxoplasma pneumonia can manifest asinterstitial pneumonia/diffuse alveolar damage ornecrotising pneumonia.29 Diagnosis of toxoplasmosis is

 based on the detection of the protozoa in body tissues.Antibody levels can be increased without active disease.A real-time PCR based assay in bronchoalveolar lavage(BAL) fluid has been reported in immunocompromisedHIV–positive patients.30 Toxoplasmosis can be treatedwith a combination of pyrimethamine and sulfadiazine.

Pulmonary Babesiosis

Babesiosis is caused by hemoprotozoan parasites,Babesia microti and Babesia divergens.31 Humans get theinfection by the bite of an infected tick, Ixodes scapularis

 but can also be infected from a contaminated bloodtransfusion. Co-infection with ehrlichiosis and Lymedisease is an important characteristic of these tick-borneillnesses.32 The parasites attack the red blood cells andcan be misdiagnosed as Plasmodium. The symptoms arefever, drenching sweats, tiredness, loss of appetite,myalgia and headache. Acute respiratory distresssyndrome occurring a few days after initiation of themedical therapy is the important pulmonarymanifestation.33 Specific diagnosis is made by theexamination of a Giemsa-stained thin blood smear,

DNA amplification using PCR or detection of specificantibody.31  Treatment is with a combination of clindamycin (600 mg every 6 hrs) and quinine (650 mgevery 8 hrs) or Atovaquone (750 mg every 12 hrs) andazithromycin (500-600 mg on first day and 250-600 mgon subsequent days) for 7-10 days.34,35

HELMINTHIC PARASITES

Helminthic parasites belonging to all three classes(Cestoidea, Trematoda and Nematoda) causepulmonary diseases in humans.

Cestodes

Hydatid lung disease in man is caused by the Cestodes,Echinococcus granulosus and Echinococcus multilocularis.

Pulmonary Hydatid Disease

The parasite species that cause hydatid disease in manare Echinococcus granulosus  and Echinococcusmultilocularis. Hydatid cysts are formed mainly in theliver and lungs. Pulmonary alveolar echinococcosis

(AE) is due to hematogeneous dissemination fromhepatic lesions.36 The adult E. granulosus resides in thesmall intestine of the definitive hosts, mainly dogs. Theintermediate hosts including man are infected byingestion of eggs excreted in the faeces of the dogs.Pulmonary symptoms include cough, fever, dyspnoeaand chest pain. Signs and symptoms can occur due to

compression of adjacent tissue by the cysts. Rupture of the cysts into a bronchus may result in haemoptysis andexpectoration of cystic fluid containing parasitemembrane, and can cause anaphylactic shock,respiratory distress, asthma-like symptoms, persistentpneumonia and sepsis.37,38 Rupture into the pleuralspace results in pneumothorax, pleural effusion andempyema. Eosinophilia and elevated IgE levels areseen when the hydatid cyst ruptures.39   Chestradiographs show solitary or multiple round opacitiesmimicking lung tumours.40 Ultrasonography using aportable ultrasound scanner has been found as reliable,inexpensive and rapid technique in community-based

screening surveys for cystic echinococcosis.41

 Thecrescent sign, Cumbo’s sign (onion peel sign), water-lilysign and air-fluid level are seen on chest radiographyand computed tomography (CT).37 Inverse crescentsign, signet ring sign and serpent sign are recognised asfeatures of pulmonary hydatid cysts in computerisedtomograpic scans.42,43  Experimentally, it has beendemonstrated that magnetic resonance imaging candetect early pulmonary AE.44 Immunodiagnostic testsusing purified E. multilocularis antigens have gooddiagnostic sensitivity and specificity for the diagnosis of AE.45

Treatment of hydatid cyst is primarily surgical.Parenchyma preserving surgery (cystotomy alone or

cystotomy and capitonnage) is the preferred treatment.Radical surgery including pneumonectomy, lobectomyand segmentectomy should be avoided. 46–48

Pharmacotherapy with albendazole or mebendazole hasalso been found to be useful especially in recurrent andmultiple cysts. The treatment of AE is radical surgicalresection of the entire parasitic lesion. In inoperablecases, pharmacotherapy with mebendazole,albendazole or praziquantel is given continuously formany years.49

Trematodes

Pulmonary schistosomiasis and paragonimiasis arecaused by trematode parasites.

Pulmonary Schistosomiasis

The schistosomes that cause human disease areSchistosoma haematobium, Schistosoma mansoni  andSchistosoma japonicum. Rarely infections with Schistosomaintercalatum and Schistosoma mekongi are reported fromAfrica and Far-East respectively.50  The schistosome eggsare passed in urine (S. haematobium) or in faeces (S.

Page 4: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 4/18

52

mansoni and S. japonicum) by the infected humans. Theeggs released in fresh water are then ingested by snails(intermediate host) in which the eggs hatch and developinto cercariae. The infective cercariae are excreted intothe water and these cercariae penetrate human skin orare ingested to penetrate the gut. The final habitat of S.haematobium is urinary bladder vesicle beds and of S.

mansoni and S. japonicum is the mesenteric beds. Theparasites can cause Schistosoma dermatitis at the site of skin penetration. Pulmonary schistosomiasis canmanifest clinically as an acute form and a chronic form.Acute symptoms can develop three to eight weeks afterskin penetration.51,52 The acute form, also known asKatayama syndrome, present with fever, chills, weightloss, diarrhoea, abdominal pain, myalgia and urticariaand is seen in non-immune patients.52,53 Pulmonarymanifestations include shortness of breath, wheezingand dry cough. Small pulmonary nodules in CT have

 been described in acute schistosomiasis.54 Patients withchronic schistosomiasis present with features of 

pulmonary hypertension and cor-pulmonale.55,56

Massive haemoptysis and lobar consolidation andcollapse have been reported in schistosomiasis. 57

Diagnosis of chronic schistosomiasis is based on thedemonstration of eggs in stool or urine by directmicroscopy or rectal/bladder biopsy. Peripheral bloodeosinophilia with mild leucocytosis, abnormal liverfunction test and elevated IgE levels are reported duringthis phase. Hepatosplenomegaly due to portalhypertension has been reported in patients infectedwith S. mansoni or S. japonicum.50 Acute schistosomiasisat presentation can be treated with corticosteroids alonefollowed by praziquantel (20-30 mg/kg orally in twodoses within 12 hours). Praziquantel can be repeated

several weeks later to eradicate the adult flukes.Artemether, an artemisinin derivative has also found to

 be useful in acute form as the drug has been found toact on juvenile forms of the schistosomes. Chronicschistosomiasis can also be treated with praziquantelwith the same dosage.50

Pulmonary Paragonimiasis

Pargonimiasis is a food borne zoonoses and is presentthroughout the world. However, 90% of the cases occurin Asia where nearly 20 million people are infected.58

Paragonimiasis is caused by infection with Paragonimusspecies and manifest as subacute or chronicinflammation of the lung. The species that are known tocause paragonimiasis in man are P. westermani,P. miyazakii, P. mexicanus, P. skrjabini, P. africanus,P. uterobilateralis, P. kellicotti, P. phillipinensis andP. heterotremus.59-61  The main species that causeparagonimiasis in man is Paragonimus westermani. Adultworms live in the lungs and the eggs are voided in thesputum or faeces. The eggs hatch in the fresh water torelease miracidiae which are ingested by the firstintermediate host, fresh water snails. The micracidiae

develop into cercariae in the snail and are released intothe water. The cercariae then invade the secondintermediate host, crustaceans (crabs or crayfish) anddevelop into infective metacercariae. The man getsinfection, when raw or undercooked crabs or crayfishesinfected with infective metacercariae are ingested.59  Theparasite from the human gut passes through several

organs and tissues to reach the lung. 59 Pulmonaryparagonimiasis manifests as fever, chest pain, chroniccough and haemoptysis.62 Chest radiographs may showinfiltrative, nodular and cavitating shadows.63 Pleuraleffusion or pneumothorax is an important finding inparagonimiasis.64 Computed tomographic scan mayshow single or multiple nodules in the lungparenchyma or pleura. Eggs can be demonstrated insputum samples, BAL fluid or lung biopsy specimens. P.westermani adult excretory-secretory products werecomposed of cysteine proteases and these molecules areinvolved in immunological events during infection.65, 66

Immunoglobulin G4 (IgG4) antibodies to an excretory-

secretory product of P. h eterotremus had accuracy,sensitivity, specificity and positive and negativepredictive values of 97.6%, 100%, 96.9%, 90% and 100%,respectively.67  Peripheral blood eosinophilia andelevated serum IgE levels are seen in more than 80% of patients with paragonimiasis.59,64 Paragonimiasis can betreated with praziquantel (75 mg/kg/day for threedays), bithionol (30 to 40 mg/kg in 10 days on alternatedays), niclofolan (2 mg/kg as a single dose) ortriclabendazole (20 mg/kg in two equal doses).59,68,69

Nematodes

Pulmonary diseases caused by nematode parasites are

pulmonary ascariasis, pulmonary ancylostomiasis,pulmonary strongyloidiasis, tropical pulmonaryeosinophilia, pulmonary dirofilariasis, visceral larvamigrans and pulmonary trichinellosis.

Pulmonary Ascariasis

Ascaris lumbricoides is the most common intestinalhelminthic infection.70 Both fertilised and unfertilisedeggs are passed in the faeces and released in the soil.71

The fertilised eggs embryonate within two to fourweeks and become infectious depending on theenvironmental conditions (moist, warm and shadedsoil). Infection occurs through soil contamination of hands or food with eggs and then swallowed. The eggshatch into larvae in the small intestine. These first-stagelarvae moult into second-stage larvae in the lumen of the small intestine. The second-stage larvae penetratethe wall of the intestine and travel via capillaries andlymphatics to the hepatic circulation and to the rightside of the heart and then reach the lungs. The second-stage larvae moult twice more in the alveoli to producethird and fourth-stage larvae. The fourth-stage larvaeare formed 14 days after ingestion, travel up to the

Parasitic Lung Diseases V.K. Vijayan

Page 5: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 5/18

2008; Vol. 50 The Indian Journal of Chest Diseases & Allied Sciences 53

trachea and are then swallowed to reach back the smallintestine. It takes approximately 10 days for the fourth-stage larvae to reach small intestine from the lung. Theeggs are produced 10 – 25 days after initial ingestion.Adult worms live approximately one to two years in theintestine.

The mechanical trauma due to adult worms and

migrating larvae is one of the factors responsible for thepathophysiologic consequences of Ascaris infection. Themigrating larvae can induce granuloma formation witheosinophils, neutrophils and macrophages. In the lung,the larvae produce a hypersensitivity reaction. Thismay result in peribronchial inflammation, increasedmucus production in the bronchi and bronchospasm. Inaddition to peripheral blood and tissue eosinophilia,Ascaris infection produces both specific and polyclonalIgE.72 Elevated levels of antibodies (IgG4) to Ascarislumbricoides have also been reported.73

1. Clinical features. Symptoms in ascariasis may bedue to the migrating larvae or due to the adult worms.

Pulmonary migration of larvae is usuallyasymptomatic. Respiratory symptoms are due to larvalpulmonary migration, airway hyperreactivity and

 bronchospasm. Symptomatic pulmonary disease mayrange from mild cough to a Loffler’s syndrome. 1, 74

Loffler’s syndrome is a self-limiting inflammation of thelungs and is associated with blood and lungeosinophilia. This syndrome can occur as a result of parasitic infestations (especially ascariasis in children)and exposure to various drugs. Patients may presentwith general symptoms of malaise, loss of appetite,fever lasting two to three days, headache and myalgia.The respiratory symptoms include chest pain, coughwith mucoid sputum, haemoptysis, shortness of breath

and wheezing.75

 There may be rapid respiratory rateand rales can be heard on auscultation. Leucocytosisparticularly eosinophilia is an important laboratoryfinding. Chest radiographs demonstrate unilateral or

 bilateral, transient, migratory, non-segmental opacitiesof various sizes. These opacities are often peripherallysituated and appear to be pleural based.76 The severityof symptoms will depend upon the larval burden.Rarely chronic eosinophilic pneumonia or symptoms of upper airway obstruction can occur.77, 78

2. Diagnosis. A diagnosis of pulmonary disease due toascariasis can be made in an endemic region in a patientwho presents with dyspnoea, dry cough, fever andeosinophilia. Sputum may show Charcot-Leydencrystals and the chest radiograph may reveal fleetingpulmonary infiltrates. Because of the occurrence of respiratory symptoms during larval pulmonarymigration, stool examination usually does not showAscaris eggs and stool samples may be negative untiltwo to three months after respiratory symptoms occur,unless the patient was previously infected. However,larvae can sometimes be demonstrated in respiratory orgastric secretions.78   It has been suggested thatmeasurement of Ascaris specific IgG4 by ELISA may be

useful in the serodiagnosis of ascariasis.79

3. Treatment. Pulmonary disease due to ascariasisusually does not require any treatment, as it is a self-limiting disease. However, the persistence of gastrointestinal ascariasis may result in repeatedepisodes of respiratory symptoms due to larvalmigration. In order to eradicate Ascaris lumbricoides from

the intestine, specific anti-helminthic treatment is given.Mebendazole and albendazole have been found to beequally effective in the treatment of ascariasis. 80

Mebendazole is given in a dose of 100 mg twice a dayfor three days. It can be also given as a single-dose of 500 mg. Albendazole is given as a single-dose of 400mg. The safety of mebendazole and albendazole duringpregnancy has not been established. A single-dose of pyrantel pamoate (11 mg/kg, maximum dose onegram) and piperazine citrate (50 to 75 mg/kg/day fortwo days) are also useful in the treatment. Pediatricdose is same as adult dose. Ivermectin has also beenfound to be useful.81

Pulmonary Ancylostomiasis

Hookworm disease in humans results from infectionswith two species, Ancylostoma duodenale and Necatoramericanus . Hookworm is a habitual blood sucker.Necator americanus causes a daily per worm blood loss of 0.01 to 0.04 mL and A. duodenale of 0.05 to 0.30 mL.82,83

Adult N. americanus can live for three to five years butA. duodenale for one year only.83, 84 Female N. americanusproduces 5000 to 10,000 eggs per day and female A.duodenale produces 10,000 to 30,000 eggs per day.83, 84

Man is the only definitive host. The eggs containingsegmented ova with four blastomers are passed out in

the faeces. A rhabditiform larva develops from each eggin the soil and this larva moults twice and develops intoa filariform larva, which is infective to man. Thefilariform larva penetrates through the intact skin.Necator americanus larvae can infect man only throughthe skin whereas A. duodenale larvae can enter thehuman host via the oral route in addition to the entrythrough the skin.83 These larvae reach pulmonarycirculation through the lymphatics and venules. Thelarvae then pierce the alveolar walls and ascend the

 bronchi, trachea, larynx and pharynx and are ultimatelyswallowed to reach the upper part of small intestine.The interval between the time of skin penetration andlaying of eggs by adult worms is about six weeks.Ancylostoma duodenale larvae can developmentally getarrested in the gut or muscle and restart developmentwhen environmental conditions become favourable.85

The arrested larvae can enter the mammary glands frommaternal somatic tissue and this is responsible forvertical transmission of ancylostomiasis to infants.86

Bronchitis and bronchopneumonia can occur whenthe larvae break through the pulmonary capillaries toenter the alveolar spaces. Chronic blood loss can resultin iron deficiency anemia. Pulmonary larval migration

Page 6: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 6/18

54

can elicit peripheral blood eosinophilia. Hookwormlarvae release a family of proteins known asancylostoma-secreted proteins (ASP). Hookworms cansecrete low-molecular weight polypeptides that inhibitclotting factor Xa and tissue factor VIIa. 87 Thesehookworm anticoagulants can facilitate blood loss thatresults from destruction of capillaries of the intestinal

mucosa.1. Clinical features. Ancylostoma dermatitis which

manifests as intense pruritis, erythema and rash(ground itch) occurs at the site of skin penetration.During pulmonary larval migration, patients maypresent with fever, cough, wheezing and transientpulmonary infiltrates in chest radiographs. This isassociated with blood and pulmonary eosinophilia. Theother characteristic feature is iron deficiency anemia dueto chronic blood loss. In severe hookworm anemia,patients may present with fatigue, exertional dyspnoea,poor concentration and cardiac murmurs. Duringmassive infection from oral ingestion of hookworm

larvae, patients can present with nausea, vomiting,cough, dyspnoea and eosinophilia and this condition istermed as Wakana disease. Prominent gastrointestinalsymptoms in hookworm disease are abdominal pain,nausea, anorexia and diarrhoea.

2. Diagnosis. A direct microscopical examination of stool demonstrates the presence of characteristichookworm eggs. Concentration method may be usedwhen the infection is light. Eosinophilia in theperipheral blood is a prominent finding. A peripheral

 blood smear examination will reveal microcytichypochromic anemia. A polymerase chain reaction(PCR) to differentiate between A. duodenale and N.americanus has been developed.88

3. Treatment. Both mebendazole and albendazole areuseful in the treatment of hookworm. Mebendazole isgiven as 100 mg twice daily for three days andalbendazole is given as a single dose of 400 mg.Pyrantel pamoate at a dose of 11mg/kg orally(maximum 1g) as a single dose has also been found to

 be useful. Recent studies have demonstrated thativermectin can also be used in the treatment of hookworm infections. Anemia can be treated with oralferrous sulphate tablets.

Pulmonary Strongyloidiasis

Strongyloides stercoralis is seen worldwide, but commonin South America, South East Asia, sub-Saharan Africaand the Appalachian region of the United States of America.89  The parasitic females live in the wall(mucous membrane) of the small intestine of man,especially in the lamina propria of the duodenum andproximal jejunum. The parasitic males remain in thelumen of the gut and they have no capacity to penetratethe mucus membrane. Eggs are laid by female parasitesand contain larvae ready to hatch. The rhabditiformlarvae emanating from the eggs pierce the mucous

membrane and reach the lumen of the intestine. Theselarvae are then passed with the faeces and eggs are,therefore, not found in the faeces. The rhabditiformlarvae can metamorphose into filariform larvae in thelumen of the bowel. These filariform larvae canpenetrate the intestinal epithelium or perianal skinwithout leaving the host. This is responsible for auto-

infection and for persistence of infection for 20 to 30years in persons who have left the endemic areas. 90 Theunique feature of the life cycle of S. stercoralis is that itcan complete its life cycle either in the human host or inthe soil. The rhabditiform larvae that are voided withthe faeces can undergo two distinct cycles in the soil:direct (host-soil-host) and indirect cycle. In direct cycle,the rhabditiform larvae directly metamorphose intofilariform larvae and can infect man through skin. Inindirect cycle, the rhabditiform larvae mature into free-living sexual forms (males and females). A secondgeneration of rhabditiform larvae is then produced andthese are then transformed into filariform larvae. The

filariform larvae can penetrate directly through the skin,invade the tissues, penetrate into the venous orlymphatic channels and are carried by the blood streamto the heart and then to the lungs. They pierce thepulmonary capillaries and enter the alveoli. Theselarvae, then, migrate to the bronchi, trachea, larynx andepiglottis and are swallowed back into the intestine. Inthe duodenum and jejunum, they develop into sexualforms to continue the life cycle.

The cutaneous reaction, as a result of penetration of the skin by the filariform larvae may be due to theimmediate hypersensitivity reaction. The cell mediatedimmunity that develops following primary infectionprevents re-infection.91 As a result, the larvae and adult

worms remain confined to the intestine and the tissueinvasion is prevented in immunocompetantindividuals. When there is immunosuppression,autoinfection is exaggerated and leads tohyperinfection. In this situation, the number of migrating larvae increases tremendously anddisseminate into many organs including lungs,meninges, brain, lymph nodes and kidneys. Theparasite can produce hyperinfection syndrome inindividuals with deficient cell mediated immunity asseen in patients with immunosuppressive therapy(especially corticosteroids) and other immune deficientstates (malnutrition, lymphoma, leukemia, etc.).92,93

Human immunodeficiency virus infected patients are ata higher risk of dissemination and this may occurwithout elevations of IgE or eosinophils.94

Newly hatched larvae are found in the LieberkuhnCrypts within the tunnels formed by the migration of parthenogenic females. A tegument derived from theparasite and host surrounds the larvae and protects theparasite and reduces any disadvantage caused byimmaturity. These larvae are capable of activemovement through the epithelium and causemechanical damage to the host. 95 The mechanical

Parasitic Lung Diseases V.K. Vijayan

Page 7: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 7/18

2008; Vol. 50 The Indian Journal of Chest Diseases & Allied Sciences 55

damage caused by the worms and the hostinflammatory response can result in abdominal pain,diarrhoea and mal-absorption. These can be aggravated

 by the associated secondary bacterial infections. Duringmigration of filariform larvae through the lungs,

 bronchopneumonia and haemorrhages in the alveolican occur. These areas are infiltrated with eosinophils.

This is associated with elevated IgE and eosinophilia inthe blood. As the larvae penetrate the intestinalmucosa, gram-negative bacteria from the gut are carried

 by the larvae on their cuticle. In addition, small breaksin the mucous membrane as the larvae penetrate theintestine also facilitate the entry of enteric bacteria intothe blood stream. The inflammation that follows suchinvasion of larvae leads to disseminatedstrongyloidiasis and this is usually fatal. It has beensuggested that hyperinfection syndrome anddisseminated strongyloidiasis can be distinguishedfrom each other.96 In hyperinfection syndrome, severesymptoms are referable to the organs usually involved

in parasitic life cycle, the lung and the intestine. In thissituation, the larval load is very high in faeces andsputum. In disseminated strongyloidiasis, there iswidespread involvement of organs that are notordinarily part of the life cycle.96  In the lung, massivepulmonary bleeding can occur due to alveolarmicrohaemorrhages. As a result of invasion of bacteriaalong with larvae, diffuse and patchy

 bronchopneumonia and pulmonary abscess can occur.1. Clinical features. It has been observed that 15% to

30% of chronically infected people may beasymptomatic.97 Although symptoms in individualswith chronic Strongyloides stercoralis infection are usuallymild, it can persist for many years due to autoinfection.

This may occasionally progress to the hyperinfectionsyndrome with high mortality especially inimmunosuppresed individuals. The relative risk of S.stercoralis  infection is increased in elderly men andpatients who had recently used corticosteroids, had ahematologic malignancy and had prior gastricsurgery.98, 99 Other risk factors include chronic lungdisease, use of histamine blockers or chronic debilitatingillness.100 Strongyloidiasis is a chronic relapsing illness of mild to moderate severity characterised by gastro-intestinal complaints (diarrhoea, pain, tenderness,nausea, vomiting), peripheral blood eosinophilia andhypoalbuminemia. 97,101

Pulmonary signs and symptoms include cough,shortness of breath, wheezing and haemoptysis. 102 Inpatients at high risk for strongyloidiasis, adultrespiratory distress syndrome and septicemia due tointestinal transmural migration of bacteria can occur asa result of pulmonary hyperinfection or disseminatedstrongyloidiasis.92,97,103,104  In addition, acute anemia,acute renal failure and systemic inflammatory responsesyndrome are also reported in hyperinfection.92

Strongyloidiasis can manifest as eosinophilic pleuraleffusion in both immunocompetent and immuno-

compromised individuals.105, 106  Rare pulmonarymanifestations include acute respiratory failure due torespiratory muscle paralysis,107 granulomatous reactionin the lung with interlobular septal fibrosis,108 syndromeof inappropriate secretion of antidiuretic hormone109

and pulmonary microcalcifications.110 A paradoxictherapeutic response of asthma to glucocorticosteroids,

in which bronchial asthma symptoms worsened aftertreatment with parenteral corticosteroids, has beendescribed in patients with Strongyloides super-infections.111 Exacerbations of chronic obstructivepulmonary disease112 and worsening of symptoms inidiopathic pulmonary fibrosis113 have also been reportedin Strogyloides stercoralis infection.

2. Diagnosis. In immunocompetent patients withstrongyloidiasis, the parasite load is usually low and thelarval output is irregular. As a result, the diagnosis of strongyloidiasis by examination of a single stoolspecimen using conventional techniques usually fails todetect larvae in up to 70% of cases.114 The diagnostic

yield can be increased by examination of several stoolspecimens on consecutive days. Examination of stool byagar plate culture method was found to be superior todirect smear and modified Baermann technique.115-117

Strongyloides stercoralis larvae can be demonstrated induodenal aspirate.75 In disseminated disease, larvae andadult parasites can be seen in sputum, urine,

 bronchoalveolar lavage fluid and other body fluids.118,119

A serological test using Centers for Disease Control(CDC) enzyme immunoassay (EIA) for detection of antibodies to strogyloidiasis was found to have asensitivity of 94.6% in patients with proven infection.120

3. Treatment.   Ivermectin, thiabendazole oralbendazole can be used for the treatment of 

strongyloidiasis in immunocompetent individualswithout complications.121,122 Ivermectin is given as 200µg/kg orally for one or two days. Thiabendazole isgiven orally as 25 mg/kg twice a day for two days.Albendazole 400 mg twice a day for five days has beenfound to be useful in the treatment of strongyloidiasis.As treatment with thiabendazole is effective only inabout 70% of patients, it has to be repeated. In immuno-compromised individuals with disseminatedstrongyloidiasis, the dose of thiabendazole has to bedoubled and the duration of treatment may be severalweeks. In addition, appropriate treatment of bacterialinfection as a result of intestinal transmural migration of 

 bacteria has to be instituted. Corticosteroids, if givenhave to be discontinued or tapered off.

Tropical Pulmonary Eosinophilia

Tropical pulmonary eosinophilia (TPE) is characterised by cough, dyspnoea and nocturnal wheezing, diffusereticulo-nodular infiltrates in chest radiographs andmarked peripheral blood eosinophilia. 123-126  Thesyndrome results from immunologic hyper-responsiveness to human filarial parasites, Wuchereria

Page 8: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 8/18

56

bancrofti and Brugia malayi.126  Tropical pulmonaryeosinophilia, one of the main causes of pulmonaryeosinophilia in the tropical countries, is prevalent infilarial endemic regions of the world especially South-East Asia.126,127 Frimodt-Moller and Barton128 in 1940described a group of patients from a sanatorium of South India who had fever, cough, and chest pain and

weight loss in association with massive bloodeosinophilia. These patients had extensive bilateralmiliary mottling in chest radiographs and were wronglydiagnosed as miliary tuberculosis. However, they werein good physical condition and did not have a highmortality as observed in miliary tuberculosis. Theydescribed this entity as “a pseudo-tuberculosiscondition associated with eosinophilia”. The name,“Tropical Eosinophilia” to this syndrome was coined byWeingarten in 1943129 who described 81 patients withsevere spasmodic bronchitis, leucocytosis and very higheosinophilia and disseminated mottlings of both thelungs. The disease was also noticed in individuals

residing in countries where filariasis was not endemic, but had visited endemic countries earlier.130,131

Tropical pulmonary eosinophilia, an occult form of filariasis, is endemic in the Indian subcontinent, South-East Asia and South Pacific islands. It has beenestimated that at least 120 million persons are infectedwith mosquito-borne lymphatic filariasis worldwide.132

However, TPE is seen in only less than 1% of filarialinfections.133 As the number of individuals travelingfrom filarial endemic areas to other parts of the worldhas increased tremendously, TPE is being increasinglyreported from countries which are not endemic tofilarial infection.130,131  Therefore, TPE should beconsidered in the differential diagnosis, if a patient

traveling from a filarial endemic region presents with“asthma-like” symptoms.134 Conversely, individualsfrom non-endemic areas visiting filarial endemicregions are more prone to develop TPE as they do nothave natural immunity against filarial infectioncompared to endemic normal subjects.

Various studies have shown that filarial infection isthe cause of TPE. 125,126  A close epidemiologicalrelationship between filarial infection and TPE wasobserved, as it had been reported that TPE was mostfrequently encountered in those regions where filariasiswas endemic.124 A positive filarial complement fixationtest (FCFT) and a positive immediate reaction to

intradermal skin tests with Dirofilaria immitis antigenshave been reported in patients with TPE. 135

Histopathological examinations had demonstratedmicrofilariae in the lungs, liver and lymph nodes of patients with TPE.136-138 The microfilariae were sheathedand had the anatomical features of Wuchereriabancrofti.136 Even though microfilariae in the blood areseen very rarely in TPE, experimental studies carriedout in a human volunteer by injection of infective larvaeof zoonotic filariasis had resulted in a syndrome similar

to that of TPE and these observations suggested that thedisease was due to filariasis. 139  The usefulness of diethylcarbamazine citrate (DEC), an antifilarial drug,in the treatment of TPE further focuses attention on itsfilarial aetiology.140,141 Elevated concentrations of filarialspecific IgG and IgE have also been reported in TPE. 142

Further evidence of human filarial infection is provided

 by the fact that maximal positive results of leukocyteadhesion phenomenon have been observed in sera frompatients with TPE using W. bancrofti compared with D.repens  and D. immitis.14 3  Biopsy of a lump in thespermatic cord in a patient with TPE showeddegenerating adult female filarial worm with uteri fullof microfilariae.144 Ultrasound examination of the scrotalarea of a patient with TPE had demonstrated livingadult Wuchereria bancrofti in the lymphatic vessels of thespermatic cord.145 The demonstration that basophilsfrom patients with TPE released greater amounts of histamine when cells were challenged with Brugia orWuchereria antigens than with Dirofilaria antigen

suggested that TPE resulted from immunologic hyper-responsiveness to human filarial parasites.142

1. Pathogenesis. The histopathological study in TPEhad shown widely scattered nodules of varying sizes (1to 5 mm) over lung surface.146 Open lung biopsy studieshave demonstrated that three types of histopathologicalreactions can be seen in TPE137,147 : (i)  interstitial,peribronchial and perivascular exudates consisting of histiocytes in patients with a short duration of symptoms (less than three weeks), (ii) acute eosinophilicinfiltrations of interstitial, peribronchial andperivascular tissues leading to the formation of eosinophilic abscesses and eosinophilic

 bronchopneumonia, and granuloma with foreign body

type giant cells in those with 1-3 months of disease, and(iii) a mixed cell type of infiltration consisting of histiocytes, eosinophils and lymphocytes with wellmarked interstitial fibrosis after six months. Apredominant histiocytic response developed two yearsafter the onset of the disease and ultimately progressedto fibrosis with marked scarring.124 In the end stage, apicture resembling fibrosing alveolitis with honey-combing might develop in some cases, if untreated.124

Lung biopsies after one month’s treatment withdiethylcarbamazine demonstrate incompletehistological regression, although symptoms subsidewithin seven days of therapy and peripheral

eosinophilia return to normal.147

The current concept of the pathogenesis of TPEsuggests that it begins with a lung parenchymalinflammation in individuals highly sensitisedimmunologically to filarial parasites. The microfilariaereleased from adult worms living in lymphatics arecleared in the pulmonary circulation, degenerate andrelease their antigenic constituents which trigger a localinflammatory process.124,138 Though the lung bears themajor brunt of the disease as a result of trapping of 

Parasitic Lung Diseases V.K. Vijayan

Page 9: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 9/18

2008; Vol. 50 The Indian Journal of Chest Diseases & Allied Sciences 57

microfilariae in the pulmonary circulation, the antigenicmaterial released from the microfilariae can reach thesystemic circulation and cause extra pulmonarymanifestations. Bronchoalveolar lavage (BAL) studieshave demonstrated that TPE is characterised by intenseeosinophilic inflammatory process in the lowerrespiratory tract.148 The concentration of eosinophils in

the lower respiratory tract is many folds greater thanthat in the blood, suggesting that eosinophilsaccumulate selectively in the lung parenchyma.148

Electron microscopic examination of lung eosinophilshad shown marked alterations consisting of severedegranulation with loss of both the cores and theperipheral portions of the granules, suggesting thateosinophils were in an activated state. 148   The

 bronchospasm in TPE may result from leukotrienesreleased by the eosinophils.149 In order to explore themechanisms underlying the eosinophil-mediatedinflammation of TPE, many of the cytokines,chemokines, eosinophilic granular proteins and

leukotrienes were studied in BAL fluid, serum andsupernatants from pulmonary and blood leucocytes.150

Of the many mediators studied, eosinophil-derivedneurotoxin (EDN) and macrophage inflammatoryprotein-1! (MIP-1!) levels were higher for TPE patientsthan for the non-TPE control groups. Eosinophil-derived neurotoxin levels were also higher in the BALfluid than in the serum, suggesting compartmenta-lisation of the response. These findings suggest thatEDN, an RNAse capable of damaging the lungepithelium, plays an important role in the pathogenesisof TPE.150

Patients with TPE show striking elevations of totalIgE, IgG (Hypergammaglobulinemia) and filarial

specific IgG, IgM and IgE antibodies in peripheral bloodand lung epithelial lining fluid (ELF).148,151 A major IgE-inducing antigen (Bm23-25) of the filarial parasite,Brugia malayi has been identified from patients withtropical pulmonary eosinophilia.152  This Bm23-25antigen has been found to be the homolog of theenzyme, gamma-glutaryl transpeptidase light chainsubunit153 and expresses mainly in the infective L3larvae that have been shown to induce specific IgEantibodies in infection-free endemic normalsubjects.154,155 In addition, Brugia malayi gamma-glutaryltranspeptidase can induce IgG1 and IgE antibodyproduction in patients with TPE.154 Bronchoalveolar

lavage fluid of patients with TPE contains IgEantibodies that recognise Brugia malayi antigen, Bm23-25.152 There is a molecular mimicry between the parasitegamma-glutaryl transpeptidase and the human gamma-glutaryl transpeptidase present on the surface of pulmonary epithelium.152,156 A marked reduction in lungELF filarial-specific IgG and IgE levels within 6-14 daysof therapy with diethylcarbamazine has beenobserved.151 Immunoblot comparison of the antigenrecognition patterns of lung ELF and serum antigens

have recognised a distinct subset of filarial specificantigens recognised by the antibodies in the peripheral

 blood. 151  Thus, the profound antibody response tofilarial infection especially to filarial gamma-glutaryltranspeptidase, observed in the lungs of patients withTPE, plays an important role in the pathogenesis of tropical eosinophilia.155

2. Clinical features. Tropical pulmonary eosinophilia isa systemic disease involving mainly the lungs, but otherorgans such as liver, spleen, lymph nodes, brain, gastro-intestinal tract, etc., may also be involved.124,157  Thedisease occurs predominantly in males, with a male tofemale ratio of 4:1, and is mainly seen in older childrenand young adults between the age 15-40 years.124,126 Thesystemic symptoms include fever, weight loss andfatigue.

Patients with TPE usually present with respiratorysymptoms that include paroxysmal cough,

 breathlessness, and wheeze and chest pain. Thesymptoms occur predominantly at night, but can persist

during the day. Severe cough can lead to fractured ribs.Sputum is usually scanty, viscous and mucoid. Thesputum often shows clumps of eosinophils,124 and rarelyCharcot-Leyden crystals are observed.129   Onexamination, patients are often breathless. Bilateralscattered rhonchi and rales may be heard onauscultation.

Leucocytosis with an absolute increase in eosinophilsin the peripheral blood is the hallmark of TPE.Spontaneous fluctuations in the eosinophil count canoccur. Absolute eosinophil counts are usually morethan 3000 cells/mm3 and may range from 5000 to80000.158 Erythrocyte sedimentation rate is elevated in90% of cases and returns to normal following specific

treatment. 124   Microfilariae are rarely seen in theperipheral blood. As patients with TPE especially fromendemic areas can be simultaneously infected withother helminthic parasites, stool examination mayreveal ova or larvae of other helminths (Ascaris ,Ancylostoma , whipworm, and Strongyloides) in 20% of patients with TPE. This observation does not deter thephysician from making a diagnosis of TPE, if otherconditions for diagnosis are fulfilled.

The chest radiological features of TPE includereticulonodular shadows predominantly seen in midand lower zones and miliary mottling of 1-3 mm indiameter often indistinguishable from miliary

tuberculosis. In addition, there may be prominent hilawith heavy vascular markings.159-162 Twenty percent of patients have a normal chest radiograph.124  In patientswith a long standing history, a few patients have honey-comb lungs.124  Radiological improvement occurs onspecific therapy with DEC, but some degree of radiological abnormality persists in some patients.163,164

Computed tomographic scan studies have shown bronchiectasis , air trapping, calcificat ion andmediastinal lymphadenopathy in patients with TPE.165

Page 10: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 10/18

58

Lung function tests reveal mainly a restrictiveventilation defect with superimposed airwaysobstruction.166-169 Single breath carbon monoxide transferfactor (TLCO) is reduced in 88% of untreated patientswith TPE.168 The reduction in TLCO is due to reducedpulmonary membrane diffusing capacity (Dm) whichitself is due to a reduction in single breath alveolar

volume (VA).168, 169 Pulmonary capillary blood volume(Vc) is normal.169 Following three weeks treatment withDEC, although there were significant rise in variouspulmonary function parameters, forced expiratoryvolume in the first second (FEV

1), forced vital capacity

(FVC), TLCO, Dm and expiratory flow rates continuedto be significantly lower than the control subjects.164, 169,170

Single breath transfer factor and total lung capacity(TLC) were negatively correlated with total number of inflammatory cells (alveolar macrophages,lymphocytes, eosinophils and neutrophils) in BALfluid.171 The lung eosinophils were negatively correlatedwith TLCO and KCO (diffusion per liter of alveolar

volume), but not with FVC, FEV1 or TLC; whereasalveolar macrophages and lymphocytes were associatedwith a reduction in lung volumes but not with TLCO,suggesting that different cell types might be responsiblefor different aspects of lung damage.171

Arterial hypoxemia (PaO2  < 80 mmHg) was

observed in 41% of untreated TPE patients. 172 Most of them had only mild arterial hypoxemia.163, 172-175

Hypocapnia (PaCO2 < 35 mmHg) was present in 16% of 

patients.172 Arterial PO2 and PCO

2 improve following

treatment at one month and normal PaO2 and PaCO

2

are maintained in most patients one year after thetreatment.163 Ventilation-perfusion scintiscanning hasdemonstrated that hypoxemia in some patients with

TPE is due to disturbed ventilation-perfusionrelationship.175

Lymphadenopathy may occur in TPE, especially inchildren.12 4  Cardiovascular changes especiallyelectrocardiographic abnormalities,176, 177 pericarditis178 ,pericardial effusion,179 and cor-pulmonale180 have also

 been reported. Tropical pulmonary eosinophilia mayalso present with gastro-intestinal,124 skeletal muscle181

and central nervous system manifestations.157

3. Diagnosis. Infestations with helminths (cestodes,nematodes and trematodes) are the commonest causesof pulmonary eosinophilia in tropical countries.182

Helminths that cause pulmonary eosinophilia

frequently are Ascaris, Ancylostoma, Strongyloides,Toxocara, Schistosoma, Paragonimus, Trichinella, andEchinococcus,   besides occult fi lariasis . Tropicalpulmonary eosinophilia of filarial aetiology maysometimes be clinically indistinguishable from TPE-likesyndrome caused by other helminths, except that someof these patients do not respond to DEC. In addition,elevated levels of antifilarial antibodies (IgG, IgG4 andIgE) are also observed in patients with TPE-likesyndrome.183 It has also been demonstrated that sera

from people of an area, not endemic for filariasis, butharboring intestinal helminths, have antifilarialantibodies that recognise antigens of microfilariae of Wuchereria bancrofti.184 These observations underscorethe need for developing a new diagnostic test todifferentiate filarial TPE from TPE-like syndromes,caused by other helminths. Other infectious causes of 

pulmonary eosinophilia include infections with Brucella,Coccidioides, Corynebacterium pseudotuberculosis and Mycobacterium tuberculosis.185,186 Non-infectious causes of pulmonary eosinophilia include allergic broncho-pulmonary aspergillosis (ABPA), bronchial asthma,acute eosinophilic pneumonia, chronic eosinophilicpneumonia, idiopathic hypereosinophilic syndrome,cryptogenic pulmonary fibrosis, Wegner’sgranulomatosis, lymphomatoid granulomatosis,eosinophilic granuloma of the lung, Churg-Strausssyndrome and drug hypersensitivity reactions. Till adiagnostic test that can differentiate filarial TPE fromother TPE-like syndrome is available, the following

diagnostic criteria can be used for the diagnosis of TPE:(a) appropriate exposure history (mosquito bite) in anendemic area of filariasis, (b) a history of paroxysmalnocturnal cough and breathlessness, (c)  chestradiographic evidence of pulmonary infiltrations, (d)leucocytosis in blood, (e) peripheral blood eosinophilsmore than 3000 cells per cumm, (f) elevated serum IgElevels, (g) elevated serum antifilarial antibodies (IgGand/or IgE), and (h)   a clinical response todiethylcarbamazine.123

4. Treatment. The efficacy of organic arsenicals in thetreatment of TPE was reported by Weingarten. 129

However, the serious toxicity of arsenicals made itunacceptable for the treatment of TPE. The efficacy of 

diethylcarbamazine in TPE was proved by Ganatra andLewis140 and Danaraj.187 Baker et al141 had shown in acontrolled trial that DEC in a dose of 5 mg/kg/day forseven days was sufficient in the treatment of TPE.Danaraj187 treated patients from Singapore with a higherdose of 18 and 30 mg/kg. The standard treatmentrecommended by the World Health Organization fortreatment of TPE is oral DEC (6 mg/kg per day) forthree weeks.188   One month after the start of thetreatment most patients show marked symptomatic andradiographic improvement; and significant improve-ment in almost all aspects of lung functions.164, 168-170

However, there is an incomplete reversal of clinical,

hematological, radiological and physiological changesin TPE one month after a three-week course of DEC.164

In addition, mild alveolitis characterised byhypercellular lavage fluid due to a significant increasein alveolar macrophages and eosinophils persists at onemonth.189 Patients evaluated 12 ± 2 months following astandard three-week course of DEC were found to havemild, persistent symptoms referable to the lung. Chestradiographic abnormalities, blood eosinophilia,elevated serum IgE and filarial specific IgG and lung

Parasitic Lung Diseases V.K. Vijayan

Page 11: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 11/18

2008; Vol. 50 The Indian Journal of Chest Diseases & Allied Sciences 59

function changes consistent with chronic mildinterstitial lung disease persist.190 These patients havepersistent eosinophilic alveolitis and the lowerrespiratory tract inflammatory cells releasespontaneously exaggerated amounts of superoxide (O

2)

and hydrogen peroxide (H2O

2).190   Treatment with

prednisolone significantly reduced the lower respiratory

tract inflammation and the release of oxidants. 191

Patients with chronic eosinophilic alveolitis at 12thmonth had a significantly lower TLCO compared topatients with normal eosinophils in lower respiratorytract.192 Thus, following a standard three-week course of DEC therapy, most patients show improvement, but notcomplete resolution of TPE and many are left withchronic respiratory tract inflammation and a mild formof interstitial lung disease.124,163,190 Relapses followingtreatment occur in 20% of patients followed up for fiveyears.124  Because of persistent mild interstitial lungdisease and high relapse rates in TPE, it has beenrecommended that repeated monthly course of DEC at

2-3 monthly intervals for a period of 1-2 years may beuseful.124

Earlier workers had shown that steroids wereeffective in the treatment of TPE.193 Since interstitial lungdisease is found to occur in a proportion of patients withTPE despite standard therapy with DEC, a controlledclinical trial is needed to know the optimum dose andduration of DEC therapy with or without steroids.Ivermectin, an antifilarial drug,194 has not been found to

 be useful in the treatment of tropical pulmonaryeosinophilia.195

Pulmonary Dirofilariasis

Pulmonary dirofilariasis is a zoonotic infection caused byfilarial nematodes, Dirofilaria immitis and Dirofilaria repens.Though the parasite has been named as “dog heartworm”, there are evidences that D. immitis is a vascularparasite.196 Humans are accidental hosts of this parasitewhich is transmitted to man by the mosquito. Theparasites are usually seen in the pulmonary artery wherethey produce an embolism ultimately leading to theformation of a pulmonary nodule or “coin lesion”.Human dirofilariasis is increasingly been reportedworldwide.197, 198 Nearly 50% of the subjects infected withdirofilariasis are asymptomtic. Clinical symptoms arechest pain, cough, fever, haemoptysis and dyspnoea.

Computed tomographic scan may show a well-definednodule with smooth margin connected to an arterial branch.199 Positron emission tomography (PET) scan candemonstrate hyper-metabolic activity in a pulmonaryinfarct secondary to dirofilariasis.200 A PCR-baseddiagnosis of D. repens in human pulmonary dirofilariasishas been reported.201  A definitive histopathologicaldiagnosis of pulmonary dirofilariasis can be made intissue specimens obtained by wedge biopsy, video-assisted thoracoscopy or rarely by fine needle biopsy.198

There is no specific treatment for human dirofilariasis.

Visceral Larva Migrans

Toxocara  larva migrans syndromes are importantzoonotic infections.202,203 Certain nematode parasiteswhen enter into an unnatural host (e.g., man) may not

 be able to complete their life cycle and their progress isarrested in the “unnatural host”. If the entry of such

parasitic larvae is through skin penetration, it causescutaneous larva migrans (creeping eruption). If theentry is via oral route, visceral larva migrans result.Cutaneous larva migrans is caused by the parasite,Ancylostoma brazeliense which is a parasite of dogs andcats. The common parasites that cause visceral larvamigrans (VLM) and esonophilic lung disease in man area dog ascarid (Toxocara canis) and less commonly a catascarid (Toxocara catis). Human toxocariasis occurs in allparts of the world wherever there is a large pool of infected dogs.204, 205

Adult Toxocara canis and Toxocara catis live in theintestines of dogs and cats, respectively. In dogs, there is

vertical transmission of larvae from infected pregnant bitches to their puppies. In cats, lactogenic transmissionoccurs to kittens. Older animals acquire infection byingestion of eggs of the parasites from contaminatedsoil. The life cycle is then completed in definitive hosts,dogs and cats. Eggs are passed by the animals in thefaeces and embryonate in the soil. These embryonatedtoxocara eggs, when ingested by an intermediate host(e.g., man), hatch into infective larvae in the intestine.The infective larvae penetrate the intestinal wall and arecarried by the circulation to many organs includingliver, lungs, muscles, central nervous system and eye.The progress of the larvae is arrested in these sites of theintermediate host by the formation of a granulomatous

lesion. In man, the larvae never develop into adultworms. Therefore, infected man never excretes toxocaraeggs in the faeces.

Visceral larva migrans is characterised byleucocytosis and eosinophilia. The larva induces agranulomatous reaction in the tissues containingeosinophils and multinucleated giant cells. Larvae canget encapsulated within the granuloma where they areeither destroyed or persist for many years in a viablestate. Granulomata are found in the liver, lungs, centralnervous system and eyes. Later fibrosis and calcificationoccur. Toxocara canis larvae produces excretory-secretoryproteins which may be responsible for inducing a Th-2-

type CD4 cellular immune response206

 and this ischaracterised by eosinophilia and IgE production.Toxocara canis larva has a labile mucinous surface coat,207

and eosinophils have been shown to bind on thissurface.208 Larval antigens induce release of histamine209

and also can cross-react with human A and B bloodgroup antigens.210

1. Clinical features. Human toxocariasis can manifestin different forms: visceral larva migrans, ocular larvamigrans (OLM), neurological toxocariasis, commontoxocariasis and “covert” toxocariasis.203 Visceral larva

Page 12: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 12/18

60

migrans is commonly seen in children. However, mostof children infected with Toxocara sp. are asymptomatic.The main symptoms in patients with visceral larvamigrans are fever, cough, wheezing, seizures, anemiaand fatigue. Pulmonary manifestations are reported in80% of cases and patients may present with severeasthma. Scattered rales and rhonchi are heard on

auscultation. There will be intense blood eosinophilia.Chest radiograph may reveal focal patchy infiltrates. Insome cases, severe eosinophilic pneumonia may lead torespiratory distress.211-213 Other clinical features includegeneralised lymph node enlargement, hepatomegalyand splenomegaly. Ocular larva migrans due to T. canismay present with pain in the eyes, white pupil,strabismus or unilateral loss of vision. Neurologicalmanifestations of VLM include eosinophilic meningitis,encephalitis, seizures, arachnoiditis and myelopathy.Common toxocariasis is described mainly in adults andmanifests as weakness, pruritis, rash, difficult breathingand abdominal pain. This syndrome is associated with

eosinophilia, increased total serum IgE level andelevated antibody titers to T. canis.203, 214 In “covert”toxocariasis, children present with fever, anemia,headache, abdominal pain, sleep disturbances, nausea,vomiting, pharyngitis, pneumonia, cough, wheeze,cervical lymphadenitis, hepatomegaly and limb pains.215

A quarter of patients with “covert” toxocariasis have noeosinophilia.

2. Diagnosis. Visceral larva migrans (VLM) is usuallyreported in young children with a history of pica. Ahistory of exposure to puppies or dogs supports thediagnosis of VLM. These children usually present withfever, cough, wheezing, eosinophilia and hepatomegaly.Chest radiograph may show patchy infiltrates. Non

specific changes include hypergamma-globulinemiaand elevated isohemaglutinin titers to A and B bloodgroup antigens. Serological tests by ELISA methodusing excretory-secretory proteins obtained fromcultured T. canis may be useful in the diagnosis.216

Cross-reactivity with other helminths limits theusefulness of this test in endemic areas. Detection of IgEantibodies by ELISA217 and toxocara excretory-secretoryantigens by Western blotting procedure have also beenreported for diagnosis.218 However, serodiagnosticprocedures can not distinguish between past andpresent infections. Histopathological examination of lung or liver biopsy specimens may demonstrategranulomas with eosinophils, multinucleated giant cellsand fibrosis. Since man is not the definitive host of Toxocara sp., eggs or larvae cannot be demonstrated inthe faeces.

3. Treatment. Visceral larva migrans is a self-limitingdisease and there may be spontaneous resolution.Therefore, mild to moderately symptomatic patientsneed not require any drug therapy. Patients with severeVLM can be treated with thiabendazole, mebendazoleor diethylcarbamazine. Treatment with antihelminthicsmay exacerbate the inflammatory reactions in the

tissues due to the killing of larvae. It is, therefore,advised to combine antihelminthic treatment withcorticosteroids. Diethylcarbamazine can be given in adose of 6 mg/kg/day for 21 days. 219 Mebendazole can

 be prescribed in a dose of 20 to 25 mg/kg/day for 21days.220 Albendazole 10 mg/kg/day for five days has

 been shown to have a moderate effect compared to

thiabendazole.221

Pulmonary Trichinellosis

Human trichinellosis is an important food-bornezoonosis. Five species of Trichinella (T. spiralis , T. nativa ,T. nelsoni , T. britovi and T. pseudospiralis) can infectman.222 The most important species that infect man isTrichinella spiralis. The parasite has a direct life cyclewith complete development is one host (pig, rat orman). However, two hosts are required to complete thelife cycle and for the preservation of the species fromextinction. Man gets infection from raw and partiallycooked pork, when infected pig’s muscle containing

larval trichinellae is eaten by man. The infective larvaein the muscle are surrounded by a host capsule which isa modified striated muscle known as “nurse” cell. Inthe stomach, the “nurse” cell is digested and the freelarva is liberated. The larvae develop into adults (malesand females) in the duodenum and jejunum. Thenewborn larvae produced by female parasites passthrough the lymphatics or blood vessels to reach thestriated muscles.223 The larvae undergo encystment inthe muscle and a host capsule develops around thelarvae. Later on, it may get calcified. The life cycle iscompleted when infected muscle is ingested by asuitable host.

The Th-2 cells play an important role in the

pathogenesis of the disease. These cells release IL-5 andIL-4 and these cytokines are responsible for eosinophiliaand increased IgE production. 224   The commonsymptoms of trichinellosis are muscle pain, periorbitaloedema, fever and diarrhoea.225,226  Pulmonarysymptoms include dyspnoea, cough and pulmonaryinfiltrates. Dyspnoea may be due to the involvement of diaphragm.227  Leucocytosis, eosinophilia and elevatedlevels of serum muscle enzymes (creatinephosphokinase, lactate dehydrogenase, aldolase andamino transferase) are important laboratory findings.An enzyme-linked immunosorbant assay (ELISA) fordetection of anti-Trichinella antibodies using excretory-

secretory antigens may be useful in the diagnosis of T.spiralis.228 A definitive diagnosis can be made by muscle biopsy (usually deltoid muscle) that may demonstratelarvae of T. spiralis.227   Symptomatic treatment of trichinellosis includes analgesics and corticosteroids.Specific treatment is with mebendazole 200 to 400 mgthree times a day for three days followed by 400 to 500mg three times a day for 10 days. Albendazole can begiven in a dosage of 400 mg per day for three daysfollowed by 800 mg per day for 15 days. Trichinellosiscan be prevented by consuming properly cooked pork.

Parasitic Lung Diseases V.K. Vijayan

Page 13: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 13/18

2008; Vol. 50 The Indian Journal of Chest Diseases & Allied Sciences 61

REFERENCES

1. Loffler W. Zur differential-diagnose der lungeninfiltrieurgen: 11.Uber fluchtige succedan – infiltrate (mit eosinophile). Bietr KlinTuberk 1932; 79: 368-92.

2. Vijayan VK. How to diagnose and manage commonparasitic pneumonias. Curr Opin Pulm Med 2007; 13: 218-24.

3. Ackers JP, Mirelman D. Progress in research on Entamoebahistolytica pathogenesis. Curr Opin Microbiol 2006; 9: 367-73.4. Shamsuzzaman SM, Hashiguchi Y. Thoracic amoebiasis. Clin

Chest Med 2002; 23: 479-92.5. Lichtenstein A, Kondo AT, Visvesvara GS, Fernandez A,

Paiva EF, Mauad T, et al. Pulmonary amoebiasis presentingas superior vena cava syndrome. Thorax 2005; 60: 350-2.

6. Hamzah Z, Petmitr S, Mungthin M, Leelayoova S,Chavalitshewinkoon-Petmir P. Differential detection of Entamoeba histolytica, Entamoeba dispar  and Entamoebamoshkovskii  by a single-round PCR assay.  J Clin Microbiol2006; 44: 3196-200.

7. Haque R, Petri WA Jr. Diagnosis of amoebiasis inBangladesh. Arch Med Res 2006; 37: 273-6.

8. Tanyuksel M, Petri WA Jr. Laboratory diagnosis of amoebiasis. Clin Microbiol Rev 2003; 16: 713-29.

9. Garcia–Rubio I, Martinez–Cocera C, Santos Magadan S,Rodriguez-Jimenez B, Vasquez-cortes S. Hypersensitivityreactions to metronidazole. Allergol Immunopathol (Madr)2006; 34: 70-2.

10. Bansal D, Malla N, Mahajan RC. Drug resistance inamoebiasis. Indian J Med Res 2006; 123: 115-8.

11. Leon-Sicairos N, Reyes-Lopez M, Ordaz-Pichardo C, de laGarza M. Microbicidal action of lactoferrin and lactoferricinand their synergistic effect with metronidazole in Entamoebahistolytica. Biochem Cell Biol 2006; 84: 327-36.

12. Lotter H, Tannich E. The current status of an amoebiasisvaccine. Arch Med Res 2006; 37: 292-6.

13. Piscopo TV, Mallia AC. Leishmaniasis. Postgrad Med J  2006;82: 649-57.

14. Russo R, Laguna F, Lopez-Velez R, Medrano FJ, Roenthal E,Cacopardo B, et al. Visceral leishmaniasis in those infected

with HIV: clinical aspects and other opportunistic infections.Ann Trop Med Parasitol 2003; 97 (Suppl. 1): S99-S105.15. Benzie AA, Goldin RD, Walsh J. A case report of seronegative

pulmonary leishmaniasis in an HIV-hepatitis C co-infectedpatient. HIV Med 2006; 7(Suppl.1):36 (abstract no. P100).

16. Morales P, Torres JJ, Salavert M, et al. Visceral leishmaniasisin lung transplantation. Transplantation Proc 2003; 35: 2001-03.

17. Croft SL, Engel J. Miltefosine:discovery of the anti-leishmanial activity of phospholipid derivatives. Trans R SocTrop Med Hyg 2006; 100 (Suppl. 1): 54-8.

18. Dondorp AM, Desakorn V, Pongtavornpinyo W, et al.Estimation of the total parasite biomass in acute falciparummalaria from plasma PfHRP2. PLoS Med  2005;2:e204(doi:10.1371/journal. pmed.0020204). (Accessed August 30,2007 at http://www.ncbi.nlm.nih.gov/sites/entrez.

19. Lomar AV, Vidal JE, Lomar FP, Barbas CV, Matos GJ, BoulosM. Acute respiratory distress syndrome due to vivaxmalaria: case report and literature revirew. Braz J Infect Dis2005; 9: 425-30.

20. Maguire GP, Handojo T, Pain MC, Kenangalem E, Price RN,Tjitra E, et al. Lung injury in uncomplicated and severefalciparum malaria: a longitudinal study in Papua,Indonesia. J Infect Dis 2005; 192: 1966-74.

21. Mharakurwa S, Simoloka C, Thuma PE, Shiff CJ, Sullivan DJ.PCR detection of Plasmodium falciparum in human urine andsaliva samples. Malar J 2006; 5: 103.(doi:10.1186/1475-2875-5-103).

22. Djimde A, Doumbo OK, Cortese JF, Kayentao K, Doumbo S,Diourte Y, et al. A molecular marker for chloroquine-resistantfalciparum malaria. N Engl J Med 2001; 344: 257-63.

23. Laufer MK, Thesing PC, Eddington ND, Masonga R,Dzinjalamala FK, Takala SL, et al. Return of chloroquineantimalarial efficacy in Malawi. N Engl J Med 2006; 355: 1959-66.

24. Mutabingwa TK. Artemisinin-based combination therapies

(ACTs): best hope for malaria treatment but inaccessible tothe needy! Acta Trop 2005; 95: 305-15.25. World Health Organization. Guidelines for the treatment of 

malaria. Geneva: World Health Organization, 2006; 1-266.26. Talwar A, Fein AM, Ahluwalia G. Pulmonary and critical

care aspects of severe malaria. In: Sharma OP, editor LungBiology in Health and Disease: Tropical Lung Diseaes; 2nd edn.New York: Taylor and Francis Group; 2006: pp 255-77.

27. Greenwood BM, Bojang K, Whitty CJ, Targett GA. Malaria.Lancet 2005; 365: 1487-98.

28. Dodds EM. Toxoplasmosis. Curr Opin Ophthalmol 2006; 17:557-61.

29. Nash G, Kerschmann RL, Herndier B, Dubey JP. Thepathological manifestations of pulmonary toxoplasmosis inthe acquired immunodeficiency syndrome. Hum Pathol 1994;25: 652-8.

30. Petersen E, Edvinsson B, Lundgren B, Benfield T, EvengardB. Diagnosis of pulmonary infection with Toxoplasma gondiiin immunocompromised HIV-positive patients by real-timePCR. Eur J Clin Microbiol Infect Dis 2006; 25: 401-4.

31. Krause PJ. Babesiosis. Med Clin North Am 2002; 86:361-73.32. Swanson SJ, Neitzel D, Reed KD, Belongia EA. Co infections

acquired from ixodes ticks. Clin Microbiol Rev 2006; 19: 708-27.

33. Boustani MR, Lepore TJ, Gelfand JA, Lazarus DS. Acuterespiratory failure in patients treated for babesiosis. Am J Respir Crit Care Med 1994; 149: 1689-91.

34. Krause PJ. Babesiosis diagnosis and treatment. Vector BorneZoonotic Dis 2003; 3: 45-51.

35. Raju M, Salazar JC, Leopold H, Krause PJ. Atovaquone andazithromycin treatment for babesiosis in an infant. PediatrInfect Dis 2007; 26: 181-3.

36. Gottstein B, Reichen J. Hydatid lung disease(echinococcosis/hydatidosis). Clin Chest Med 2002; 23: 397-408.

37. Kuzucu A. Parasitic diseases of the respiratory tract. CurrOpin Pulm Med 2006; 12: 212-21.

38. Fanne RA, Khamaisi M, Mevorach D, Leitersdorf E,Berkman N, Laxer U, et al. Spontaneous rupture of lungechinococcal cyst causing anaphylactic shock andrespiratory distress syndrome. Thorax 2006; 61: 550.

39. Savani DM, Sharma OP. Eosinophilic lung disease in thetropics. Clin Chest Med 2002; 23: 377-96.

40. Gencer M, Ceylan E. Pulmonary echinococcosis withmultiple nodules mimicking metastatic lung tumor in chestradiography. Respiration (DOI. 10.1159/000091141) Jan 23,2006. (Accessed August 30, 2007 at http://www.ncbi.nlm.nih.gov/sites/entrez/.

41. Kilimcioglu AA, Ozkol M, Bayindir P, Girginkardesler N,Ostan I, Ok UZ. The value of ultrasonography alone inscreening surveys of cystic ecchinococcosis in children inTurkey. Parasitol Int 2006; 55: 273-5.

42. Erdem CZ, Erdem LO. Radiological characteristics of pulmonary hydatid disease in children: less commonradiological appearances. Eur J Radiol 2003; 45: 123-8.

43. Kilic D, Tercan F, Sahin E, Bilen A, Hatipoglu A. Unusualradiologic manifestations of the echinococcus infection in thethorax. J Thorac Imaging 2006; 21: 32-6.

44. Asanuma T, Kawahara T, Inanami O, Nakao M, Nakaya K,Ito A, et al.  Magnetic resonance imaging of alveolar

Page 14: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 14/18

62

echinococcosis experimentally induced in the rat lung. J Vet Med Sci 2006; 68: 15-20.

45. Ito A, Sako Y, Yamasaki H, Mamuti W, Nakaya K, Nakao M,et al. Development of Em 18-immunoblot and Em 18-ELISAfor specific diagnosis of alveolar echinococcosis. Acta Trop2003; 85: 173-82.

46. Kavukcu S, Kilic D, Tokat AO, Kutlay H, Cangir AK, Enon S,et al. Parenchyma-preserving surgery in the management of 

pulmonary hydatid cysts. J Invest Surg 2006; 19: 61-8.47. Hasdiraz L, Oguzkaya F, Bilgin M. Is lobectomy necessary inthe treatment of pulmonary hydatid cysts? ANZ J Surg 2006;76: 488-90.

48. Dincer SI, Demir A, Sayar A, Gunluoglu MZ, Kara HV,Gurses A. Surgical treatment of pulmonary hydatid disease:a comparison of children and adults. J Pediatr Surg 2006; 41:1230-6.

49. Gottstein B, Reichen J. Hydatid lung disease. In: Sharma OP,editor Lung Biology in Health and Disease: Tropical LungDisease; 2nd edn. New York: Taylor and Francis Group; 2006:327-50.

50. Schwartz E. Pulmonary schistosomiasis. Clin Chest Med 2002;23: 433-43.

51. Schwartz E, Rozenman J, Perelman N. Pulmonarymanifestations of early Schistosoma infection in nonimmune

travelers. Am J Med 2000; 109: 718-22.52. Walt F. The Katayama syndrome. S Afr Med J 1954; 28: 89-93.53. Doherty JF, Moody AH, Wright SG. Katayama fever; an

acute manifestation of schistosomiasis. Br Med J 1996; 313:1071-2.

54. Nguyen LQ, Estrella J, Jett EA, Grunvald EL, Nocholson L,Levin DL. Acute schistosomiasis in nonimmune travelers:chest CT findings in 10 patients. Am J Roentgenol 2006; 186:1300-3.

55. Lapa MS, Ferreira EV, Jardim C, Martins BC, Arakaki JS,Souza R. Clinical characteristics of pulmonary hypertensionpatients in two reference centers in the city of Sao Paulo. RevAssoc Med Bras 2006; 52: 139-43.

56. Morris W, Knauer M. Cardiopulmonary manifestations of schistosomiasis. Semin Respir Infect 1997; 12: 159-70.

57. Sersar SI, Abulmaaty RA, Elnahas HA, Moussa SA, Shisa

UA, Ghafar WA, et al. A diagnostic dilemma of right lowerlobe collapse caused by pulmonary bilharsiasis. Heart LungCirc 2006; 15: 50-2.

58. Strobel M, Veasna D, Saykham M, Wei Z, Tran DS, Valy K,et al. Pleuro-pulmonary paragonimiasis. Med Mal Infect 2005;35: 476-81.

59. Nakamura–Uchiyama F, Mukae H, Nawa Y.Paragonimiasis: a Japanese perspective. Clin Chest Med 2002;23: 409-20.

60. Blair D, Xu ZB, Agatsuma T. Paragonimiasis and the genusparagonimus. Adv Parasitol 1999; 42: 113-222.

61. King CH. Pulmonary flukes. In: Mahamoud AAF, editorLung Biology in Health and Disease: Parasitic Lung Disease .New York: Marcel Dekker; 1997: pp 157-169.

62. Xu ZB. Studies on clinical manifestations, diagnosis andcontrol of paragonimiasis in China. Southeast Asian J Trop

 Med Pub Health 1991; 22 (Suppl. 1): 345-8.63. Suwanik R, Harinasuta C. Pulmonary paragonimiasis: an

evaluation of roentgen findings in 38 positive sputumpatients in an endemic area in Thailand. Am J Roentgenol1959; 81: 236-44.

64. Mukae H, Taniguchi H, Matsumoto N, Liboshi H, Ashitani J,Matsukura S, et al.  Clinicoradiologic features of pleuropulmonary Paragonimus westermani on Kyushu Island,

 Japan. Chest 2001; 120: 514-20.65. Lee EG, Na BK, Bae YA, Kim SH, Je EY, Ju JW, et al.

Identification of immunodominant excretory-secretorycysteine proteases of adult Paragonimus westermani by

proteome analysis. Proteomics 2006; 6: 1290-300.66. Na BK, Kim SH, Lee EG, Kim TS, Bae YA, Kang I, et al.

Critical roles for excretory-secretory cysteine proteasesduring tissue invasion of Paragonimus westermani newlyexcysted metacercariae. Cell Microbiol 2006; 8: 1034-46.

67. Wongkham C, Intapan PM, Maleewong W, Miwa M.Evaluation of human IgG subclass antibodies in theserodiagnosis of Paragonimus heterotremus. Asian Pac J Allergy

Immunol 2005; 23: 205-11.68. Velez ID, Ortega JE, Velasquez LE. Paragonimiasis: a reviewfrom Columbia. Clin Chest Med 2002; 23: 421-31.

69. Keiser J, Engels D, Buscher G, Utzinger J. Triclabendazole forthe treatment of fascioliasis and paragonimiasis. Expert OpinInvestig Drugs 2005; 14: 1513-26.

70. Crompton DWT. How much human helminthiasis is there inthe world? J Parasitol 1999; 85: 379-403.

71. Peng W, Zhou X, Gasser RB. Ascaris egg profiles in humanfaeces: biological and epidemiological implications.Parasitology 2003; 127: 283-90.

72. Yazicioglu M, Ones U, Yalcin I. Peripheral and nasaleosinophilia and serum total immunoglobulin E levels inchildren with ascariasis. Turk J Pediatric 1996; 38: 477-84.

73. Santra A, Bhattacharya T, Chowdhury A, Ghosh A, GhoshN, Chatterjee BP, et al. Serodiagnosis of ascariasis with

specific IgG4 antibody and its use in epidemiological study.Trans R Soc Trop Med Hyg 2001; 95: 289-292.74. Ford RM. Transient pulmonary eosinophilia and asthma: a

review of 20 cases occurring in 5702 asthma sufferers. AmRev Respir Dis 1996; 93: 797-803.

75. Liu LX, Weller PF. Strongyloidiasis and other intestinalnematode infections. Infect Dis Clin North Am 1993; 7: 655-82.

76. Citro LA, Gordon ME, Miller WT. Eosinophilic lung disease(or how to slice PIE). Am J Roentgenol Rad Ther Nuc Med 1973;117: 787-97.

77. Rexroth G, Keller C. Chronic course of eosinophilicpneumonia in infection with Ascaris lumbricoides .Pneumologie 1995; 49: 77-83.

78. Sarinas PS, Chitkara RK. Ascariasis and hookworm. SeminRespir Infect 1997; 12: 130-7.

79. Bhattacharya T, Santra A, Mazumdar DN, Chatterjee BP.

Possible approach for serodiagnosis of ascariasis byevaluation of immunoglobulin G4 response using Ascarislumbricoides somatic antigen. J Clin Microbiol 2001; 39: 2991-4.

80. St Georgiev V. Pharmacotherapy of ascariasis. Expert OpinPharmacother 2001; 2: 223-39.

81. Belizaro VY, Amarillo ME, de Leon WU, de los Reyes AE,Bugayong MG, Macatangay BJ. A comparison of the efficacyof single dose of albendazole, ivermectin and diethyl-carbamazine alone or in combination against Ascaris andTrichuris spp. Bull World Health Organ 2003; 81: 35-42.

82. Roche M, Layrise M. Nature and causes of hookwormanemia. Am J Trop Med Hyg 1996; 15: 1031-1102.

83. Hoagland KE, Schad GA. Necator americanus  andAncylostoma duodenale: life history parameters andepidemiological implications of two sympatric hookworms

on humans. Exp Parasitol 1978; 44: 36-49.84. Hotez P. Human hookworm infection. In: Farthing MJG,

Keusch GT, Wakelin D, editors Intestinal Helminths. London:Chapman and Hall; 1995: pp 129-50.

85. Nawalinski TA, Schad GA. Arrested development inAncylostoma duodenale: cause of self-induced infections inman. Am J Trop Med Hyg  1974; 23: 895-8.

86. Yu Sen-Hai, Jiang Ze-xiao, Xu Long-Qi. Infantile hookwormdisease in China: a review. Acta Trop 1995; 59: 265-70.

87. Cappello M, Clyne LP, MacPhedram P, Hotez PJ.Ancylostoma factor Xa inhibitor: partial purification and itsidentification as a major hookworm-derived anticoagulant in

Parasitic Lung Diseases V.K. Vijayan

Page 15: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 15/18

2008; Vol. 50 The Indian Journal of Chest Diseases & Allied Sciences 63

vitro. J Infect Dis 1993; 167: 1474-7.88. Howdon JM. Differentiation between the human hookworm

Ancylostoma duodenale and Necator americanus using PCR-RFLP. J Parasitol 1996; 82: 642-7.

89. Genta RM. Global prevalence of strongyloidiasis: criticalreview with epidemiologic insights into the prevention of disseminated disease. Rev Infect Dis 1989; 5: 755-67.

90. Scowden EB, Schaffner W, Stone WJ. Overwhelming

strongyloidiasis: an unappreciated opportunistic infection. Medicine (Baltimore) 1978; 57: 527-44.91. Neva FA. Biology and immunology of human

strongyloidiasis. J Infect Dis 1986; 153: 397-406.92. Casati A, Cornero G, Muttini S, Tresoldi M, Gallioli G, Torri

G. Hyperacute pneumonitis in a patient with overwhelmingStrongyloides stercoralis  infection. Eur J Anesthesiol 1996; 13:498-501.

93. Genta RM, Miles P, Fields K. Opportunistic Strongyloidesstercoralis infection in lymphoma patients: report of a caseand review of the literature. Cancer 1989; 63: 1407-11.

94. Lessman KD, Can S, Talavera W. Disseminated Strongyloidesstercoralis in human immunodeficiency virus-infectedpatients: treatment failure and review of literature. Chest1993; 104: 119-22.

95. Dionisio D, Manneschi LI, di Lollo S, Orsi A, Tani A, Papucci

A, et al. Strongyloides stercoralis: ultra structural study of newly hatched larvae within human duodenal mucosa. J Clin Pathol 2000; 53: 110-6.

96. Sanchez P, Guzman AP, Guillen SM, Adell RI, Estruch AM,Gonzalo IN, et al. Endemic strongyloidiasis on the SpanishMediterranean coast. Q J Med 2001; 94: 357-68.

97. Wehner JH, Kirsch CM. Pulmonary manifestations of strongyloidiasis. Semin Respir Infect 1997; 12: 122-9.

98. Alzer PD, Milder JE, Banwell JG, Kilgore G, Klein M, ParkerR. Epidemiologic features of Strongyloides stercoralis infectionin an endemic area of United States. Am J Trop Med Hyg 1982;31: 313-9.

99. Davidson RA, Fletcher RH, Chapman LE. Risk factors forstrongyloidiasis: a case-control study. Arch Intern Med 1984;144: 321-4.

100. Woodring JH, Halfhill H 2nd, Berger R, Reed JC, Moser N.

Clinical and imaging features of pulmonary strongyloidiasis.South Med J  1996; 89: 10-19.101. Milder JE, Walzer PD, Kilgore G, Rutherford I, Klein M.

Clinical features of Strongyloides stercoralis infection in anendemic area of the United States. Gastroenterology 1981; 80:1481-8.

102. Nwokolo C, Imohiosen E. Strongyloidiasis of respiratorytract presenting as “asthma”. Br Med J 1973; 2: 153-4.

103. Woodring JH, Halfhill H 2nd, Reed JC. Pulmonarystrongyloidiasis: clinical and imaging features. Am J Roentgenol 1994; 162: 537-42.

104. Ghoshal UC, Ghoshal U, Jain M, Kumar A, Aggarwal R,Misra A, et al. Strongyloides stercoralis infestation associatedwith septicemia due to intestinal transmural migration of 

 bacteria. J Gastroenterol Hepatol 2002; 17: 1331-3.105. Goyal SB. Intestinal strongyloidiasis manifesting as

eosinophilic pleural effusion. South Med J  1998; 91: 768-9.106. Premanand R, Prasad GV, Mohan A, Gururajkumar A,

Reddy MK. Eosinophilic pleural effusion and presence of filariform larva of Strongyloides stercoralis in a patient withmetastatic squamous cell carcinoma deposits in the pleura.Indian J Chest Dis Allied Sci 2003; 45: 121-4.

107. da Silva OA, Amarol CF, da Silveira JC, Lopez M, Pittella JE.Hypokalemic respiratory muscle paralysis followingStrongyloides stercoralis hyperinfection: a case report. Am J Trop Med Hyg 1981; 30: 69-73.

108. Lin AL, Kessimian N, Benditt JO. Restrictive pulmonarydisease due to interlobar septal fibrosis associated with

disseminated infection by Strongyloides stercoralis. Am J RespirCrit Care Med 1995; 151: 205-09.

109. Reddy TS. Syndrome of inappropriate secretion of antidiuretic hormone and nonpalpable purpura in a womanwith Strongyloides stercoralis. Am J Med Sci 2003; 325: 288-91.

110. Caceres MA, Genta RM. Pulmonary microcalcification asso-ciated with Strongyloides stercoralis. Chest 1988; 94: 862-5.

111. Sen P, Gil C, Estrellas B, Middleton JR. Corticosteroid –

induced asthma: a manifestation of limited hyperinfectionsyndrome due to Strongyloides stercoralis. South Med J 1995;88: 923-7.

112. Ossorio MA, Brovon PE, Fields CL, Roy TM. Exacerbationof chronic obstructive pulmonary disease due tohyperinfection with Strongyloides stercoralis. J Ky Med Assoc1990; 88: 233-7.

113. Marriota S, Pallone G, Li Bianchi E, Gilardi G, Bisetti A.Strongyloides stercoralis hyperinfection in a case of idiopathicpulmonary fibrosis. Panminerva Med 1996; 38: 45-7.

114. Siddiqui AA, Berk SL. Diagnosis of Strongyloides stercoralisinfection. Clin Infect Dis 2001; 33: 1040-7.

115. Koga K, Kasuya S, Khamboonruang C, Sukavat K,Nakamura Y, Tani S , et al. An evaluation of the agar platemethod for the detection of Strogyloides stercoralis in northernThailand. J Trop Med Hyg 1990; 93: 183-8.

116. Hernadez-Chavarria F, Avendano L. A simple modificationof the Baermann method for diagnosis of strongyloidiasis. Mem Inst Oswaldo Cruz (Rio de Janeiro) 2001; 96: 805-7.

117. De Kaminsky RG. Evaluation of three methods forlaboratory diagnosis of Strogyloides stercoralis  infection. J Parasitol 1993; 79: 277-80.

118. Chu E, Whitlock WL, Dietrich RA. Pulmonary hyper-infection syndrome with Strogyloides stercoralis. Chest 1990;97: 1475-7.

119. Williams J, Ninley D, Dralle W, Berk SL, Verghese A.Diagnosis of pulmonary strongyloidiasis by bronchoalveolarlavage. Chest 1988; 94: 643-4.

120. Loutfy MR, Wilson M, Keystone JS, Kain KC. Serology andeosinophil count in the diagnosis and management of strongyloidiasis in a non-endemic area. Am J Trop Med Hyg2002; 66: 749-52.

121. Datry A, Hilmarsdoltir I, Mayorga-Sagastume R, LyaguobiM, Gaxotte P, Biligui S, et al. Treatment of Strongyloidesstercoralis  infection with ivermectin compared withalbendazole: results of an open study of 60 cases. Trans R SocTrop Med Hyg 1994; 88: 344-5.

122. Pitisuttihum P, Supanaranoud W, Chindanoud A. Arandomized comparative study of albendazole in chronicstrongyloidiasis.South East Asian J Trop Med Pub Health 1995;26: 735-8.

123. Vijayan VK. Immunopathogenesis and treatment of eosinophilic lung diseases in the tropics. In: Sharma OP,editor Tropical Lung Disease (Lung Biology in Health andDisease). New York: Marcel Dekker Inc., 2006; 211: 195-239.

124. Udwadia FE. Tropical eosinophilia. In: Herzog H, editorPulmonary Eosinophilia: Progress in Respiration Research. Basel:S. Karger; 1975: pp 35-155.

125. Ottesen EA, Nutman TB. Tropical pulmonary eosinophilia.Ann Rev Med 1992; 43: 417-24.

126. Vijayan VK. Tropical pulmonary eosinophilia. Curr OpinPulm Med 2007; 13: 428-33.

127. Ottesen EA. Immunological aspects of lymphatic filariasisand onchocerciasis. Trans R Soc Trop Med Hyg 1984; 73(suppl.): 9-18.

128. Frimodt-Moller C, Barton RM. A pseudo-tuberculosiscondition associated with eosinophilia. Indian Med Gaz 1940;75: 607-13.

129. Weingarten RJ. Tropical eosinophilia. Lancet 1943; 1: 103-5.130. Stuiver PC, Wismans PJ, Schornagel R. Tropical eosinophilia

Page 16: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 16/18

64

is an important disease in the Netherlands. Ned TijdschrGeneeskd 1991; 135: 283-6.

131. Hayashi K, Horiba M, Shindou J, Sumida T, Takekoshi A.Tropical eosinophilia in a man from Sri Lanka. Nihon KyobuShikkan Gakkai Zasshi 1996; 34: 1411-5.

132. World Health Organization, Division of Control of TropicalDiseases. Lymphatic filariasis infection and disease: controlstrategies, 1994 (TDR/CTD/FIL/Penang); pp 1-30.

133. Johnson S, Wilkinson R, Davidson RN. Tropical respiratorymedicine. IV: Acute tropical infection and the lung. Thorax1994; 49: 714-8.

134. Jiva TM, Israel RH, Poe RH. Tropical pulmonary eosinophiliamasquerading as acute bronchial asthma. Respiration 1996;63: 55-8.

135. Danaraj TJ, Schacher JF. I/D test with Dirofilaria immitisextract in eosinophilic lung (Tropical eosinophilia). Am J Trop

 Med Hyg 1959; 8: 640-3.136. Webb JKB, Job OK, Gault EW. Tropical eosinophilia:

demonstration of microfilariae in lung, liver and lymphnodes. Lancet 1960; 1: 835-42.

137. Joshi VV, Udwadia FE, Gadgil RK. Etiology of tropicaleosinophilia: a study of lung biopsies and review of published reports. Am J Trop Med Hyg 1969; 18: 231-40.

138. Danaraj TJ, Pachecco G, Shanmugaratnam K, Beaver PC.

The etiology and pathology of eosinophilic lung (Tropicaleosinophilia). Am J Trop Med Hyg 1966; 15: 183-9.139. Buckley JJC. Tropical pulmonary eosinophilia in relation to

filarial infections (Wuchereria sp.) of animals. Trans R Soc Trop Med Hyg 1958; 52: 335-6.

140. Ganatra RD, Lewis RA. Diethylcarbamazine (Hetrazan) intropical eosinophilia. India J Med Sci 1955; 9: 672-81.

141. Baker SJ, Rajan KJ, Davadutta S. Treatment of tropicaleosinophilia: a controlled trial. Lancet 1959; 2: 144-7.

142. Ottesen EA, Neva FA, Paranjape RS, Tripathy SP,Thiruvengadam KV, Beaver MA. Specific allergicsensitization to filarial antigens in tropical pulmonaryeosinophilia. Lancet 1979; 1: 1158-61.

143. Viswanathan R, Bagai RC, Saran R. Leukocyte adhesionphenomenon in pulmonary eosinophilia (Tropicaleosinophilia). Am Rev Respir Dis 1973; 107: 298-300.

144. Perera CS, Perera LM, de Silva C, Abeywickreme W,Dissanaike AS, Ismail MM. An eosinophilic granulomacontaining an adult female Wuchereria bancrofti in a patientwith tropical pulmonary eosinophilia. Trans R Soc Trop MedHyg 1992; 86: 542.

145. Dreyer G, Noroes J, Rocha A, Addiss D. Detection of livingadult Wuchereria bancrofti  in a patient with tropicalpulmonary eosinophilia. Braz J Med Biol Res 1996; 29: 1005-8.

146. Viswanathan R. Post mortem appearances in tropicaleosinophilia. Indian Med Gaz 1947; 82: 49-50.

147. Udwadia FE, Joshi VV. A study of tropical eosinophilia.Thorax 1964; 19: 548-54.

148. Pinkston P, Vijayan VK, Nutman TB, Rom WN, O’DonnellKM, Cornelius MJ, et al.  Acute tropical pulmonaryeosinophilia: characterization of the lower respiratory tract

inflammation and its response to therapy. J Clin Invest 1987;80: 216-25.

149. Gliech GJ, Loegering DA. Immunobiology of eosinophils.Ann Rev Immunol 1984; 2: 429-59.

150. O’Bryan L, Pinkston P, Kumaraswami V, Vijayan VK,Yenokida G, Rosenberg HF, et al. Localized eosinophildegranulation mediates disease in tropical pulmonaryeosinophilia. Infect Immun 2003; 71: 1337-42.

151. Nutman TB, Vijayan VK, Pinkston P, Steel R, Crystal RG,Ottesen EA. Tropical pulmonary eosinophilia: analysis of antifilarial antibody localized to the lung. J Infect Dis  1989;160: 1042-50.

152. Lobos E, Ondo A, Ottesen EA, Nutman TB. Biochemical andimmunologic characterization of a major IgE-inducingfilarial antigen of Brugia malayi and implications for thepathogenesis of tropical pulmonary eosinophilia. J Immunol1992; 149: 3029-34.

153. Lobos E, Zahn R, Weiss N, Nutman TB. A major allergen of lymphatic filarial nematodes is a parasite homolog of thegamma-glutaryl transpeptidase. Mol Med 1996; 2: 712-24.

154. Lobos E, Nutman TB, Hothersall JS, Moncada S. Elevatedimmunoglobulin E against recombinant Brugia malayigamma-glutaryl transpeptidase in patients with bancroftianfilariasis: association with tropical pulmonary eosinophiliaor putative immunity. Infect Immun 2003; 71: 747-53.

155. Freedman DO, Nutman TB, Ottesen EA. Protectiveimmunity in bancroftian filariasis: selective recognition of a43-KD larval stage antigen by infection-free individuals in anendemic area. J Clin Invest 1989; 83: 14-22.

156. Gounni AS, Spanel-Borowski K, Palacios M, Heusser C,Moncada S, Lobos E. Pulmonary inflammation induced by arecombinant Brugia malayi gamma-glutaryl transpeptidasehomolog: involvement of human autoimmune process. Mol

 Med 2001; 7: 344-54.157. Ravindran M. Tropical eosinophilia presenting with

neurological features. Br Med J  1979; 2: 1262.

158. Cooray JH, Ismail MM. Re-examination of the diagnosticcriteria of tropical pulmonary eosinophilia. Respir Med 1999;93: 655-9.

159. Kamat SR, Pimparkar SD, Store SD, Warrier NVU, Fakey YC.Study of clinical, radiological and pulmonary functionpatterns and response to treatment in pulmonaryeosinophilia. Indian J Chest Dis 1970; 12: 91-100.

160. Basu SP. X-ray appearance in lung fields in tropicaleosinophilia. Indian Med Gaz 1954; 89: 212-7.

161. Islam N. Radiological features of tropical eosinophilia.  J  Trop Med Hyg 1965; 68: 117-80.

162. Khoo FY, Danaraj TJ. The roentgenographic appearances of eosinophilic lung. Am J Roentgenol 1960; 86: 251-9.

163. Vijayan VK Tropical eosinophilia: bronchoalveolar lavageand pulmonary pathophysiology in relation to treatment.Ph.D. Dissertation, University of Madras, Madras, India,

1988.164. Vijayan VK, Kuppurao KV, Sankaran K, Venkatesan P,

Prabhakar R. Tropical eosinophilia: clinical andphysiological response to diethylcarbamazine. Respir Med1991; 85: 17-20.

165. Sandhu M, Mukhopadhyay S, Sharma SK. Tropicalpulmonary eosinophilia: a comparative evaluation of plainchest radiography and computed tomography. AustralasRadiol 1996; 40: 32-7.

166. Ray D. Lung function in tropical eosinophilia. Indian J ChestDis 1974; 16: 368-73.

167. Poh SC. The course of lung function in treated tropicaleosinophilia. Thorax 1974; 29: 710-2.

168. Vijayan VK, Kuppurao KV, Sankaran K, Venkatesan P,Prabhakar R. Diffusing capacity in acute untreated tropicaleosinophilia. Indian J Chest Dis Allied Sci 1988; 30: 71-7.

169. Vijayan VK, Kuppurao KV, Sankaran K, Venkatesan P,Prabhakar R. Pulmonary membrane diffusing capacity andcapillary blood volume in tropical eosinophilia. Chest 1990;97: 1386-9.

170. Kuppurao KV, Vijayan VK, Venkatesan P, Sankaran K. Effectof treatment on maximal expiratory flow rates in tropicaleosinophilia. Ceylon Med J  1993; 38: 78-80.

171. Vijayan VK, Sankaran K, Venkatesan P, Kuppurao KV.Correlation of lower respiratory tract inflammation withchanges in lung function and chest roentgenograms inpatients with untreated tropical pulmonary eosinophilia.Singapore Med J  1991; 32: 122-5.

Parasitic Lung Diseases V.K. Vijayan

Page 17: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 17/18

2008; Vol. 50 The Indian Journal of Chest Diseases & Allied Sciences 65

172. Vijayan VK, Kuppurao KV, Venkatesan P, Prabhakar R.Arterial hypoxemia in acute tropical pulmonaryeosinophilia. Lung India 1988; 6: 183-5.

173. Ray D. Relapsing tropical eosinophilia. Indian J Chest DisAllied Sci 1977; 19: 65-71.

174. Ray D. Arterial desaturation in tropical eosinophilia. Indian J Chest Dis Allied Sci 1984; 26: 34-7.

175. Ray D, Jayachandran CA. Ventilation-perfusion scinti-

scanning in tropical pulmonary eosinophilia. Chest 1993; 104:497-500.176. Vakil RJ. Cardiovascular involvement in tropical

eosinophilia. Br Heart J  1961; 23: 578-86.177. Johny KV, Ananthachari MD. Cardiovascular changes in

tropical eosinophilia. Am Heart J  1965; 69: 591-8.178. Singh M, Sharma SK, Patney RM. Tropical eosinophilic

pericarditis. Indian Heart J  1974; 26: 261-3.179. D’Arbera VSE. Tropical eosinophilia: some atypical cases of 

clinical interest. Med J Malaya 1958; 12: 559-62.180. Quah BS, Anuar AK, Rowani MR, Pennie RA. Cor

pulmonale: an unusual presentation of tropical eosinophilia.Ann Trop Pediatr 1997; 17: 17-8.

181. Kedarnath, Agarwal SN. An unusual case of tropicaleosinophilia. J Trop Med Hyg 1963; 66: 175-7.

182. Spry CJF. Lung diseases associated with eosinophils. In: Kay

AB, Goetzl EJ, editors Current Perspective in Allergy.Edinburgh: Churchill Livingstone; 1982: pp 67-77.183. Rocha A, Dreyer G, Poindexter RW, Ottesen EA. Syndrome

resembling tropical eosinophilia but of non-filarial etiology:serological findings with filarial antigens. Trans R Soc Trop

 Med Hyg 1995; 89: 573-5.184. Alves LC, Brayner FA, Silva LF, Pimentel RC, Rocha A,

Peixoto CA. Immunocytochemical localization of antigensrecognized by tropical pulmonary eosinophilia andindividuals with intestinal helminth antisera in microfilaria of Wuchereria bancrofti. J Submicrose Cytol Pathol 2002; 34: 211-6.

185. Vijayan VK. Allergic bronchopulmonary helminthiasis(ABPH). Lung India 1991; 9: 145-8.

186. Vijayan VK, Reetha AM, Jawahar MS, Sankaran K,Prabhakar R. Pulmonary eosinophilia in pulmonarytuberculosis. Chest 1992; 101: 1708-9.

187. Danaraj TJ. The treatment of eosinophilic lung (Tropicaleosinophilia) with diethylcarbamazine. Quart J Med 1958; 27:243-63.

188. World Health Organization. Final report: Joint WPRO/SEARO Working Group on Brugian Filariasis. Manila: WHO;1979: pp 1-47.

189. Vijayan VK, Sankaran K, Venkatesan P, Prabhakar R. Effectof diethylcarbamazine on the alveolitis of tropicaleosinophilia. Respiration 1991; 58: 255-9.

190. Rom WN, Vijayan VK, Cornelius MJ, Kumaraswami V,Prabhakar R, Ottesen EA, et al. Persistent lower respiratorytract inflammation associated with interstitial lung disease inpatients with tropical pulmonary eosinophilia followingtreatment with diethylcarbamazine. Am Rev Respir Dis 1990;142: 1088-92.

191. Vijayan VK. Role of steroids in chronic tropical eosinophilia:

a bronchoalveolar lavage study (abstract). Indian J Chest DisAllied Sci 1987; 29: vii.

192. Vijayan VK. Changes in respiratory function and lunginflammation in patients with tropical eosinophilia one yearafter treatment with diethylcarbamazine. Indian J ClinPharmacol Therap 1997; 15: 5-7.

193. Sanjivi KS, Thiruvengadam KV, Friedman HC. Tropicaleosinophilia treated with cortisone. Dis Chest 1955; 28: 88-90.

194. Ottesen EA, Vijayasekaran V, Kumaraswami V, PerumalPillai SV, Sadanandam A, Frederik S, et al. A controlled trialof ivermectin and diethylcarbamazine in lymphatic filariasis.N Engl J Med 1990; 322: 1113-7.

195. Vijayan VK. Ivermectin in the treatment of tropicaleosinophilia (Abstract). Chest 2000; 118(Suppl. 4): 247s.

196. Theis JH. Public health aspects of dirofilariasis in the UnitedStates. Vet Parasitol 2005; 133: 157-80.

197. Bronshstein AM, Supriaga VG, Stavrovski BI, et al. Humandirofilariasis in the Moscow region.  Med Parazitol (Mosk)2003; 3: 51-6.

198. Miyoshi T, Tsubouchi H, Iwasaki A, Shiraishi T, Nabeshima

K, Shirakusa T. Human pulmonary dirofilariasis: a casereport and review of the recent Japanese literature.Respirology 2006; 11: 343-7.

199. Oshiro Y, Murayama S, Sunagawa U, Nakamoto A, Owan I,Kuba M, et al.  Pulmonary dirofilariasis: computedtomographic findings and correlation with pathologicfeatures. J Comput Assist Tomogr 2004; 28: 796-800.

200. Moore W, Franceschi D. PET findings in pulmonarydirofilariasis. J Thorac Imaging 2005; 20: 305-6.

201. Rivasi F, Boldorini R, Criante P, et al. Detection of Dirofilaria(Nochtiella) repens DNA by polymerase chain reaction inembedded paraffin tissues from two human pulmonarylocations. APMIS 2006; 114: 566-73.

202. Beaver PC, Syncler CH, Carrera GM. Chronic eosinophiliadue to visceral larva migrans. Pediatrics 1952; 9: 7-19.

203. Magnaval JF, Glickman LT, Dorchies P, Morassin B. Highlights

of human toxocariasis. Korean J Parasitol 2001; 39: 1-11.204. Glickman LT, Schantz PM. Epidemiology and pathogenesisof zoonotic toxocariasis. Epidemiol Rev 1981; 3: 230-50.

205. Thompson DE, Bundy DAP, Cooper ES, Schantz PM.Epidemiological characteristics of Toxocara canis infection of children in a Caribbean community. Bull World Health Organ1986; 64:283-90.

206. Del Prete GF, De Carli M, Mastromauro C, Biagiotti R,Macchia D, Falagiani P, et al. Purified protein derivative of 

 Mycobacterium tuberculosis and excretory-secretary antigen(s)of Toxocara canis expand in vitro human T cells with stableand opposite (type 1 T helper or type 2 T helper) profile of cytokine production. J Clin Invest 1991; 88:346-50.

207. Blaxter ML, Page AP, Rudin W, Maizels RM. Nematodesurface coats: actively evading immunity. Parasitol Today1992; 8: 243-7.

208. Badley JE, Grieve RB, Rockey JH, Glickman LT. Immuno-mediated adherence of eosinophils to Toxocara canis infectivelarvae: the role of excretory-secretary antigens. ParasiteImmunol 1987; 9: 133-43.

209. Windelborg NB, Lind P, Hansen B, Nansen P, Schiotz PO.Larval exoantigens from ascarid nematodes are potentinducers of histamine release from human blood basophils.Clin Exp Allergy 1991; 21: 725-32.

210. Smith HV, Kusel JR, Girdwood RW. The productions byhuman A and B blood group like substances by in vitromaintained second stage Toxocara canis larvae; their presenceon the outer larval surfaces and in their excretions/secretions. Clin Exp Immunol 1983; 54: 625-33.

211. Feldman GJ, Parker HW. Visceral larva migrans associatedwith the hypereosinophilic syndrome and the onset of severeasthma. Ann Intern Med 1992; 116: 838-40.

212. Roig J, Romeu J, Riera C, Texido A, Domingo C, Morera J.Acute eosinophilic pneumonia due to toxocariasis with

 bronchoalveolar lavage findings. Chest 1992; 102: 294-6.213. Bartelink AK, Kortbeek LM, Huidekoper HJ, Meulenubelt J,

Van Knapen F. Acute respiratory failure due to toxocarainfections. Lancet 1993; 342: 1234.

214. Glickman LT, Magnaval JF, Domanski LM, Shofer FS, LauriaSS, Gottstein B, et al. Visceral larva migrans in French adults:a new disease syndrome? Am J Epidemiol 1987; 125:1019-34.

215. Nathwani D, Laing RB, Currie PF. “Covert” toxocariasis: acause of recurrent abdominal pain in childhood. Br J ClinPract 1992; 46: 271.

Page 18: iaet08i1p49

7/21/2019 iaet08i1p49

http://slidepdf.com/reader/full/iaet08i1p49 18/18

66

216. Spieser F, Gottstein B. A collaborative study on larvalexcretory/secretary antigens of Toxocara canis  forimmunodiagnosis of human toxocariasis with ELISA. ActaTrop 1984; 41: 361-72.

217. Magnaval JF, Fabre R, Maurieres P, Charlet JP, De Larrard B.Evaluation of an immunenzymatic assay detecting specificanti-Toxocara immunoglobulin E for diagnosis and posttreatment follow up of human toxocariasis.  J Clin Microbiol1992; 30:2269-74.

218. Magnaval JF, Fabre R, Maurieres P, Charlet JP, De Larrard B.Application of the Western-blotting procedure for theimmunodiagnosis of human toxocariasis. Parasitol Res 1991;77: 697-702.

219. Rasmussen LN, Dirdal M, Birkeback NH. “CovertToxocariasis” in a child treated with low-dosediethylcarbamazine. Acta Paediatr 1993; 82: 116-8.

220. Magnaval JF. Comparative efficacy of diethylcarbamazineand mebendazole for the treatment of human toxocariasis.Parasitology 1995; 110: 529-33.

221. Sturchler D, Schubarth P, Gualzata M, Gottstein B, Orettli A.Thiabendazole vs albendazole in treatment of toxocariasis: aclinical trial. Ann Trop Med Parasitol 1989; 83: 473-8.

222. Pozio E, La Rosa G, Murrell KD, Lichtenfels JR. Taxonomicrevision of the genus Trichinella.  J Parasitol 1992; 78: 654-9.

223. Despommier DD. How does Trichinella spiralis make itself athome? Parasitol Today 1998; 14: 318-23.

224. Dessein AJ, Parker WL, James SL, David JR. IgE antibodyand resistance to infection. I. Selective suppression of the IgEantibody response in rats diminishes the resistance and theeosinophil response in Trichinella infections. J Exp Med 1981;

153: 423-36.225. Capo V, Despommier DD. Clinical aspects of infections with

Trichinella spp. Clin Microbiol Rev 1996; 9: 47-54.226. Sheldon JH. An outbreak of trichinosis near Wolverhampton.

Lancet 1941; 1: 203-5.227. Bruschi F, Murrell K. Trichinellosis. In: Guerrant RL, Walker

DH, Weller PF, editors Tropical Infectious Diseases: Principles,Pathogens and Practice; Vol. II. (Elsevier Science HealthScience Divn). Philadelphia: Churchill Livingstone; 1999: pp917-25.

228. Engvall E, Ljungstrom I. Detection of human antibodies toTrichinella spiralis by enzyme-linked immunosorbent assay(ELISA). Acta Pathol Microbiol Scand 1975; 83: 231-7.

Parasitic Lung Diseases V.K. Vijayan