Вы находитесь на странице: 1из 8

Eur Respir J 2001; 18: Suppl.

32, 93s100s Printed in UK all rights reserved

Copyright #ERS Journals Ltd 2001 European Respiratory Journal ISSN 0904-1850 ISBN 1-904097-01-4

REVIEW

Drug-induced inltrative lung disease


Ph. Camus, P. Foucher, Ph. Bonniaud, K. Ask

Drug-induced inltrative lung disease. Ph. Camus, P. Foucher, Ph. Bonniaud, K. Ask. #ERS Journals Ltd 2001. ABSTRACT: An increasing number of drugs are recognized to induce distinctive patterns of inltrative lung disease (ILD), ranging from benign inltrates to lifethreatening adult respiratory distress syndromes. In addition to drugs, biomolecules such as proteins and cytokines, and medicinal plants are also capable of inducing respiratory disease, some being severe and/or irreversible. For several reasons it is difcult to estimate the exact frequency of drug-induced inltrative lung disease (DI-ILD). The risk for DI-ILD and the clinical patterns vary depending on a variety of host and drug factors. Although establishing the diagnosis is often difcult, systematic evaluation of the possible role of almost any kind of drugs in ILD is warranted, as stopping a drug may favourably inuence prognosis. However, prognosis depends on the drug and the type of DI-ILD. Corticosteroids may suppress the inammatory reaction, but for many drugs, proof of the effect of corticosteroids is lacking. Advances in prevention and prediction are needed. A user-friendly database of respiratory adverse drug reactions was made available on the web, to provide quick information in this area. Eur Respir J 2001; 18: Suppl. 32, 93s100s.

Service de Pneumologie et de Reanima tion Respiratoire, Centre Hospitalier Universitaire de Dijon, et Unite de Pharmacologie Toxicologie Pulmonaire, Universite de Bourgogne, Dijon, France. Correspondence: Ph. Camus, Service de Pneumologie et de Reanimation Respiratoire, CHU de Dijon, Dijon, France. Fax: 33 380293251 Keywords: Drug-induced lung disease drugs lung toxicity patients registry web technology

In the relatively recent past, conventional (i.e. nonillicit) drugs (as opposed to medical or surgical procedures) have emerged as relatively common and signicant inducers of diffuse inltrative lung disease (ILD) [15], as well as of pleural and pulmonary vascular disease [57]. Here, the term "inltrative" will be preferred to "interstitial", as drugs can induce alveolar lling processes, in addition to affecting the interstitium [810]. Drugs as aetiological agents of ILD are interesting for two main reasons: 1) they offer a coherent explanation to a sizeable number of ILD cases seen in clinical practice [11]; and 2) early withdrawal of the causative drug will often lead to improvement or even cure of the ILD. In addition to the utility of a catalogue of causative drugs and clinical pictures, at present [5, 11], it is necessary to look ahead, and to ask further questions in order to prepare the future of iatrogenic lung disorders. It is beyond the scope of the present paper to review all drug-induced respiratory diseases [1113], but rather to point out novel aspects, and raise questions that need to be addressed. For a more complete overview of drug-induced respiratory diseases, refer to the Pneumotox1 website [11] and to comprehensive review articles [13, 14]. Data Processing From 1950 onwards, a considerable amount of information on drug-induced (DI)-ILD has been

gathered [15], and made available to the clinician, as regards which drugs can cause adverse effects in the lung, and what pattern of lung reaction can be expected with the use of a given drug [14, 14, 1618]. During that period of time, accrual of information has been almost relentless [15]. In addition, novel patterns of respiratory involvement and newer offending compounds have been described, as new drugs or combinations were made available to the clinician, and postmarketing experience on drugs expanded. In the past, information on DI-ILD was essentially available in the form of conventional articles [15]. A supplemental source of information was recently provided, in the form of the free and continuously updated Pneumotox1 website [11]. The site was designed and constructed to provide the interrogator with quick and easy access to formatted data, and relevant literature on drug-induced respiratory disease. At the time of writing, the site9s database contained 4,320 references on DI-ILD available on-line. An overview of this accumulated literature shows marked heterogeneity in reports. This illustrates the need to improve existing diagnostic criteria [19] and to provides publication guidelines for all reports on DI-ILD in the future. In addition to drugs, biomolecules can also induce ILD, and include the following. 1) Interferons (used in the treatment of chronic myelogenous leukemia and of hepatitis C), which have been linked to the

94s

PH. CAMUS ET AL.

development of cutaneous or thoracic sarcoidosis [2023] or bronchiolitis obliterans with organizing pneumonia (BOOP) [24]; this is of interest to the general understanding of the pathogenesis of these diseases. 2) Immunoglobulins and anti-thymocyte globulin, which may induce pulmonary inltrates or pulmonary oedema [25, 26]. 3) Substances which modulate the growth, release or maturation of blood cells or progenitors, which have been shown to induce severe ILD, and even the acute respiratory distress syndrome (ARDS); examples include all-trans-retinoic acid (ATRA) [27, 28], used in the management of promyelocytic leukemia, and granulocyte (G)- or granulocyte monocyte (GM)-colony stimulating factor (CSF) [29, 30], used to restore white blood cell counts in patients with bone marrow hypoplasia, mainly of iatrogenic origin. 4) Transfusions, which can induce the transfusion-related lung injury (TRALI) syndrome, which consists of the rapid development of pulmonary inltrates or pulmonary oedema, following transfusion of blood or blood products containing antileukocyte or antihuman leukocyte antigen (antiHLA) antibodies of donor origin [31, 32]. 5) A novel pattern of pulmonary complications has been described following stem cell transplantation, in the form of pulmonary cytolytic thrombi originating from donor cells [33]. Thus, whereas DI-ILDs were formerly regarded as interstitial tissue reactions to drugs, the three latter patterns suggest that they may also result from damage to pulmonary vessels, during the transit, aggregation and/or sequestration of activated blood cells or progenitors within the pulmonary circulation. Treatments with natural products, such as herbs and other "dietary supplements", will also require close attention, as they increasingly appear capable of causing serious diseases [3437]. Well-documented examples are the association of the intake of ephedra with death from cardiovascular causes, of comfrey with hepatic veno-occlusive disease, of germander with acute hepatitis, of aristolochic acid in a chinese herb with urothelial cancer [36, 37], and of shrub leafs containing Sauropus androgynus with bronchiolitis obliterans [3842]. Demographics and epidemiology It is difcult to estimate the exact frequency of DI-ILD for several reasons, including the following. 1) The use of plain chest radiography is likely to underestimate subclinical forms of DI-ILD, compared with high-resolution computed tomography (HRCT) [43]. This has been exemplied by asymptomatic patients taking amiodarone [44], who may have opacities detectable at HRCT or at autopsy [45]. 2) Many drugs that induce ILD are used primarily by nonrespiratory physicians [4649], and underdiagnosis is possible. 3) In oncology patients, where DI-ILD is common and the differential diagnosis is broad, the ultimate diagnosis will often remain presumptive, because the severe clinical status of the patient precludes the use of invasive diagnostic techniques. The role of drugs in the pathogenesis of the "delayed

pulmonary toxicity syndrome" seen in this context remains to be delineated [5053]. 4) Estimates of DIILD frequency would require systematic notication of drug-induced adverse effects to centralized drugmonitoring agencies (as performed in several countries, including France; but a Europe-wide reporting system is needed), and estimates of the number of exposed patients. Therefore, any estimate of the frequency of DI-ILD is probably an underestimate. Risk factors A major, but largely unresolved question is why, out of the treated population of patients, only some individuals will develop DI-ILD (see the report by NEMERY et al. [54] in this Supplement). A limiting dose has only been identied for a few drugs [55]. For amiodarone and bleomycin, toxicity was once considered likely only if high doses had been administered, but several recent reports of side-effects after low doses of these drugs tend to suggest that there is no real safe dose [56, 57]. For most drugs there is no apparent role of dosage or duration of treatment in relation to the likelihood of developing adverse effects, i.e. development remains largely unexpected and idiosyncratic. Other explanations for the unequal risks among patients include the following. 1) Prior respiratory reactions to the drug, to a congener, or to unrelated compounds. Beyond occasional mentions in case reports [5862], little data are available in that area. 2) Type of underlying disease for which the drug is being given. Diseases such as rheumatoid arthritis [63] or ulcerative colitis [50], may increase the relative risk of developing respiratory disease from diseasemodifying drugs. 3) Occupational factors, such as exposure to asbestos, which may potentiate the noxious respiratory effects of ergot drugs [6466]. 4) Activation and detoxication pathways of drugs and chemicals, which differ quantitatively and qualitatively among individuals [67]; also, some ethnic groups may be at increased risk of developing adverse reactions to drugs [46, 6870]. 5) Hepatic or systemic reactions to drugs were occasionally linked to acetylator or HLA phenotype [71], but in contrast to these, very few studies have addressed the possibility of such a relation in patients with DI-ILD. The overall rarity and scattering of cases may be an explanation of why such analysis has not yet been possible. 6) The role of drugs taken concomitantly may be important, via their possible impact on cytochrome P450 systems, on detoxication pathways, or via altered pharmacokinetics of the offending drug [72]. More information is needed in this area. 7) Hazardous associations have been reported, such as the concurrent administration of several chemotherapeutic agents, of chemotherapy and radiation, and of chemotherapy (even remote) with high inspired concentrations of oxygen. Inadvertent mixing of potentially pneumotoxic treatments may lead to an unexpected pulmonary toxicity, known as recall pneumonitis [7376], which may develop at doses of each offending agent individually considered to be safe and nontoxic. 8) Bleomycin may exhibit

DRUG-INDUCED INFILTRATIVE LUNG DISEASE

95s

enhanced pulmonary toxicity in a patient with renal failure [77]. On the other hand, a slow, as opposed to a rapid infusion, may decrease the likelihood of bleomycin pulmonary toxicity [78]. Although these risks are well recognized, it is unclear that all these hazards are really taken into account and implemented sufciently in clinical practice. There are also examples of individuals who differ in their risk of developing pulmonary disease following an apparently similar pulmonary insult [79]. For instance, why only a fraction of patients with denite amiodarone pneumonitis will ultimately develop irreversible pulmonary brosis, while others will recover, remains largely unexplained. Despite identication of some risk factors, prediction of a DI-ILD remains difcult. Only for a limited number of drugs (bleomycin, nitrosoureas, amiodarone), is monitoring of patients who receive the drug advisable, but even this is debatable [80]. Clinical patterns The clinicoradiographical correlates of DI-ILD range from subclinical opacities [81] to a picture of white lungs with the criteria of ARDS [11, 82, 83]. Several drugs (including amiodarone, bleomycin, colony stimulating factors, methotrexate, mitomycin, nitrofurantoin) [11], and several histopathological patterns (acute cellular-type nonspecic interstitial pneumonia, acute eosinophilic pneumonia, acute organizing pneumonia (alternatively called BOOP), acute pulmonary oedema or alveolar haemorrhage) may lead to the clinicoradiographical and gasexchange pattern of ARDS [11]. Accordingly, pulmonary physicians should inform their colleagues in the intensive care eld, that drugs can cause lifethreatening respiratory failure and ARDS. In the patient with migratory opacities with or without chest pain that could be suggestive of organizing pneumonia, a careful drug history should also be taken, as this clinical pattern may relate to exposure to drugs, or radiation [8486]. Among the mild forms of DI-ILD, transient inltrates have been described following administration of drugs (such as hydrochlorthiazide, nitrofurantoin, novel anticancer agents) [87], proteins [26], colony stimulating factors [29], and blood products [31]. Understandably, the histopathological background of these transient and benign inltrates remains almost unknown [88], as no biopsy was taken in these cases. While in most instances, the pulmonary reaction in DI-ILD is conned to the lung, or restricted to the lung and liver [89], it may occasionally be part of a generalized and systemic syndrome [60, 9095]. Examples, include the following. 1) The drug-induced systemic lupus erythematosus syndrome [96], which may result from exposure to a wide array of drugs [11] (e.g. hydralazine, beta blockers, nonsteroidal antiinammatory drugs and many others). 2) Drug-induced hypersensitivity syndromes, with involvement of liver, brain, heart, digestive system, bone marrow, lymph nodes or any combination of these, which mostly

follows the exposure to anticonvulsants [60, 94, 95, 97, 98]. 3) Drug-induced alveolar haemorrhage with concomitant renal failure from penicillamine [99]. 4) ANCA-positive drug-induced angiitis with or without pulmonary capillaritis and haemorrhage, recently related to the use of the antithyroid drug propyl-thiouracil [100105], in addition to a few other compounds [106109]. 5) The drug-induced ChurgStrauss syndrome, occasionally described in the past following the use of aspirin [110], macrolides [111, 112], and more recently described in asthma patients on leukotriene antagonists [111114]. Children may also develop DI-ILD [115]. Chemotherapy, radiotherapy or their combination in early childhood, for instance for brain tumours or lymphoma, may lead to a pattern of progressive pulmonary retraction and brosis [116]. The pulmonary maldevelopment may be aggravated with growth, and lead to impaired lung function and loss of functional reserve, resulting in lung transplantation in adolescence, or in early adulthood. Histopathology Reports of DI-ILD in which histological data were obtained (v10% of those published to date [11]) indicate that drugs can induce many distinctive histopathological patterns of ILD (table 1). In the constellation of ILD patterns described [140142], most can be induced by drugs. Only giant-cell interstitial pneumonia and respiratory bronchiolitisinterstitial lung disease have not (yet) been related to exposure to drugs. While drugs can trigger the development of such conventional patterns of ILD as nonspecic interstitial pneumonia (cellular or brotic), eosinophilic pneumonia [59], or pulmonary brosis [143], drugs can also elicit less usual patterns such as organizing pneumonia (OP, previously designated BOOP) [86, 122, 144], or desquamative interstitial pneumonia (DIP) [124]. The two latter patterns have such a limited number of other recognized causes that the iatrogenic cause for the ILD is sometimes suspected as a result of the histological report. Other than mineral oil pneumonia [145] and, sometimes, amiodarone pneumonitis [146], the histopathological appearance of DI-ILD is almost indistinguishable from that of their counterparts occurring idiopathically, or from other causes. While some drugs (e.g. minocycline, nitrofurantoin) induce quite stereotyped reactions in the lung (eosinophilic pneumonia and the cellular type of nonspecic interstitial pneumonia, respectively) [11], other drugs (e.g. amiodarone, bleomycin), can induce a palette of variegated histopathological patterns in different patients [11]. As an example, patterns described under the term "amiodarone pneumonitis" may include nonspecic interstitial pneumonia (cellular or brotic type), alveolar lling by foamy macrophages, organizing pneumonia, diffuse alveolar damage, interstitial lung brosis, a pattern of "usual interstitial pneumonia", or any combination thereof depending on the patient and, possibly, the site of the lung biopsy [11, 146]. The reasons why some drugs

96s

PH. CAMUS ET AL.

Table 1. Types of inltrative lung disease which may be induced by drugs (pulmonary oedema was omitted) Type of inltrative lung disease Prototypic drug(s) Frequency Severity Literature Number of causative drugs [11]

NSIP-cellular Granulomatous IP Pseudo-sarcoidosis Eosinophilic pneumonia Organizing pneumonia

Methotrexate, nilutamide Common Mild [18, 46] 89 BCG, methotrexate Uncommon Mild [18, 117, 118] 8 Interferons (antiretroviral therapy?) Uncommon Mild [20, 23, 119] 3 Antibiotics, NSAID, ACE inhibitors Common Can be severe [58, 120, 121] 95 Amiodarone, bleomycin, interferons, Common Can be severe [83, 84, 23 nitrofurantoin, radiations, statins 122, 123] DIP Nitrofurantoin Rare Moderate [124] 3 LIP Phenytoin Very rare? Mild [125] 1 NSIP-brotic, or IPF-like Amiodarone, chemoptherapy, ergots Quite Severe [8, 48, 126] 34 common AIP/DAD Chemotherapy, gold, methotrexate* Quite Usually [127132] 10 and most common severe chemotherapies Alveolar lling process with Amiodarone, mineral oil Quite Variable [9, 10, 45] 2 foamy macrophage common Alveolar haemorrhage Anticoagulants, brinolytic agents, Unusual Variable [133137] 25 pulmonary haemosiderosis** penicillamine, PTU Proteinosis-like Busulfan Rare ? [138, 139] 1 NSIP: nonspecic interstitial pneumonia; IP: interstitial pneumonia; DIP: desquamative interstitial pneumonia; LIP: lymphocytic interstitial pneumonia; IPF: idiopathic pulmonary brosis; AIP: acute (or accelerated) interstitial pneumonia; DAD: diffuse alveolar damage; PTU: propyl-thiouracil; BCG: bacille Calmette-Guerin; NSAID: nonsteroid anti inammatory; ACE: angiotensin converting enzyme. *: as per gold or methotrexate, it may be difcult to distinguish "hyperdense" cellular NSIP, from true DAD; **: in addition, alveolar haemorrhage may complicate most cases of acute cellular interstitial pneumonia; ?: too few data available.

are capable of inducing several patterns of ILD, and why a given patient will develop a given pattern remain unknown. A notable clue that suggests the drug as a cause, is the fact that the ILD (e.g. nonspecic interstitial pneumonia, organizing pneumonia) will demonstrate a relative refractoriness to steroids, or will recur while the patient is still receiving relatively high dosages of steroids [124]. Of note, long-term treatment with steroids, which are occasionally needed to control severe DI-ILD, expose the patient to signicant risks, including that of severe opportunistic infections in the lung or elsewhere. Imaging The pattern and topographic distribution of opacities in patients with DI-ILD are highly variable. Occasionally, imaging features are very suggestive, if not pathognomonic, of certain types of DI-ILD. Examples include the "foamy" opacities of lipidic density seen on HRCT of patients with mineral oil pneumonia [9], and the asymmetrical, nonsegmental opacities common to many patients with amiodarone lung [147]. Other imaging features are suggestive of a given histopathological pattern of DI-ILD, like the migratory, occasionally painful opacities of BOOP [85], the "melted candlewax-like" opacities in nitrofurantoin lung or in some OP cases [43, 148], the image of "photographic negative of pulmonary oedema" of eosinophilic pneumonia [149], and the distinctive mosaic pattern of desquamative interstitial pneumonia [150].

Future outlook In order to improve current knowledge of DI-ILD, and to have it move beyond the purely descriptive art, which was necessary up to now, it is necessary to: 1) rene and homogenize the diagnostic criteria of all forms of drug-induced respiratory diseases among different countries, to give future publications a common and interpretable framework; 2) help develop and serve drug monitoring systems, in conjunction with responsible agencies, be they country-based or European, with the objective of exhaustively collecting and formatting data on drug-induced adverse respiratory reactions; 3) collect data on associated or other risk factors, including genetic prole, occupational exposure, history (even remote) of exposure to drugs [151]; and 4) evaluate the long-term consequences of drug-induced respiratory diseases, in order to better guide pulmonary physicians in the management (including the handling of steroids) and follow-up of aficted patients. With these data at hand in the future, broader understanding, better prevention, earlier detection, and overall reduction of the severity of drug-induced inltrative lung disease should follow. This may help lawmakers to accurately delineate elds of responsibility, and prepare ways to compensate patients adequately.

Acknowledgements. The authors wish to thank C. Sartini-Camus for invaluable help in rening the manuscript.

DRUG-INDUCED INFILTRATIVE LUNG DISEASE

97s

References 1. 2. 3. 4. 5. 6. White JP, Ward MJ. Drug-induced adverse pulmonary reactions. Adv Drug React Acute Pois Rev 1985; 4: 183211. Cooper JAD, White DA, Matthay RA. Drug-induced pulmonary disease. Part II: noncytotoxic drugs. Am Rev Respir Dis 1986; 133: 488505. Cooper JAD, White DA, Matthay RA. Drug-induced pulmonary disease. Part I: cytotoxic drugs. Am Rev Respir Dis 1986; 133: 321340. Rosenow ECI, Myers JL, Swensen SJ, Pisani RJ. Drug-induced pulmonary disease: an update. Chest 1992; 102: 239250. Foucher P, Biour M, Blayac JP, et al. Drugs that may injure the respiratory system. Eur Respir J 1997; 10: 265279. Delcroix M, Kurz X, Walckiers D, Demedts M, Naeije R. High incidence of primary pulmonary hypertension associated with appetite suppressants in Belgium. Eur Respir J 1998; 12: 271276. Morelock SY, Sahn SA. Drugs and the pleura. Chest 1999; 116: 212221. Coudert B, Bailly F, Andre F, Lombard JN, Camus P. Amiodarone pneumonitis: bronchoalveolar lavage ndings in 15 patients and review of the literature. Chest 1992; 102: 10051012. Gondouin A, Manzoni P, Ranfaing E, et al. Exogenous lipid pneumonia: a retrospective multicentre study of 44 cases in France. Eur Respir J 1996; 9: 14631469. Franquet T, Gimenez A, Bordes R, Rodriguez-Arias JM, Castella J. The crazy-paving pattern in exogenous lipoid pneumonia: CT-pathologic correlation. Am J Roentgenol 1998; 170: 315317. Foucher P, Camus Ph. Pneumotox on the Web. http:// www.pneumotox.com. Last update: July 2001. Rosenow ECI. Drug-induced pulmonary disease. Dis Month 1994; 40: 253312. Cooper JAJ. Drug-induced lung disease. Adv Intern Med 1997; 42: 23168. Whitcomb ME, Domby WR. Drug-induced lung disease. Primary Care 1978; 5: 411423. Camus P. Respiratory disease induced by drugs. Eur Respir J 1997; 10: 260264. Pollak PT. Clinical organ toxicity of antiarrhythmic compounds: Ocular and pulmonary manifestations. Am J Cardiol 1999; 84: 37r45r. Ben-Noun LL. Drug-induced respiratory disorders. Incidence, prevention and management. Drug Safety 2000; 23: 145164. Imokawa S, Colby TV, Leslie KO, Helmers RA. Methotrexate pneumonitis: review of the literature and histopathological ndings in nine patients. Eur Respir J 2000; 15: 373381. Fournier M, Camus P, Benichou C, et al. Pneumo pathies interstitielles: criteres d9imputation a un ` ` medicament. Presse Med 1989; 18: 13331336. Pietropaoli A, Modrak J, Utell M. Interferon-alpha therapy associated with the development of sarcoidosis. Chest 1999; 116: 569572. Savoye G, Goria O, Herve S, et al. Probable cutaneous sarcoidosis associated with combined interferonalpha and ribavirin therapy for chronic hepatitis C. Gastroenterol Clin Biol 2000; 24: 679680. Fiorani C, Sacchi S, Bonacorsi G, Cosenza M. Systemic sarcoidosis associated with interferon-alpha 23. 24. 25.

26.

27. 28.

7. 8.

29. 30.

9. 10.

31. 32. 33.

11. 12. 13. 14. 15. 16. 17. 18.

34. 35. 36. 37. 38.

19. 20. 21.

39. 40.

41. 42.

22.

treatment for chronic myelogenous leukemia. Haematologica 2000; 85: 10061007. Vander Els NJ, Gerdes H. Sarcoidosis and IFN-alpha treatment. Chest 2000; 117: 294. Ogata K, Koga T, Yagawa K. Interferon-related bronchiolitis obliterans organizing pneumonia. Chest 1994; 106: 612613. Dooren MC, Ouwehand WH, Verhoeven AJ, von dem Borne AEGK, Kuijpers RWAM. Adult respiratory distress syndrome after experimental intravenous gamma-globulin concentrate and monocyte-reactive IgG antibodies. Lancet 1998; 352: 16011602. Maillard N, Foucher P, Caillot D, Durand C, Sgro C, Camus P. Transient pulmonary inltrates during treatment with anti-thymocyte globulin. Respiration 1999; 66: 279282. van de Loodsdrecht AA, van Imhoff GW. All-transretinoic acid related pulmonary syndrome. Neth J Med 1999; 54: 131132. Raanani P, Segal E, Levi I, et al. Diffuse alveolar hemorrhage in acute promyelocytic leukemia patients treated with ATRA - A manifestation of the basic disease or the treatment. Leukemia Lymphoma 2000; 37: 605610. Couderc LJ, Stelianides S, Frachon I, et al. Pulmonary toxicity of chemotherapy and G/GM-CSF: a report of ve cases. Respir Med 1999; 93: 6568. Ruiz-Arguelles GJ, Arizpe-Bravo D, Sanchez-Sosa S, Rojas-Ortega S, Moreno-Ford V, Ruiz-Arguelles A. Fatal G-CSF-induced pulmonary toxicity. Am J Hematol 1999; 60: 8283. Popovsky MA. Case Records of the Massachusetts General Hospital - Case 401998. N Engl J Med 1998; 339: 20052012. Kopko PM, Holland PV. Transfusion-related lung injury. Br J Haematol 1999; 105: 322329. Woodard JP, Gulbahce E, Shreve M, et al. Pulmonary cytolytic thrombi: a newly recognized complication of stem cell transplantation. Bone Marrow Transplant 2000; 25: 293300. Tojima H, Yamazaki T, Tokudome T. Two cases of pneumonia caused by Sho-saiko-to. Nippon Kyobu Shikkan Gakkai Zasshi 1996; 34: 904910. Ernst E. Harmless herbs? A review of the recent literature. Am J Med 1998; 104: 170178. Nortier JL, et al. Urothelial carcinoma associated with the use of a chinse herb (Aristolochia fangchi). N Engl J Med 2000; 342: 16861692. Kessler DA. Cancer and herbs. N Engl J Med 2000; 342: 17421743. Temaru R, Yamashita N, Matsui S, Ohta T, Kawasaki A, Kobayashi M. A case of drug induced pneumonitis caused by Saiboku-To. Nippon Kyobu Shikkan Gakkai Zasshi 1994; 32: 485490. Oketani N, Saito H, Ebe T. Pneumonitis due to Hangeshashin-to. Nippon Kyobu Shikkan Gakkai Zasshi 1996; 34: 938988. Hatakeyama S, Tachibana A, Morita M, Suzuki K, Okano H. Five cases of pneumonitis induced by ShoSaiko-to. Nippon Kyobu Shikkan Gakkai Zasshi 1997; 35: 505510. Higenbottam TW. Bronchiolitis obliterans following the ingestion of an Asian shrub leaf. Thorax 1997; 52: S68S72. Chang YL, Yao YT, Wang NS, Lee YC. Segmental necrosis of small bronchi after prolonged intakes of

98s

PH. CAMUS ET AL.

43. 44.

45.

46. 47. 48. 49. 50.

51.

52.

53.

54.

55.

56.

57.

58.

59. 60.

Sauropus androgynus in Taiwan. Am J Respir Crit Care Med 1998; 157: 594598. Ellis SJ, Cleverley JR, Muller NL. Drug-induced lung disease: High-resolution CT ndings. Am J Roentgenol 2000; 175: 10191024. Standertskjold-Nordenstam CG, Wandtke JC, Hood WJ Jr, Zugibe FT, Butler L. Amiodarone pulmonary toxicity. Chest radiography and CT in asymptomatic patients. Chest 1985; 88: 143145. Bedrossian CW, Warren CJ, Ohar J, Bhan R. Amiodarone pulmonary toxicity: cytopathology, ultrastructure, and immunocytochemistry. Ann Diagn Pathol 1997; 1: 4756. Ptzenmeyer P, Foucher P, Piard F, et al. Nilutamide pneumonitis: a report on eight patients. Thorax 1992; 47: 622627. Camus P, Piard F, Ashcroft T, Gal AA, Colby TV. The lung in inammatory bowel disease. Medicine (Baltimore) 1993; 72: 151183. Ptzenmeyer P, Foucher P, Dennewald G, et al. Pleuropulmonary changes induced by ergoline drugs. Eur Respir J 1996; 9: 10131019. Camus P, Colby TV. The lung in inammatory bowel disease. Eur Respir J 2000; 15: 510. Rubio C, Hill ME, Milan S, O9Brien MER, Cunningham D. Idiopathic pneumonia syndrome after high dose chemotherapy for relapsed Hodgkin9s disease. Br J Cancer 1997; 75: 10441048. Wilczynski SW, Erasmus JJ, Petros WP, Vredenburgh JJ, Folz RJ. Delayed pulmonary toxicity syndrome following high-dose chemotherapy and bone marrow transplantation for breast cancer. Am J Respir Crit Care Med 1998; 157: 565573. Kantrow SP, Hackman RC, Boeckh M, Myerson D, Crawford SW. Idiopathic pneumonia syndrome Changing spectrum of lung injury after marrow transplantation. Transplantation 1997; 63: 10791086. Bhalla KS, Wilczynski SW, Abushamaa AM, et al. Pulmonary toxicity of induction chemotherapy prior to standard or high-dose chemotherapy with autologous hematopoietic support. Am J Respir Crit Care Med 2000; 161: 1725. Nemery B, Bast A, Behr J, et al. Interstitial lung disease induced by exogenous agents: factors governing susceptibility. Eur Respir J 2001; 18: Suppl. 32, 30s42s. Aronin PA, Mahaley MSJ, Rudnick SA, et al. prediction of BCNU pulmonary toxicity in patients with malignant gliomas: an assessment of risk factors. N Engl J Med 1980; 303: 183188. Real E, Roca MJ, Vinuales A, Pastor E, Grau E. Life threatening lung toxicity induced by low doses of bleomycin in a patient with Hodgkin9s disease. Haematologica 1999; 84: 667668. Alliot C, Tabuteau S, Desablens B, Aubry P, Andrejak M. Fatal pulmonary brosis after a low cumulated dose of bleomycin: Role of alpha1-antitrypsin deciency? Am J Hematol 1999; 62: 198199. Kautekeete ML, Bourgeois N, Potvin P, et al. Hypersensitivity with hepatotoxicity to mesalazine after hypersensitivity to sulfasalazine. Gastroenterology 1992; 103: 19351937. Sitbon O, Bidel N, Dussopt C, et al. Minocycline pneumonitis and eosinophilia: a report on 8 patients. Arch Intern Med 1994; 154: 16331640. Acker CG, Greenberg A. Angioedema induced by the

61.

62. 63.

64.

65.

66. 67. 68.

69.

70. 71.

72. 73. 74.

75.

76. 77.

angiotensin II blocker losartan. N Engl J Med 1995; 333: 1572. de Vriese ASP, Philippe J, Van Renterghem DM, De Cuyper CA, Hindryckx EGJ, Louagie A. Carbamazepine hypersensitivity syndrome: report of 4 cases and review of the literature. Medicine (Baltimore) 1995; 74: 144150. Dietz A, Hubner C, Andrassy K. Makrolid-Antibiotika induzierte Vaskulitis (Churg-Strauss-Syndrom). Laryngo-Rhino-Otol 1998; 77: 111114. Salaf F, Manganelli P, Carotti M, Subiaco S, Lamanna G, Cervini C. Methotrexate-induced pneumonitis in patients with rheumatoid arthritis and psoriatic arthritis: report of ve cases and review of the literature. Clin Rheumatol 1997; 16: 296304. Hillerdal G, Lee J, Blomkvist A, et al. Pleural disease during treatment with bromocriptine in patients previously exposed to asbestos. Eur Respir J 1997; 10: 27112715. Knoop E, Mairesse M, Lenclud C, Gevenois PA, De Vuyst P. Pleural effusion during bromocriptine exposure in two patients with preexisting asbestos pleural plaques: a relationship? Eur Respir J 1998; 10: 28982901. De Vuyst P, Ptzenmeyer P, Camus P. Asbestos, ergot drugs and the pleura. Eur Respir J 1997; 10: 2695 2698. de Wildt SN, Kearns GL, Leeder JS, van den Anker JN. Cytochrome P450 3A - Ontogeny and drug disposition. Clin Pharmacokinet 1999; 37: 485505. Suriyachan D, Satyapan N, Pulsri Y, Singhakowinta A, Tanprasert P. ACE inhibitors and cough in Thai patients: a preliminary report. Asia Pacic J Pharmacol 1995; 10: 1719. Voigt MD, Workman B, Lombard C, Kirsch RE. Halothane hepatitis in a South African population. Frequency and the inuence of gender and ethnicity. S A Med J 1997; 87: 882885. Ajayi AAL, Adigun AQ. Angioedema and cough in Nigerian patients receiving ACE inhibitors. Br J Clin Pharmacol 2000; 50: 8182. Russell GI, Bing RF, Jones JA, Thurston H, Swales JD. Hydralazine sensitivity: clinical features, autoantibody changes and HLA-DR phenotype. Q J Med 1987; 65: 84552. Lin JH, Lu AYH. Inhibition and induction of cytochrome P450 and the clinical implications. Clin Pharmacokinet 1998; 35: 361390. Gilson AJ, Sahn SA. Reactivation of bleomycin lung toxicity following oxygen administration. A second response to corticosteroids. Chest 1985; 88: 304306. Schweitzer VG, Juillard GJF, Bajada CL, Parker RG. Radiation recall dermatitis and pneumonitis in a patient treated with paclitaxel. Cancer 1995; 76: 10691072. Ma LD, Taylor GA, Wharam MD, Wiley JM. Recall pneumonitis: adriamycin potentiation of radiation pneumonitis in two children. Radiology 1993; 187: 465467. Thomas PS, Agrawal S, Gore M, Geddes DM. Recall lung pneumonitis due to carmustine after radiotherapy. Thorax 1995; 50: 11161118. Sleijfer S, van der Mark TW, Schraffordt Koops H, Mulder NH. Enhanced effects of bleomycin on pulmonary function disturbances in patients with decreased renal function due to cisplatin. Eur J Cancer 1996; 32A: 550552.

DRUG-INDUCED INFILTRATIVE LUNG DISEASE

99s

78.

79.

80. 81. 82.

83.

84.

85. 86. 87. 88. 89. 90. 91.

92.

93.

94.

95. 96.

Gerson R, Tellez-Bernal E, Lazaro-Leon M, et al. Low toxicity with continuous infusion of high-dose bleomycin in poor prognostic testicular cancer. Am J Clin Oncol 1993; 16: 323326. Avila JJ, Lympany PA, Pantelidis P, Welsh KI, Black CM, duBois RM. Fibronectin gene polymorphisms associated with brosing alveolitis in systemic sclerosis. Am J Respir Cell Mol Biol 1999; 20: 106112. Cottin V, Tebib J, Massonnet B, Souquet PJ, Bernard J-P. Pulmonary function in patients receiving longterm low-dose methotrexate. Chest 1996; 109: 933938. Wolkowicz J, Sturgeon J, Rawji M, Chan CK. Bleomycin-induced pulmonary function abnormalities. Chest 1992; 101: 97101. Dunsford ML, Mead GM, Bateman AC, Cook T, Tung K. Severe pulmonary toxicity in patients treated with a combination of docetaxel and gemcitabine for metastatic transitional cell carcinoma. Ann Oncol 1999; 10: 943947. Abramov Y, Elchalal U, Schenker JG. Pulmonary manifestations of severe ovarian hyperstimulation syndrome: a multicenter study. Fertility Sterility 1999; 71: 645651. Crestani B, Kambouchner M, Soler P, et al. Migratory bronchiolitis obliterans organizing pneumonia after unilateral radiation therapy for breast carcinoma. Eur Respir J 1995; 8: 31821. Faller M, Quoix E, Popin E, et al. Migratory pulmonary inltrates in a patient treated with sotalol. Eur Respir J 1997; 10: 21592162. Cazzato S, Zompatori M, Baruzzi G, et al. Bronchiolitis obliterans-organizing pneumonia: an Italian experience. Respir Med 2000; 94: 702708. Ramanathan RK, Belani CP. Transient pulmonary inltrates: a hypersensitivity reaction to Paclitaxel. Ann Intern Med 1996; 124: 278. Geller M, Dickie HA, Kass DA, Hafez GR, Gillepsie JJ. The histopathology of acute nitrofurantoin-associated pneumonitis. Ann Allergy 1976; 37: 275279. Reinhart HH, Reinhart E, Korlipara P, Peleman R. Combined nitrofurantoin toxicity to liver and lung. Gastroenterology 1992; 102: 13961399. Brain C, Mac Ardle B, Levin S. Idiosyncratic reactions to carbamazepine mimicking viral infection in children. BMJ 1984; 289: 354. Matuschak GM. Pseudosepsis syndrome, multiplesystem organ failure, and chronic salicylate intoxication. Inhibition of regulatory eicosanoids? Chest 1991; 100: 11881189. Kaufman D, Pichler W, Beer JH. Severe episode of high fever with rash, lymphadenopathy, neutropenia, and eosinophilia after minocycline therapy for acne. Arch Intern Med 1994; 154: 19831984. Knowles SR, Shapiro L, Shear NH. Serious adverse reactions induced by minocycline: report of 13 patients and review of the literature. Arch Dermatol 1996; 132: 934939. Bourezane Y, Salard D, Hoen B, Vandel S, Drobacheff C, Laurent R. DRESS (Drug Rash with Eosinophilia and Systemic Symptoms) syndrome associated with nevirapine therapy. Clin Infect Dis 1998; 27: 1321 1322. Marik P. Anticonvulsant hypersensitivity syndrom occurring as sepsis with multiorgan dysfucntion. Pharmacotherapy 1999; 19: 346348. Skaer TL. Medication-induced systemic lupus erythematosus. Clin Ther 1992; 14: 496506.

97. 98. 99.

100.

101.

102.

103.

104.

105. 106. 107.

108.

109. 110. 111. 112. 113. 114. 115.

Khosla R, Butman AN, Hammer DF. Simvastatininduced lupus erythematosus. South Med J 1998; 91: 873874. Stridhar MK, Abdulla A. Fatal lupus-like syndrome and ARDS induced by uvastatin. Lancet 1998; 352: 114. Phillips D, Phillips B, Mannino D. A case study and national database report of progressive systemic sclerosis and associated conditions. J Womens Health 1998; 7: 10991104. Ohtsuka M, Yamashita Y, Doi M, Hasegawa S. Propylthiouracil-induced alveolar haemorrhage associated with antineutrophil cytoplasmic antibody. Eur Respir J 1997; 10: 14051407. Chastain MA, Russo GG, Boh EE, Chastain JB, Falabella A, Millikan LE. Propylthiouracil hypersensitivity: Report of two patients with vasculitis and review of the literature. J Am Acad Dermatol 1999; 41: 757764. Choi HK, Merkel PA, Cohen-Tervaert JW, Black RM, McCluskey RT, Niles JL. Alternating antineutrophil cytoplasmic antibody specicity. Drug-induced vasculitis in a patient with Wegener9s granulomatosis. Arthr Rheum 1999; 42: 384388. Dhillon SS, Singh D, Doe N, Qadri AM, Ricciardi S, Schwarz MI. Diffuse alveolar hemorrhage and pulmonary capillaritis due to propylthiouracil. Chest 1999; 116: 14851488. Gunton JE, Stiel J, Caterson RJ, McElduff A. Antithyroid drugs and antineutrophil cytoplasmic antibody positive vasculitis. A case report and review of the literature. J Clin Endocrinol Metab 1999; 84: 1316. Morita S, Ueda Y, Eguchi K. Anti-thyroid druginduced ANCA-associated vasculitis: A case report and review of the literature. Endocrine J 2000; 47: 467470. Elkayam O, Yaron M, Caspi D. Minocycline-induced arthritis associated with fever, livedo reticularis, and p-ANCA. Ann Rheum Dis 1996; 55: 769771. Laux-End R, Inaebnit D, Gerber HA, Bianchetti MG. Vasculitis associated with levamisole and circulating autoantibodies. Archives of Disease in Childhood 1996; 75: 355356. Salerno SM, Ormseth EJ, Roth BJ, Meyer CA, Christensen ED, Dillard TA. Sulfasalazine pulmonary toxicity in ulcerative colitis mimicking clinical features of Wegener9s granulomatosis. Chest 1996; 110: 556 559. Choi HK, Merkel PA, Niles JL. ANCA-positive vasculitis associated with allopurinol therapy. Clin Exp Rheum 1998; 16: 743744. Schmitz Schumann M, Palca A, Simon U, Blaser K. Aspirin-induced asthma and Churg-Strausssyndrome. Eur J Clin Invest 1998; 28: Suppl. 1, A49. Green RL, Vayonis AG. Churg-Strauss syndrome after zarlukast in two patients not receiving systemic steroid treatment. Lancet 1999; 353: 725726. Pedvis S, Anastakis D, Inman R. Churg-Strauss syndrome associated with zarlukast (abstract). J Rheumatol 1999; 26: 1630. Katz RS, Papernik M. Zarlukast and Churg-Strauss syndrome. JAMA 1998; 279: 1949. Knoell DL, Lucas J, Allen JN. Churg-Strauss syndrome associated with zarlukast. Chest 1998; 114: 332334. Fauroux B, Meyer-Milsztain A, Boccon-Gibbod L, et al. Cytotoxic drug-induced pulmonary disease in infants and children. Pediatr Pulmonol 1994; 18: 347 355.

100s

PH. CAMUS ET AL.

116. O9Driscoll BR, Hasleton PS, Taylor PM, Poulter LW, Gattamaneni HR, Woodcock AA. Active lung brosis up to 17 years after chemotherapy with carmustine (BCNU) in childhood. N Engl J Med 1990; 323: 378 382. 117. LeMense GP, Strange C. Granulomatous pneumonitis following intravesical BCG. What therapy is needed? Chest 1994; 106: 16241626. 118. Mooren FC, Lerch MM, Ullerich H, Burger H, Domschke W. Systemic granulomatous disease after intravesical BCG instillation. BMJ 2000; 320: 219. 119. Naccache JM, Antoine M, Wislez M, et al. Sarcoidlike pulmonary disorder in human immunodeciency virus-infected patients receiving antiretroviral therapy. Am J Respir Crit Care Med 1999; 159: 20092013. 120. Ptzenmeyer P, Meier M, Zuck P, et al. Piroxicaminduced pulmonary inltrates and eosinophilia. J Rheumatol 1994; 21: 15731577. 121. Benzaquen-Forner H, Dournovo P, TandjaouiLambiotte H, et al. Pneumopathie hypoxemiante sous traitement par IEC. Rev Mal Respir 1998; 15: 804810. 122. Camus P, Lombard JN, Perrichon M, et al. Bronchiolitis obliterans organising pneumonia in patients taking acebutolol or amiodarone. Thorax 1989; 44: 711715. 123. Cohen MB, Austin JHM, Smith-Vaniz A, Lutzky J, Grimes MM. Nodular bleomycin toxicity. Am J Clin Pathol 1989; 92: 101104. 124. Bone RC, Wolfe J, Sobonya RE, et al. Desquamative interstitial pneumonia following long-term nitrofurantoin therapy. Am J Med 1976; 60: 697701. 125. Chamberlain DW, Hyland RH, Ross DJ. Diphenylhydantoin-induced lymphocytic interstitial pneumonia. Chest 1986; 90: 458460. 126. Sobol SM, Rakita L. Pneumonitis and pulmonary brosis associated with amiodarone treatment: a possible complication of a new antiarrythmic drug. Circulation 1982; 65: 819824. 127. Israel CM. Severe pneumonitis as complication of low-dose methotrexate therapy in psoriasis-related polyarthritis. Reply. Dtsch Med Wochenschr 1995; 120: 12241225. 128. Suwa A, Hirakata M, Satoh S, Mimori T, Utsumi K, Inada S. Rheumatoid arthritis associated with methotrexate-induced pneumonitis: Improvement with i.v. cyclophosphamide therapy. Clin Exp Rheumatol 1999; 17: 355358. 129. Leng PH, Murillo B, Fraire A. Acute interstitial pneumonitis occurring after consolidation chemotherapy with high-dose cytarabine for acute myelogenous leukemia. Chest 1999; 116: 409410. 130. Attar EC, Ervin T, Janicek M, Deykin A, Godleski J. Acute interstitial pneumonitis related to gemcitabine. J Clin Oncol 2000; 18: 697698. 131. Wilschut FA, Cobben NAM, Thunnissen FBJM, Lammers LJS, Wouters EFM, Drent M. Recurrent respiratory distress associated with carbamazepine overdose. Eur J Respir Dis 1997; 10: 21632165. 132. Woolley J, Collett J, Goldstein D. Diffuse alveolar damage following a single administration of a cyclophosphamide containing chemotherapy regimen. Aust N Z J Med 1997; 27: 605606. 133. Louie S, Gamble CN, Cross CE. Penicillamine associated pulmonary hemorrhage. J Rheumatol 1986; 13: 963966. 134. Santalo M, Domingo P, Fontcuberta J, Franco M,

135. 136. 137.

138.

139.

140.

141. 142.

143. 144.

145. 146. 147.

148. 149.

150.

151.

Nolla J. Diffuse pulmonary hemorrhage associated with anticoagulant therapy. Eur J Respir Dis 1986; 69: 114119. Bucknall CE, Adamson MR, Banham SW. Non fatal pulmonary haemorrhage associated with nitrofurantoin. Thorax 1987; 42: 475476. Sleiman C, Raffy O, Roue C, Mal H. Fatal pulmonary hemorrhage during high-dose valproate monotherapy. Chest 2000; 117: 613. Yigla M, Sprecher E, Azzam Z, Guralnik L, Kapeliovich M, Krivoy N. Diffuse alveolar hemorrhage following thrombolytic therapy for acute myocardial infarction. Respiration 2000; 67: 445448. Aymard JP, Gyger M, Lavallee R, Legresley LP, Desy M. A case of pulmonary alveolar proteinosis complicating chronic myelogenous leukemia. A peculiar pathologic aspect of busulfan lung? Cancer 1984; 53: 954956. Maehara K, Shiraki M, Hara I, Koike S, Matsumiya T. Possible association of drug therapy with pulmonary alveolar proteinosis (PAP) in patients with malignant haematologicaldisorders. Jpn J Pharmacol 1999; 79: 285P. Colby TV, Lombard C, Yousem SA, Kitaichi M. Atlas of pulmonary surgical pathology. In: Mitchell J, ed. Atlases in Diagnostic Surgical Pathology. Philadelphia, WB Saunders, Harcourt Brace Jovanovich, 1991; p. 380. Colby TV. Surgical pathology of non-neoplastic lung disease. Modern Pathol 2000; 13: 343358. Katzenstein ALA, Myers JL. Nonspecic interstitial pneumonia and the other idiopathic interstitial pneumonias: Classication and diagnostic criteria. Am J Surg Pathol 2000; 24: 13. Theile A, Kemper P. Bleomycin-induced pulmonary brosis after chemotherapy of an ovarian granulosa cell tumor. Pneumologie 1998; 52: 325332. Cameron RJ, Kolbe J, Wilsher ML, Lambie N. Bronchiolitis obliterans organising pneumonia associated with the use of nitrofurantoin. Thorax 2000; 55: 249351. Bandla HPR, Davis SH, Hopkins NE. Lipoid pneumonia: A silent complication of mineral oil aspiration. Pediatrics 1999; 103: E191E194. Myers JL, Kennedy JI, Plumb VJ. Amiodarone lung: pathologic ndings in clinically toxic patients. Hum Pathol 1987; 18: 349354. Ren H, Kuhlman JE, Hruban RH, Fishman EK, Wheeler PS, Hutchins GM. CT-pathology correlation of amiodarone lung. J Comput Assist Tomogr 1990; 14: 760765. Cameron C, Bayliff CD, Patterson NAM. Nitrofurantoin-associated acute pulmonary toxicity. Can J Hosp Pharm 1999; 52: 8386. Gaensler EA, Carrington CB. Peripheral opacities in chronic eosinophilic pneumonia: the photographic negative of pulmonary edema. Am J Roentgenol 1977; 128: 113. Hartman TE, Primack SL, Swensen SJ, Hansell D, McGuiness G, Muller NL. Desquamative interstitial pneumonia: thin-section CT ndings in 22 patients. Radiology 1993; 187: 787790. Hubbard R, Venn A, Smith C, Cooper M, Johnston I, Britton J. Exposure to commonly prescribed drugs and the etiology of cryptogenic brosing alveolitis. Am J Respir Crit Care Med 1998; 157: 743747.

Вам также может понравиться