1
|
Zhou C, Deng H, Yang Y, Wang F, Lin X, Liu M, Xie X, Luan T, Zhong N. Cancer therapy-related interstitial lung disease. Chin Med J (Engl) 2025; 138:264-277. [PMID: 39402974 PMCID: PMC11771665 DOI: 10.1097/cm9.0000000000003149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Indexed: 01/29/2025] Open
Abstract
ABSTRACT With the increasing utilization of cancer therapy, the incidence of lung injury associated with these treatments continues to rise. The recognition of pulmonary toxicity related to cancer therapy has become increasingly critical, for which interstitial lung disease (ILD) is a common cause of mortality. Cancer therapy-related ILD (CT-ILD) can result from a variety of treatments including chemotherapy, targeted therapy, immune checkpoint inhibitors, antibody-drug conjugates, and radiotherapy. CT-ILD may progress rapidly and even be life-threatening; therefore, prompt diagnosis and timely treatment are crucial for effective management. This review aims to provide valuable information on the risk factors associated with CT-ILD; elucidate its underlying mechanisms; discuss its clinical features, imaging, and histological manifestations; and emphasize the clinical-related views of its diagnosis. In addition, this review provides an overview of grading, typing, and staging treatment strategies used for the management of CT-ILD.
Collapse
Affiliation(s)
- Chengzhi Zhou
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Haiyi Deng
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Yilin Yang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Fei Wang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Xinqing Lin
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Ming Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Xiaohong Xie
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Tao Luan
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| |
Collapse
|
2
|
Drent M, Wijnen PA, Jessurun NT, Harmsze AM, Bekers O, Bast A. Drug-Gene Risk Stratification in Patients with Suspected Drug-Induced Interstitial Lung Disease. Drug Saf 2024; 47:355-363. [PMID: 38460070 PMCID: PMC10955005 DOI: 10.1007/s40264-024-01400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Pulmonary toxicity has been associated with drug use. This is often not recognized in clinical practice, and underestimated. OBJECTIVE We aimed to establish whether polymorphisms in certain genes corresponding with a metabolic pathway of drug(s) used are associated with pulmonary toxicity in patients with suspected drug-induced interstitial lung disease (DI-ILD). METHODS This retrospective observational study explored genetic variations in three clinically relevant cytochrome P450 (CYP) iso-enzymes (i.e., CYP2D6, CYP2C9, and CYP2C19) in a group of patients with a fibroticinterstitial lung disease, either non-specific interstitial pneumonia (n = 211) or idiopathic pulmonary fibrosis (n = 256), with a suspected drug-induced origin. RESULTS Of the 467 patients, 79.0% showed one or more polymorphisms in the tested genes accompanied by the use of drug(s) metabolized by a corresponding affected metabolic pathway (60.0% poor metabolizers and/or using two or more drugs [likely DI-ILD], 37.5% using three or more [highly likely DI-ILD]). Most commonly used drugs were statins (63.1%) with a predominance among men (69.4 vs 47.1%, p < 0.0001). Nitrofurantoin, not metabolized by the tested pathways, was prescribed more frequently among women (51.9 vs 4.5%, p < 0.00001). CONCLUSIONS In our cohort with suspected DI-ILD, 79% carried one or more genetic variants accompanied by the use of drugs metabolized by a corresponding affected pathway. In 60%, the diagnosis of DI-ILD was likely, whereas in 37.5%, it was highly likely, based on CYP analyses. This study underlines the importance of considering both drug use and genetic make-up as a possible cause, or at least a contributing factor, in the development and/or progression of fibrotic lung diseases. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov identifier NCT00267800, registered in 2005.
Collapse
Affiliation(s)
- Marjolein Drent
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine, and Life Science, Maastricht University, Maastricht, The Netherlands.
- ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, The Netherlands.
- Research Team, ILD Care Foundation, Heideoordlaan 8, 6711NR, Ede, The Netherlands.
| | - Petal A Wijnen
- ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, The Netherlands
- Research Team, ILD Care Foundation, Heideoordlaan 8, 6711NR, Ede, The Netherlands
- Department of Clinical Chemistry, Central Diagnostic Laboratory, Maastricht University Medical Centre, P Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | - Naomi T Jessurun
- Research Team, ILD Care Foundation, Heideoordlaan 8, 6711NR, Ede, The Netherlands
- Netherlands Pharmacovigilance Centre Lareb, Goudsbloemvallei 7, 5237 MH, 's-Hertogenbosch, The Netherlands
| | - Ankie M Harmsze
- ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, The Netherlands
- Department of Clinical Pharmacology, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Otto Bekers
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine, and Life Science, Maastricht University, Maastricht, The Netherlands
- Department of Clinical Chemistry, Central Diagnostic Laboratory, Maastricht University Medical Centre, P Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | - Aalt Bast
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine, and Life Science, Maastricht University, Maastricht, The Netherlands
- Research Team, ILD Care Foundation, Heideoordlaan 8, 6711NR, Ede, The Netherlands
| |
Collapse
|
3
|
Harrison M, Kavanagh G, Corte TJ, Troy LK. Drug-induced interstitial lung disease: a narrative review of a clinical conundrum. Expert Rev Respir Med 2024; 18:23-39. [PMID: 38501199 DOI: 10.1080/17476348.2024.2329612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Drug-induced interstitial lung disease (DI-ILD) is increasing in incidence, due to the use of many new drugs across a broad range of cancers and chronic inflammatory diseases. The presentation and onset of DI-ILD are variable even for the same drug across different individuals. Clinical suspicion is essential for identifying these conditions, with timely drug cessation an important determinant of outcomes. AREAS COVERED This review provides a comprehensive and up-to-date summary of epidemiology, risk factors, pathogenesis, diagnosis, treatment, and prognosis of DI-ILD. Relevant research articles from PubMed and Medline searches up to September 2023 were screened and summarized. Specific drugs including immune checkpoint inhibitors, CAR-T cell therapy, methotrexate, and amiodarone are discussed in detail. The potential role of pharmacogenomic profiling for lung toxicity risk is considered. EXPERT OPINION DI-ILD is likely to be an increasingly important contributor to respiratory disability in the community. These conditions can negatively impact quality of life and patient longevity, due to associated respiratory compromise as well as cessation of evidence-based therapy for the underlying disease. This clinical conundrum is relevant to all areas of medicine, necessitating increased understanding and greater vigilance for drug-related lung toxicity.
Collapse
Affiliation(s)
- Megan Harrison
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Grace Kavanagh
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Tamera J Corte
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Lauren K Troy
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
4
|
Motamedi M, Ferrara G, Yacyshyn E, Osman M, Abril A, Rahman S, Netchiporouk E, Gniadecki R. Skin disorders and interstitial lung disease: Part I-Screening, diagnosis, and therapeutic principles. J Am Acad Dermatol 2023; 88:751-764. [PMID: 36228941 DOI: 10.1016/j.jaad.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/26/2022] [Accepted: 10/02/2022] [Indexed: 11/07/2022]
Abstract
Numerous inflammatory, neoplastic, and genetic skin disorders are associated with interstitial lung disease (ILD), the fibrosing inflammation of lung parenchyma that has significant morbidity and mortality. Therefore, the dermatologist plays a major role in the early detection and appropriate referral of patients at risk for ILD. Part 1 of this 2-part CME outlines the pathophysiology of ILD and focuses on clinical screening and therapeutic principles applicable to dermatological patients who are at risk for ILD. Patients with clinical symptoms of ILD should be screened with pulmonary function tests and high-resolution chest computed tomography. Screening for pulmonary hypertension should be considered in high-risk patients. Early identification and elimination of pulmonary risk factors, including smoking and gastroesophageal reflux disease, are essential in improving respiratory outcomes. First-line treatment interventions for ILD in a dermatological setting include mycophenolate mofetil, but the choice of therapeutic agents depends on the nature of the primary disease, the severity of ILD, and comorbidities and should be the result of a multidisciplinary assessment. Better awareness of ILD among medical dermatologists and close interdisciplinary collaborations are likely to prevent treatment delays improving long-term outcomes.
Collapse
Affiliation(s)
- Melika Motamedi
- Division of Dermatology, University of Alberta, Edmonton, Alberta, Canada
| | - Giovanni Ferrara
- Division of Pulmonary Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Elaine Yacyshyn
- Division of Rheumatology, University of Alberta, Edmonton, Alberta, Canada
| | - Mohammed Osman
- Division of Rheumatology, University of Alberta, Edmonton, Alberta, Canada
| | - Andy Abril
- Division of Rheumatology, Mayo Clinic, Jacksonville, Florida
| | - Samia Rahman
- Division of Dermatology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Robert Gniadecki
- Division of Dermatology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW Sarcoidosis is a chronic multisystemic inflammatory disease of unknown aetiology with a wide range of highly variable clinical manifestations and unpredictable disease course. Sarcoidosis patients may present with specific organ-related symptoms involving functional impairments, and less specific symptoms. The decision whether and when to treat a sarcoidosis patient with pharmacotherapy depends on two major factors: risk of organ failure and/or death and impairment of quality of life. This decision is complex and not standardized. RECENT FINDINGS Glucocorticoids (GCs) are recommended as initial treatment, when needed. Subsequent GC-sparing alternatives frequently follow. Comorbidities or adverse drug reactions (ADRs) from drugs used in sarcoidosis treatment are sometimes very hard to differentiate from symptoms associated with the disease itself, which may cause diagnostic dilemmas. An ideal approach to minimalize ADRs would involve genetic screening prior to prescribing certain 'high-risk drugs' and therapeutic drug monitoring during treatment. Pharmacogenomic testing aims to guide appropriate selection of medicines, with the potential of reducing unnecessary polypharmacy while improving clinical outcomes. SUMMARY A multidisciplinary approach to the management of sarcoidosis may avoid unnecessary ADRs. It is important to consider the possibility of drug-induced damage in sarcoidosis, especially if the clinical situation deteriorates after the introduction of a particular drug.
Collapse
Affiliation(s)
- Marjolein Drent
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht
- ILD Center of Excellence, Department of Respiratory Medicine, St. Antonius Hospital, Nieuwegein
- ILD Care Foundation Research Team, Ede
| | - Naomi T. Jessurun
- ILD Care Foundation Research Team, Ede
- Netherlands Pharmacovigilance Centre Lareb, ‘s-Hertogenbosch
| | - Petal A. Wijnen
- ILD Care Foundation Research Team, Ede
- Central Diagnostic Laboratory, Department of Clinical Chemistry, MUMC, Maastricht, The Netherlands
| | - Otto Bekers
- Central Diagnostic Laboratory, Department of Clinical Chemistry, MUMC, Maastricht, The Netherlands
| | - Aalt Bast
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht
- ILD Care Foundation Research Team, Ede
| |
Collapse
|
6
|
Role of Drug-Gene Interactions and Pharmacogenetics in Simvastatin-Associated Pulmonary Toxicity. Drug Saf 2021; 44:1179-1191. [PMID: 34606062 PMCID: PMC8553720 DOI: 10.1007/s40264-021-01105-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 11/05/2022]
Abstract
Introduction Simvastatin has previously been associated with drug-induced interstitial lung disease. In this retrospective observational study, cases with non-specific interstitial pneumonia (NSIP) or idiopathic pulmonary fibrosis (IPF) with simvastatin-associated pulmonary toxicity (n = 34) were evaluated. Objective To identify whether variations in genes encoding cytochrome P450 (CYP) enzymes or in the SLCO1B1 gene (Solute Carrier Organic anion transporting polypeptide 1B1 gene, encoding the organic anion transporting polypeptide 1B1 [OATP1B1] drug transporter enzyme), and/or characteristics of concomitantly used drugs, predispose patients to simvastatin-associated pulmonary toxicity. Methods Characteristics of concomitantly used drugs and/or variations in the CYP or SLCO1B1 genes and drug–gene interactions were assessed. The outcome after withdrawal of simvastatin and/or switch to another statin was assessed after 6 months. Results Multiple drug use involving either substrates and/or inhibitors of CYP3A4 and/or three or more drugs with the potential to cause acidosis explained the simvastatin-associated toxicity in 70.5% (n = 24) of cases. Cases did not differ significantly from controls regarding CYP3A4, CYP2C9, or OATP1B1 phenotypes, and genetic variation explained only 20.6% (n = 7) of cases. Withdrawal of simvastatin without switching to another statin or with a switch to a hydrophilic statin led to improvement or stabilization in all NSIP cases, whereas all cases who were switched to the lipophilic atorvastatin progressed. Conclusion Simvastatin-associated pulmonary toxicity is multifactorial. For patients with this drug-induced pulmonary toxicity who need to continue taking a statin, switching to a hydrophilic statin should be considered. ClinicalTrials.gov identifier NCT00267800, registered in 2005. Supplementary Information The online version contains supplementary material available at 10.1007/s40264-021-01105-8.
Collapse
|
7
|
Lee H. Vitamin E acetate as linactant in the pathophysiology of EVALI. Med Hypotheses 2020; 144:110182. [PMID: 33254504 PMCID: PMC7422838 DOI: 10.1016/j.mehy.2020.110182] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/25/2020] [Accepted: 08/11/2020] [Indexed: 11/11/2022]
Abstract
The recent identification of Vitamin E acetate as one of the causal agents for the e-cigarette, or vaping, product use associated lung injury (EVALI) is a major milestone. In membrane biophysics, Vitamin E is a linactant and a potent modulator of lateral phase separation that effectively reduces the line tension at the two-dimensional phase boundaries and thereby exponentially increases the surface viscosity of the pulmonary surfactant. Disrupted dynamics of respiratory compression-expansion cycling may result in an extensive hypoxemia, leading to an acute respiratory distress entailing the formation of intraalveolar lipid-laden macrophages. Supplementation of pulmonary surfactants which retain moderate level of cholesterol and controlled hypothermia for patients are recommended when the hypothesis that the line-active property of the vitamin derivative drives the pathogenesis of EVALI holds.
Collapse
Affiliation(s)
- Hanjun Lee
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States.
| |
Collapse
|
8
|
Gomes ESR, Marques ML, Regateiro FS. Epidemiology and Risk Factors for Severe Delayed Drug Hypersensitivity Reactions. Curr Pharm Des 2020; 25:3799-3812. [PMID: 31694518 DOI: 10.2174/1381612825666191105115346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023]
Abstract
Severe delayed drug hypersensitivity reactions comprise different clinical entities and can involve different immune-mediated mechanisms. Common examples are severe cutaneous adverse reactions and druginduced internal organ injuries. The incidence of such reactions is overall low but seems to be on the rise reaching numbers as high as 9 per million individuals-years in the case of SJS/TEN and DRESS. Such conditions carry an important associated morbidity, and mortality can attain 40% in SJS/TEN patients, making these hypersensitivity reactions important targets when implementing preventive measures. Several risk factors have been identified for reaction severity; some are transverse, such as older age and underlying chronic diseases. The recent advances in pharmacogenetics allowed the identification of specific populations with higher risk and permitted strategic avoidance of certain drugs being HLA-B*57:01 screening in patients initiating abacavir the best successful example. In this work, we reviewed the epidemiology of SCARs and liver/kidney/lung drug-induced immune-mediated reactions. We focus on particular aspects such as prevalence and incidence, drugs involved, mortality and risk factors.
Collapse
Affiliation(s)
- Eva S R Gomes
- Allergy and Clinical Immunology Department, Centro Hospitalar e Universitario do Porto, Porto, Portugal
| | - Maria L Marques
- Allergy and Clinical Immunology Department, Centro Hospitalar e Universitario do Porto, Porto, Portugal
| | - Frederico S Regateiro
- Allergy and Clinical Immunology Department, Centro Hospitalar e Universitario de Coimbra, Coimbra, Portugal.,Institute of Immunology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Reseach (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
9
|
Matsumoto K, Nakao S, Hasegawa S, Matsui T, Shimada K, Mukai R, Tanaka M, Uranishi H, Nakamura M. Analysis of drug-induced interstitial lung disease using the Japanese Adverse Drug Event Report database. SAGE Open Med 2020; 8:2050312120918264. [PMID: 32528682 PMCID: PMC7262990 DOI: 10.1177/2050312120918264] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Objectives: Drug-induced interstitial lung disease occurs when exposure to a drug causes
inflammation and, eventually, fibrosis of the lung interstitium.
Drug-induced interstitial lung disease is associated with substantial
morbidity and mortality. The aim of this retrospective study was to obtain
new information on the time-to-onset profiles of drug-induced interstitial
lung disease by consideration of other associated clinical factors using the
Japanese Adverse Drug Event Report database. Methods: We identified and analyzed reports of drug-induced interstitial lung disease
between 2004 and 2018 from the Japanese Adverse Drug Event Report database.
The reporting odds ratio and 95% confidence interval was used to detect the
signal for each drug-induced interstitial lung disease incidence. We
evaluated the time-to-onset profile of drug-induced interstitial lung
disease and used the applied association rule mining technique to uncover
undetected relationships, such as possible risk factors. Results: The reporting odds ratios (95% confidence intervals) of drug-induced
interstitial lung disease due to temsirolimus, gefitinib, sho-saiko-to,
sai-rei-to, osimertinib, amiodarone, alectinib, erlotinib, everolimus, and
bicalutamide were 18.3 (15.6–21.3), 17.8 (16.5–19.2), 16.3 (11.8–22.4), 14.5
(11.7–18.2), 12.5 (10.7–14.7), 10.9 (9.9–11.9), 10.6 (8.1–13.9), 9.6
(8.8–10.4), 9.4 (8.7–10.0), and 9.2 (7.9–10.6), respectively. The median
durations (day (interquartile range)) for drug-induced interstitial lung
disease were as follows: amiodarone (123.0 (27.0–400.5)), methotrexate
(145.5 (67.8–475.8)), fluorouracil (86.0 (35.5–181.3)), gemcitabine (53.0
(20.0–83.0)), paclitaxel (52.0 (28.5–77.5)), docetaxel (47.0 (18.8–78.3)),
bleomycin (92.0 (38.0–130.5)), oxaliplatin (45.0 (11.0–180.0)), nivolumab
(56.0 (21.0–135.0)), gefitinib (24.0 (11.0–55.0)), erlotinib (21.0
(9.0–49.0)), temsirolimus (38.0 (14.0–68.5)), everolimus (56.0 (35.0–90.0)),
osimertinib (51.5 (21.0–84.8)), alectinib (78.5 (44.3–145.8)), bicalutamide
(50.0 (28.0–147.0)), pegylated interferon-2α (140.0 (75.8–233.0)),
sai-rei-to (35.0 (20.0–54.5)), and sho-saiko-to (33.0 (13.5–74.0)) days.
Association rule mining suggested that the risk of drug-induced interstitial
lung disease was increased by a combination of amiodarone or sho-saiko-to
and aging. Conclusion: Our results showed that patients who receive gefitinib or erlotinib should be
closely monitored for the development of drug-induced interstitial lung
disease within a short duration (4 weeks). In addition, elderly people who
receive amiodarone or sho-saiko-to should be carefully monitored for the
development of drug-induced interstitial lung disease.
Collapse
Affiliation(s)
- Kiyoka Matsumoto
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan
| | - Satoshi Nakao
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan
| | - Shiori Hasegawa
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan.,Department of Pharmacy, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Toshinobu Matsui
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan.,Department of Pharmacy, Gifu Prefectural Tajimi Hospital, Tajimi, Japan
| | - Kazuyo Shimada
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan
| | - Ririka Mukai
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan
| | - Mizuki Tanaka
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan
| | - Hiroaki Uranishi
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan.,Department of Pharmacy, Nara Medical University Hospital, Kashihara, Japan
| | - Mitsuhiro Nakamura
- Laboratory of Drug Informatics, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
10
|
Distefano G, Fanzone L, Palermo M, Tiralongo F, Cosentino S, Inì C, Galioto F, Vancheri A, Torrisi SE, Mauro LA, Foti PV, Vancheri C, Palmucci S, Basile A. HRCT Patterns of Drug-Induced Interstitial Lung Diseases: A Review. Diagnostics (Basel) 2020; 10:244. [PMID: 32331402 PMCID: PMC7236658 DOI: 10.3390/diagnostics10040244] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Interstitial Lung Diseases (ILDs) represent a heterogeneous group of pathologies, which may be related to different causes. A low percentage of these lung diseases may be secondary to the administration of drugs or substances. Through the PubMed database, an extensive search was performed in the fields of drug toxicity and interstitial lung disease. We have evaluated the different classes of drugs associated with pulmonary toxicity. Several different high resolution computed tomography (HRCT) patterns related to pulmonary drug toxicity have been reported in literature, and the most frequent ILDs patterns reported include Nonspecific Interstitial Pneumonia (NSIP), Usual Interstitial Pneumonia (UIP), Hypersensitivity Pneumonitis (HP), Organizing Pneumonia (OP), Acute Respiratory Distress Syndrome (ARDS), and Diffuse Alveolar Damage (DAD). Finally, from the electronic database of our Institute we have selected and commented on some cases of drug-induced lung diseases related to the administration of common drugs. As the imaging patterns are rarely specific, an accurate evaluation of the clinical history is required and a multidisciplinary approach-involving pneumologists, cardiologists, radiologists, pathologists, and rheumatologists-is recommended.
Collapse
Affiliation(s)
- Giulio Distefano
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Luigi Fanzone
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Monica Palermo
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Francesco Tiralongo
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Salvatore Cosentino
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Corrado Inì
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Federica Galioto
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Ada Vancheri
- Department of Clinical and Experimental Medicine, University of Catania, Regional Referral Centre for Rare Lung Disease, 95123 Catania, Italy; (A.V.); (S.E.T.)
| | - Sebastiano E. Torrisi
- Department of Clinical and Experimental Medicine, University of Catania, Regional Referral Centre for Rare Lung Disease, 95123 Catania, Italy; (A.V.); (S.E.T.)
| | - Letizia A. Mauro
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Pietro V. Foti
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Carlo Vancheri
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Stefano Palmucci
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| | - Antonio Basile
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”-University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (L.F.); (M.P.); (F.T.); (S.C.); (C.I.); (F.G.); (L.A.M.); (P.V.F.); (C.V.); (S.P.); (A.B.)
| |
Collapse
|
11
|
Tamsulosin Associated with Interstitial Lung Damage in CYP2D6 Variant Alleles Carriers. Int J Mol Sci 2020; 21:ijms21082770. [PMID: 32316326 PMCID: PMC7215842 DOI: 10.3390/ijms21082770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/14/2020] [Indexed: 11/17/2022] Open
Abstract
Drugs are serious but underestimated causative agents of interstitial lung disease (ILD). Both cytotoxic and immune mechanisms may be involved in drug-induced ILD (DI-ILD). We aimed to investigate whether polymorphisms of relevant CYP enzymes involved in the metabolization of tamsulosin might explain the pathologic mechanism of the DI-ILD in the cases with suspected tamsulosin DI-ILD. We collected 22 tamsulosin-associated DI-ILD cases at two ILD Expertise Centers in the Netherlands between 2009 and 2020. CYP2D6, CYP2C9, CYP2C19, CYP3A4, and CYP3A5 single nucleotide polymorphisms were genotyped and compared with a control group of 78 healthy Caucasian male volunteers. Nine cases were phenotyped as CYP2D6 poor metabolizers and 13 as CYP2D6 intermediate metabolizers. The phenotypes of the cases differed significantly from those of the healthy controls, with more poor metabolizers. After withdrawal of tamsulosin, the pulmonary condition of three cases had improved, six patients had stabilized, and one patient stabilized after reducing the tamsulosin dose. The described 22 cases suggest that an association between the presence of CYP2D6 allelic variants and tamsulosin-associated ILD is highly likely. These cases highlight the importance of both clinical and genetic risk stratification aimed to achieve a more accurate prevention of DI-ILD in the future and enhance the quality of life of patients.
Collapse
|
12
|
Jessurun NT, Drent M, van Puijenbroek EP, Bekers O, Wijnen PA, Bast A. Drug-induced interstitial lung disease: role of pharmacogenetics in predicting cytotoxic mechanisms and risks of side effects. Curr Opin Pulm Med 2019; 25:468-477. [PMID: 31365381 DOI: 10.1097/mcp.0000000000000590] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The diagnosis of drug-induced interstitial lung disease (DI-ILD) is challenging and mainly made by exclusion of other possible causes. Toxicity can occur as a cause of drug(s) or drug-drug interactions. In this review, we summarize the possible role of pharmacogenetics of metabolizing enzymes in DI-ILD. RECENT FINDINGS Knowledge of the genetic predispositions of enzymes involved in drug metabolization and their relation with proposed cytotoxic mechanisms of DI-ILD, in particular direct cell toxicity and free oxygen radical production is increasing. The cytochrome P450 enzyme family and other enzymes play an important role in the metabolism of all sorts of ingested, injected, or inhaled xenobiotic substances. The liver is the major site for metabolism. Metabolic cytotoxic mechanisms have however also been detected in lung tissue. Polymorphisms in genes coding for enzymes that influence metabolic activity may lead to localized (toxic) reactions and tissue damage. This knowledge may be helpful in preventing the risk of DI-ILD. SUMMARY Drug toxicity can be the consequence of absence or very poor enzyme activity, especially if no other metabolic route is available. In the case of reduced enzyme activity, it is recommended to reduce the dose or to prescribe an alternative drug, which is metabolized by a different, unaffected enzyme system to prevent toxic side effects. However, enhanced enzyme activity may lead to excessive formation of toxic and sometimes reactive metabolites. Therefore, knowing a patient's drug-metabolizing profile before drug prescription is a promising way to prevent or explain DI-ILD.
Collapse
Affiliation(s)
- Naomi T Jessurun
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
- ILD Care Foundation Research Team, Ede, The Netherlands
| | - Marjolein Drent
- ILD Care Foundation Research Team, Ede, The Netherlands
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, The Netherlands
- ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Eugène P van Puijenbroek
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
- Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Otto Bekers
- ILD Care Foundation Research Team, Ede, The Netherlands
- Department of Clinical Chemistry, Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Petal A Wijnen
- ILD Care Foundation Research Team, Ede, The Netherlands
- Department of Clinical Chemistry, Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Aalt Bast
- ILD Care Foundation Research Team, Ede, The Netherlands
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, The Netherlands
- Venlo Campus, Maastricht University, Venlo, The Netherlands
| |
Collapse
|
13
|
Skeoch S, Weatherley N, Swift AJ, Oldroyd A, Johns C, Hayton C, Giollo A, Wild JM, Waterton JC, Buch M, Linton K, Bruce IN, Leonard C, Bianchi S, Chaudhuri N. Drug-Induced Interstitial Lung Disease: A Systematic Review. J Clin Med 2018; 7:E356. [PMID: 30326612 PMCID: PMC6209877 DOI: 10.3390/jcm7100356] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Drug-induced interstitial lung disease (DIILD) occurs as a result of numerous agents, but the risk often only becomes apparent after the marketing authorisation of such agents. METHODS In this PRISMA-compliant systematic review, we aimed to evaluate and synthesise the current literature on DIILD. RESULTS Following a quality assessment, 156 full-text papers describing more than 6000 DIILD cases were included in the review. However, the majority of the papers were of low or very low quality in relation to the review question (78%). Thus, it was not possible to perform a meta-analysis, and descriptive review was undertaken instead. DIILD incidence rates varied between 4.1 and 12.4 cases/million/year. DIILD accounted for 3⁻5% of prevalent ILD cases. Cancer drugs, followed by rheumatology drugs, amiodarone and antibiotics, were the most common causes of DIILD. The radiopathological phenotype of DIILD varied between and within agents, and no typical radiological pattern specific to DIILD was identified. Mortality rates of over 50% were reported in some studies. Severity at presentation was the most reliable predictor of mortality. Glucocorticoids (GCs) were commonly used to treat DIILD, but no prospective studies examined their effect on outcome. CONCLUSIONS Overall high-quality evidence in DIILD is lacking, and the current review will inform larger prospective studies to investigate the diagnosis and management of DIILD.
Collapse
Affiliation(s)
- Sarah Skeoch
- Arthritis Research UK Centre for Epidemiology, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9PL, UK.
- Royal National Hospital for Rheumatic Diseases, Royal United Hospitals Bath NHS Foundation Trust, Bath BA1 1RL, UK.
| | - Nicholas Weatherley
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK.
| | - Andrew J Swift
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK.
| | - Alexander Oldroyd
- Arthritis Research UK Centre for Epidemiology, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9PL, UK.
| | - Christopher Johns
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK.
| | - Conal Hayton
- North West Lung Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M6 8HD, UK.
| | - Alessandro Giollo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, NIHR Leeds Biomedical Research Centre, University of Leeds, Leeds LS2 9JT, UK.
- Rheumatology Unit, Department of Medicine, University of Verona, 37134 Verona, Italy.
| | - James M Wild
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK.
| | - John C Waterton
- Bioxydyn Limited, Rutherford House, Manchester Science Park, Manchester M15 6SZ, UK.
- Centre for Imaging Sciences, Division of Informatics Imaging & Data Sciences, School of Health Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9PL, UK.
| | - Maya Buch
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, NIHR Leeds Biomedical Research Centre, University of Leeds, Leeds LS2 9JT, UK.
| | - Kim Linton
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9PL, UK.
| | - Ian N Bruce
- Arthritis Research UK Centre for Epidemiology, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9PL, UK.
- The Kellgren Centre for Rheumatology, NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M6 8HD, UK.
| | - Colm Leonard
- North West Lung Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M6 8HD, UK.
| | - Stephen Bianchi
- Academic Directorate of Respiratory Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield S10 2JF, UK.
| | - Nazia Chaudhuri
- North West Lung Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M6 8HD, UK.
| |
Collapse
|
14
|
Ferreira PG, Costa S, Dias N, Ferreira AJ, Franco F. Simultaneous interstitial pneumonitis and cardiomyopathy induced by venlafaxine. J Bras Pneumol 2015; 40:313-8. [PMID: 25029655 PMCID: PMC4109204 DOI: 10.1590/s1806-37132014000300015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 08/28/2013] [Indexed: 12/17/2022] Open
Abstract
Venlafaxine is a serotonin-norepinephrine reuptake inhibitor used as an
antidepressant. Interindividual variability and herb-drug interactions can lead to
drug-induced toxicity. We report the case of a 35-year-old female patient diagnosed
with synchronous pneumonitis and acute cardiomyopathy attributed to venlafaxine. The
patient sought medical attention due to dyspnea and dry cough that started three
months after initiating treatment with venlafaxine for depression. The patient was
concomitantly taking Centella asiatica and Fucus
vesiculosus as phytotherapeutic agents. Chest CT angiography and chest
X-ray revealed parenchymal lung disease (diffuse micronodules and focal ground-glass
opacities) and simultaneous dilated cardiomyopathy. Ecocardiography revealed a left
ventricular ejection fraction (LVEF) of 21%. A thorough investigation was carried
out, including BAL, imaging studies, autoimmune testing, right heart catheterization,
and myocardial biopsy. After excluding other etiologies and applying the Naranjo
Adverse Drug Reaction Probability Scale, a diagnosis of synchronous
pneumonitis/cardiomyopathy associated with venlafaxine was assumed. The herbal
supplements taken by the patient have a known potential to inhibit cytochrome P450
enzyme complex, which is responsible for the metabolization of venlafaxine. After
venlafaxine discontinuation, there was rapid improvement, with regression of the
radiological abnormalities and normalization of the LVEF. This was an important case
of drug-induced cardiopulmonary toxicity. The circumstantial intake of inhibitors of
the CYP2D6 isoenzyme and the presence of a CYP2D6 slow metabolism phenotype might
have resulted in the toxic accumulation of venlafaxine and the subsequent clinical
manifestations. Here, we also discuss why macrophage-dominant phospholipidosis was
the most likely mechanism of toxicity in this case.
Collapse
Affiliation(s)
| | - Susana Costa
- Department of Cardiology, Coimbra Hospital, University Center, Coimbra, Portugal
| | - Nuno Dias
- Department of Anatomopathology, Coimbra Hospital, University Center, Coimbra, Portugal
| | | | - Fátima Franco
- Department of Pulmonology, Coimbra Hospital, University Center, Coimbra, Portugal
| |
Collapse
|
15
|
Essaadouni L, Benjilali L. [Drug induced interstitial lung disease in systemic diseases]. REVUE DE PNEUMOLOGIE CLINIQUE 2013; 69:307-314. [PMID: 24183295 DOI: 10.1016/j.pneumo.2013.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 06/28/2013] [Accepted: 08/04/2013] [Indexed: 06/02/2023]
Abstract
Immunosuppressants and immunomodulators are designed to regulate excessive immune response responsible for inflammatory lesions and are prescribed more and more in internal medicine. These drugs are known for their efficiency but with a significant toxicity including interstitial lung disease (ILD). Some factors liable to pulmonary toxicity include advanced age, genetic polymorphism and the existence of prior pulmonary disease. Cytotoxicity and hypersensitivity are the main mechanisms of pulmonary toxicity. There is no universal classification of drug induced-lung disease. Theoretically, drugs may be responsible for all histological aspects of ILD. Methotrexate is the most well-known drug as a provider of ILD with a prevalence of 0.3 to 11.6%. Some cases of ILD have also been reported with the new biologics used in systemic diseases. The diagnostic approach to the suspicion of drug ILD is to eliminate non-medicinal causes of pneumonia including infections and tumors before exploring the clinical symptomatology and the results of imaging and bronchoalveolar lavage cell profile. The analysis of the clinical symptomatology check the compatibility of the chronology of clinical and/or radiological pneumonia with the medication suspected. Subsequently, data from the clinical case are compared with those of the literature. Treatment involves stopping the suspected drug. The use of corticosteroids may be required in case of signs of severity or a lingering evolution.
Collapse
Affiliation(s)
- L Essaadouni
- Service de médecine interne, faculté de médecine et de pharmacie, université Cadi-Ayyad, CHU Mohammed VI, Marrakech, Maroc
| | - L Benjilali
- Service de médecine interne, faculté de médecine et de pharmacie, université Cadi-Ayyad, CHU Mohammed VI, Marrakech, Maroc.
| |
Collapse
|
16
|
Wijnen PA, Linssen CF, Haenen GR, Bekers O, Drent M. Variant VKORC1 and CYP2C9 Alleles in Patients with Diffuse Alveolar Hemorrhage Caused by Oral Anticoagulants. Mol Diagn Ther 2012; 14:23-30. [DOI: 10.1007/bf03256350] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
17
|
Schwaiblmair M, Behr W, Haeckel T, Märkl B, Foerg W, Berghaus T. Drug induced interstitial lung disease. Open Respir Med J 2012; 6:63-74. [PMID: 22896776 PMCID: PMC3415629 DOI: 10.2174/1874306401206010063] [Citation(s) in RCA: 200] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 07/06/2012] [Accepted: 07/06/2012] [Indexed: 01/15/2023] Open
Abstract
With an increasing number of therapeutic drugs, the list of drugs that is responsible for severe pulmonary disease also grows. Many drugs have been associated with pulmonary complications of various types, including interstitial inflammation and fibrosis, bronchospasm, pulmonary edema, and pleural effusions. Drug-induced interstitial lung disease (DILD) can be caused by chemotherapeutic agents, antibiotics, antiarrhythmic drugs, and immunosuppressive agents. There are no distinct physiologic, radiographic or pathologic patterns of DILD, and the diagnosis is usually made when a patient with interstitial lung disease (ILD) is exposed to a medication known to result in lung disease. Other causes of ILD must be excluded. Treatment is avoidance of further exposure and systemic corticosteroids in patients with progressive or disabling disease.
Collapse
Affiliation(s)
- Martin Schwaiblmair
- Department of Internal Medicine I, Klinikum Augsburg, Ludwig-Maximilians-University of Munich, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Matsuno O. Drug-induced interstitial lung disease: mechanisms and best diagnostic approaches. Respir Res 2012; 13:39. [PMID: 22651223 PMCID: PMC3426467 DOI: 10.1186/1465-9921-13-39] [Citation(s) in RCA: 203] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 05/16/2012] [Indexed: 02/06/2023] Open
Abstract
Drug-induced interstitial lung disease (DILD) is not uncommon and has many clinical patterns, ranging from benign infiltrates to life-threatening acute respiratory distress syndrome. There are two mechanisms involved in DILD, which are probably interdependent: one is direct, dose-dependent toxicity and the other is immune-mediated. Cytotoxic lung injury may result from direct injury to pneumocytes or the alveolar capillary endothelium. Drugs can induce all types of immunological reactions described by Gell and Coombs; however, most reactions in immune-mediated DILD may be T cell-mediated. DILD can be difficult to diagnose; diagnosis is often possible by exclusion alone. Identifying the causative drug that induces an allergy or cytotoxicity is essential for preventing secondary reactions. One method to confirm the diagnosis of a drug-induced disease is re-exposure or re-test of the drug. However, clinicians are reluctant to place patients at further risk of illness, particularly in cases with severe drug-induced diseases. Assessment of cell-mediated immunity has recently increased, because verifying the presence or absence of drug-sensitized lymphocytes can aid in confirmation of drug-induced disease. Using peripheral blood samples from drug-allergic patients, the drug-induced lymphocyte stimulation test (DLST) and the leukocyte migration test (LMT) can detect the presence of drug-sensitized T cells. However, these tests do not have a definite role in the diagnosis of DILD. This study explores the potential of these new tests and other similar tests in the diagnosis of DILD and provides a review of the relevant literature on this topic.
Collapse
Affiliation(s)
- Osamu Matsuno
- Division of Medicine for Allergic Disease, Osaka Prefectural Medical Center for Respiratory and Allergic Diseases, Habikino City, Japan.
| |
Collapse
|
19
|
Development of cocaine-induced interstitial lung damage in two CYP2C and VKORC1 variant allele carriers. Mol Diagn Ther 2011; 15:177-80. [PMID: 21766908 DOI: 10.1007/bf03256408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Often, the connection between drug use and the development of related inflammatory damage or idiosyncratic toxicities is hard to recognize and objectify. The presence of cytochrome P450 (CYP) variant genotypes appears to be a substantial susceptibility risk factor in the development of drug-induced pulmonary adverse events. We hypothesized that the presence of variant alleles may be associated with serious complications of illicit drug use. CASE REPORT We report the cases of two cocaine users who developed a 'flu-like' syndrome with diffuse interstitial infiltrates after cocaine abuse. Genotyping for CYP (CYP2C9, CYP2C19) and vitamin K epoxide reductase complex 1 (VKORC1) allelic variants (-1639G/A and 1173C/T) was performed in these two patients. Both cases were heterozygous for VKORC1 variant alleles, and both possessed a CYP2C polymorphism (case 1: CYP2C19*1/*2; case 2: CYP2C9*1/*3). CONCLUSIONS The described drug abuse cases suggest that an association between the presence of CYP2C and VKORC1 allelic variants and cocaine-induced interstitial lung damage is highly likely. It is assumed that these polymorphisms contribute to intra-individual variability in drug response and toxicity, including cocaine response and toxicity. Moreover, the importance of including pharmacogenomics in the work-up of patients with suspected drug-induced (lung) toxicity, such as alveolar hemorrhage, is highlighted by these cases.
Collapse
|
20
|
Schwaiblmair M, Behr W, Foerg W, Berghaus T. Cytochrome P450 polymorphisms and drug-induced interstitial lung disease. Expert Opin Drug Metab Toxicol 2011; 7:1547-60. [DOI: 10.1517/17425255.2011.629185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
21
|
Nyberg F, Barratt BJ, Mushiroda T, Takahashi A, Jawaid A, Hada S, Umemura T, Fukuoka M, Nakata K, Ohe Y, Kato H, Kudoh S, March R, Nakamura Y, Kamatani N. Interstitial lung disease in gefitinib-treated Japanese patients with non-small-cell lung cancer: genome-wide analysis of genetic data. Pharmacogenomics 2011; 12:965-75. [DOI: 10.2217/pgs.11.38] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aim: To investigate potential relationships between SNPs and acute interstitial lung disease (ILD) events in Japanese non-small-cell lung cancer patients receiving gefitinib. Materials & methods: Japanese non-small-cell lung cancer patients treated with gefitinib from a prospective pharmacoepidemiological cohort with a nested case–control study component (‘CCS’; 52 ILD cases, 139 controls) and a retrospective study (28 ILD cases, 55 controls) were genotyped for nearly 500,000 SNPs. Associations between genotype and ILD were evaluated using Fisher’s exact test and logistic regression modeling, and false discovery rate analysis was used to adjust for the large number of statistical tests. Results: The CCS data provided some false discovery rate evidence that the significance of top-ranking SNPs exceeded levels expected by chance, suggesting some genuine associations. However, replication analyses using retrospective study data were not supportive and there was little evidence of strong genetic associations from a combined analysis. Adjustment of CCS analyses for clinical variables provided little additional convincing evidence. Significant gene–gene interactions between SNP pairs using CCS data were not confirmed in retrospective study replication analyses. Conclusion: Although it is not possible to exclude genetic influences in ILD etiology, common sequence variation is unlikely to explain a major component of ILD risk. Our top results may provide a useful hypothesis-generating starting point for further research. Presented, in part, at the 26th ICPE: International Conference on Pharmacoepidemiology & Therapeutic Risk Management, Brighton, UK, 19–22 August 2010. Original submitted 1 December 2010; Revision submitted 22 February 2011
Collapse
Affiliation(s)
- Fredrik Nyberg
- Global Epidemiology, AstraZeneca R&D, Mölndal, Sweden and Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Bryan J Barratt
- Research & Development Genetics, Personalised Healthcare & Biomarkers, AstraZeneca, Macclesfield, UK
| | - Taisei Mushiroda
- Research Group for Pharmacogenomics, Center for Genomic Medicine, The Institute of Physical & Chemical Research (RIKEN), Yokohama, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, Center for Genomic Medicine, The Institute of Physical & Chemical Research (RIKEN), Yokohama, Japan
| | - Ansar Jawaid
- Research & Development Genetics, Personalised Healthcare & Biomarkers, AstraZeneca, Macclesfield, UK
| | - Shuji Hada
- Clinical Development, AstraZeneca KK, Osaka, Japan
| | - Takaaki Umemura
- Patient Safety, Research & Development, AstraZeneca KK, Osaka, Japan
| | - Masahiro Fukuoka
- Department of Medical Oncology, Kinki University School of Medicine, Osaka, Japan
| | | | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Harubumi Kato
- Department of Surgery, Tokyo Medical University, Tokyo, Japan and Niizashiki Central General Hospital, Saitama, Japan
| | - Shoji Kudoh
- Japan Anti-Tuberculosis Association, Fukujuji Hospital, Tokyo, Japan and Division of Pulmonary Medicine, Infectious Diseases & Oncology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Ruth March
- Personalised Healthcare & Biomarkers, AstraZeneca, Macclesfield, UK
| | - Yusuke Nakamura
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Naoyuki Kamatani
- Laboratory for Statistical Analysis, Center for Genomic Medicine, The Institute of Physical & Chemical Research (RIKEN), Yokohama, Japan
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW To review the role of microorganisms in interstitial lung disease (ILD) and to emphasize their importance in initiation and course of ILD. RECENT FINDINGS ILD can be idiopathic but often causality such as drugs or connective tissue disease can be found. Multiple microorganisms have been associated with ILD. On the one hand, pulmonary infection can cause extensive pulmonary damage with patterns of an ILD. On the other hand, microorganisms can trigger the immune system and provoke an abnormal response- not directed against the causative pathogen- that may result in ILD. Moreover, patients with ILD often are susceptible to infection, and infections can importantly influence the course of ILD. Furthermore, not only an infection but also its treatment can result in a drug-induced pneumonitis, eventually resulting in long-term lung damage. SUMMARY Microorganisms can initiate and/or influence the course of ILD. Early recognition, adequate diagnostic evaluation and therapy are essential to prevent permanent damage. Prevention of infection in patients with established ILD is strongly recommended.
Collapse
|
23
|
Relationship between drug-induced interstitial lung diseases and cytochrome P450 polymorphisms. Curr Opin Pulm Med 2010; 16:496-502. [PMID: 20592596 DOI: 10.1097/mcp.0b013e32833c06f1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Interstitial lung disease and especially drug-induced interstitial lung disease can occur as a cause of drug(s) or drug-drug interactions. In this review we summarize the possible role of cytochrome P450 (CYP) enzymes in drug-induced interstitial lung disease. RECENT FINDINGS The CYP enzyme family plays an important role in the metabolism of all sorts of ingested, injected or inhaled xenobiotic substances. Although the liver is considered to be the major metabolism site of CYP enzymes, in recent years more CYP isoforms have been detected in lung tissue. Polymorphisms in these CYP genes can influence the metabolic activity of the subsequent enzymes, which in turn may lead to localized (toxic) reactions and tissue damage. SUMMARY Drug toxicity can be the consequence of no or very poor enzyme activity, especially if no other metabolic route is available. In the case of reduced enzyme activity, dose reduction or prescribing an alternative drug metabolized by a different, unaffected CYP enzyme is recommended to prevent toxic side effects. Therefore, knowing a patient's CYP profile before drug prescription could be a way to prevent drug-induced interstitial lung disease. Moreover, it might be helpful in explaining serious adverse effects from inhaled, injected or ingested xenobiotic substances.
Collapse
|
24
|
|
25
|
Lind JSW, Dingemans AMC, Groen HJM, Thunnissen FB, Bekers O, Heideman DAM, Honeywell RJ, Giovannetti E, Peters GJ, Postmus PE, van Suylen RJ, Smit EF. A multicenter phase II study of erlotinib and sorafenib in chemotherapy-naive patients with advanced non-small cell lung cancer. Clin Cancer Res 2010; 16:3078-87. [PMID: 20395213 DOI: 10.1158/1078-0432.ccr-09-3033] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE This multicenter, phase II study evaluates the efficacy and safety of erlotinib, an epidermal growth factor receptor (EGFR) inhibitor, plus sorafenib, a multityrosine kinase inhibitor against vascular endothelial growth factor receptors, in patients with previously untreated advanced non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN Chemotherapy-naïve patients with stage IIIB/IV NSCLC received erlotinib (150 mg once a day) and sorafenib (400 mg twice a day) until disease progression or unacceptable toxicity. The primary end point was the rate of nonprogression at 6 weeks. Secondary end points included objective response rate (ORR), time to progression, overall survival, and adverse events. Exploratory end points included pretreatment EGFR and KRAS mutation status, pharmacokinetics, and cytochrome P450 polymorphisms. RESULTS Fifty patients initiated therapy. The nonprogression rate at 6 weeks was 74%: 12 (24%) partial response and 25 (50%) stable disease. Ultimately, the ORR was 28%. Median time to progression was 5.0 months [95% confidence interval (95% CI), 3.2-6.8 months]. Median overall survival was 10.9 months (95% CI, 3.8-18.1 months). Grade 3/4 adverse events included fatigue (16%), hand-foot skin reaction (16%), rash (16%), diarrhea (14%), and hypophosphatemia (42%). There was one treatment-related fatal pulmonary hemorrhage. Patients with wild-type EGFR had a higher ORR (19%) than previously reported for single-agent erlotinib/sorafenib. Erlotinib levels were lowered. This was associated with CYP3A4 polymorphism and was possibly due to sorafenib. CONCLUSION Despite a possible drug interaction, sorafenib plus erlotinib has promising clinical activity in patients with stage IIIB/IV NSCLC and has an acceptable safety profile. Further evaluation of this combination as potential salvage therapy in EGFR mutation-negative patients and the possible drug interaction is warranted.
Collapse
Affiliation(s)
- Joline S W Lind
- Departments of Pulmonary Diseases, VU University Medical Center Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wijnen PAHM, Limantoro I, Drent M, Bekers O, Kuijpers PMJC, Koek GH. Depressive effect of an antidepressant: therapeutic failure of venlafaxine in a case lacking CYP2D6 activity. Ann Clin Biochem 2009; 46:527-30. [PMID: 19822698 DOI: 10.1258/acb.2009.009003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Understanding the mechanisms of drug metabolism and interactions can help to prevent side-effects. Not only drug interactions, environmental factors, disease processes and ageing are factors in the inter-individual metabolic capacity variance but also genetic factors probably play an important role, as is illustrated in the case presented. Besides therapeutic drug monitoring, genotyping some important cytochrome P450 (CYP450) enzymes was of additional value in explaining why the patient developed severe adverse effects and, moreover, did not experience any therapeutical effect of venlafaxine. Results indicated that the patient was a poor metabolizer for CYP2D6, the most important phase I enzyme to metabolize venlafaxine. This corroborates that polymorphisms in the CYP450 gene influence the metabolic activity of the corresponding enzymes, thus affecting the subsequent serum drug levels and their metabolites. This case highlights the potential benefit of both clinical and genetic risk stratification (pharmacogenetics) prior to treatment, either for setting the individual dose or for making a decision about using a particular drug.
Collapse
Affiliation(s)
- P A H M Wijnen
- Department of Clinical Chemistry, ILD Care Center, Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
27
|
Wijnen PAHM, Drent M, van Dieijen-Visser MP, Bekers O. Pharmacogenetic testing after a simple DNA isolation method on buccal swab samples. Pharmacogenomics 2009; 10:983-7. [PMID: 19530965 DOI: 10.2217/pgs.09.4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To evaluate whether the quality and quantity of DNA isolated from noninvasively obtained buccal swab (BS) samples, using the previously described isolation method for dried blood spot (DBS) samples was satisfactory. MATERIALS & METHODS From 25 healthy volunteers, DBS samples were obtained by the capillary finger prick method and BS samples were obtained by rubbing a sterile, dry cotton swab against the inside of their cheek. Thereafter, DNA was isolated. In addition, the quantity of the obtained DNA was measured and melting curve analyses for both sampling methods were performed to establish the quality of the obtained DNA from both the DBS and BS samples. RESULTS The derivative melting curves of the DNA samples obtained from the capillary blood and BS were comparable and highly reproducible. The mean DNA concentrations measured were 16.0 ng/microl (12.6-19.4 ng/microl) and 70.2 ng/microl (57.3-83.1 ng/microl), respectively, for the DBS and BS samples (p < 0.001). CONCLUSION The DBS DNA isolation method appeared to be extremely useful to discriminate between genotypes. This expands the possibilities of this quick and easy DNA isolation procedure. In particular, the noninvasive BS sampling method appeared to be a good alternative to invasive sampling methods.
Collapse
Affiliation(s)
- Petal A H M Wijnen
- Maastricht University Medical Centre, PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | | | | | | |
Collapse
|
28
|
Current awareness: Pharmacoepidemiology and drug safety. Pharmacoepidemiol Drug Saf 2009. [DOI: 10.1002/pds.1650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
29
|
Bijl MJ, van Schaik RHN. Research Highlights. Per Med 2009. [DOI: 10.2217/pme.09.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Monique J Bijl
- Department of Hospital Pharmacy, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron HN van Schaik
- Department of Clinical Chemistry, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
30
|
Journal Watch. Pharmaceut Med 2008. [DOI: 10.1007/bf03256737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|