Skip to main content
SAGE Open Medicine logoLink to SAGE Open Medicine
. 2020 May 6;8:2050312120918264. doi: 10.1177/2050312120918264

Analysis of drug-induced interstitial lung disease using the Japanese Adverse Drug Event Report database

Kiyoka Matsumoto 1, Satoshi Nakao 1, Shiori Hasegawa 1,2, Toshinobu Matsui 1,3, Kazuyo Shimada 1, Ririka Mukai 1, Mizuki Tanaka 1, Hiroaki Uranishi 1,4, Mitsuhiro Nakamura 1,
PMCID: PMC7262990  PMID: 32528682

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.

Keywords: Drug-induced interstitial lung disease, the Japanese Adverse Drug Event Report database, pharmacovigilance, time-to-onset profile

Introduction

Interstitial lung disease is a group of diffuse parenchymal lung disorders associated with substantial morbidity and mortality.1 Drug-induced interstitial lung disease (DIILD) occurs when drug exposure causes inflammation and eventually fibrosis of the lung interstitium.2 Chemotherapeutic drugs (e.g. bleomycin and gefitinib), amiodarone, anti-inflammatory drugs (e.g. methotrexate), biological drugs, and various other drugs can cause DIILD (www.pneumotox.com).2,3 As DIILD is considered a serious adverse event (AE) and represents a serious clinical problem, all healthcare professionals should be aware of a potential DIILD as soon as possible. Early intervention may prevent the progression of AEs and permanent changes.4 However, the detailed time-to-onset profiles of DIILD in clinical settings are not clear.

The frequency of DIILD is reported to be higher in Japan than that in other countries.5 Lung injuries related to molecular-targeted drugs have been reported. Reports related to gefitinib first occurred in 2002 in Japan and those related to the antirheumatic drug leflunomide were reported 1 year later.5 The Ministry of Health, Labor and Welfare in Japan has issued the Manual for Handling Disorders due to Adverse Drug Reactions with a focus on DIILD. AEs during the post-marketing phase in Japan are reported and managed by the Pharmaceuticals and Medical Devices Agency (PMDA). The agency has established a spontaneous reporting system (SRS) for the Japanese Adverse Drug Event Report (JADER) database. The JADER is the largest database in Japan and reflects the realities of clinical practices.

The aim of this retrospective pharmacovigilance study was to assess the incidence of DIILD by using the JADER database. We focused on the time-to-onset profile of DIILD. Furthermore, association rule mining has been proposed as a new analytical technique to identify undetected relationships such as possible risk factors between variables in the SRS database.6,7 We evaluated potential association rules between DIILD and demographics.

Materials and methods

Data source

Healthcare professionals, marketing approval holders, patients, and consumers voluntarily send AE reports to the PMDA. All AE report data were accumulated in the PMDA and were fully anonymized by the PMDA to form the JADER database. JADER data from April 2004 to June 2018 are publicly available and can be downloaded from the PMDA website (www.pmda.go.jp). For this retrospective study, we built a relational database, which integrated the data tables, by using the FileMaker Pro 13 software.

Definition of interstitial lung disease

In accordance with the terminology preferred by the Medical Dictionary for Regulatory Activities (MedDRA, www.pmrj.jp/jmo/php/indexj.php) version 19.0, we used the following preferred term (PT) for DIILD: interstitial lung disease (PT code: 10022611).

Drug selection

The number of drugs known to produce various patterns of DIILD is increasing. In this study, we first listed 82 drugs, each of which had more than 100 reported DIILD cases in the JADER database. Second, from the Drug-Induced Respiratory Disease Website (www.pneumotox.com), we listed 598 drugs from the website in the categories of interstitial/parenchymal lung disease, pulmonary edema—acute lung injury—ARDS, and pathology. From these categories, the following patterns were identified: “Interstitial/parenchymal lung disease: pneumonitis (ILD), acute, severe (may occasion an ARDS picture)” (pattern Ia, 155 listed drugs); “Interstitial/parenchymal lung disease: pneumonitis (ILD)” (pattern Ib, 329 listed drugs); “Interstitial/parenchymal lung disease: eosinophilic pneumonia (pulmonary infiltrates and eosinophilia)” (pattern Ic, 192 listed drugs); “Interstitial/parenchymal lung disease: pulmonary fibrosis (not otherwise specified)” (pattern Ig, 84 listed drugs); “Pulmonary edema—acute lung injury—ARDS” (pattern IIb, 254 listed drugs); “Pathology: cellular NSIP pattern” (pattern XVa, 51 listed drugs); “Pathology: organizing pneumonia (OP/BOOP) pattern” (pattern XVc, 70 listed drugs). Third, we compared the 598 listed drugs from the Drug-Induced Respiratory Disease Website (www.pneumotox.com) and the drugs in the JADER database with between 50 and 99 reported DIILD cases. Fourth, we listed the 18 drugs that matched the drugs in the Drug-Induced Respiratory Disease Website. Fifth, regardless of the number of reported DIILD cases related to each drug, we compared the drugs that were reported in the JADER database and drugs reported in previous studies.2,8 Ten drugs (sirolimus, simvastatin, fluvastatin, daptomycin, lapatinib, interferon beta, interferon gamma, pravastatin, pitavastatin, and ipilimumab) that were not listed by the fourth procedure were added. In total, we identified 110 (82 + 18 + 10) drugs for analysis (Table 1). Thus, Table 1 is considered to include almost all drugs that can be practically analyzed.

Table 1.

Number of reports and ROR for drug-induced interstitial lung disease.

Category ATC codea Drugs Total (n) Case (n) Non-case (n) RORb (95% CI)
Total 534688 24123
H2-receptor antagonists A02BA03 Famotidine 3469 172 3297 1.1 (0.9–1.3)
Proton pump inhibitors A02BC03 Lansoprazole 4434 240 4194 1.2 (1.1–1.4)
Aminosalicylic acid and similar agents A07EC01 Salazosulfapyridine 1864 108 1756 1.3 (1.1–1.6)
A07EC02 Mesalazine 1558 133 1425 2.0 (1.7–2.4)
Dipeptidyl peptidase 4 (DPP-4) inhibitors A10BH01 Sitagliptin 2054 148 1906 1.6 (1.4–1.9)
A10BH02 Vildagliptin 2371 74 2297 0.7 (0.5−0.9)
Platelet aggregation inhibitors B01AC04 Clopidogrel 4638 229 4409 1.1 (0.9–1.3)
B01AC05 Ticlopidine 2180 57 2123 0.6 (0.4−0.7)
B01AC23 Cilostazol 2244 107 2137 1.1 (0.9−1.3)
Direct thrombin inhibitors B01AE07 Dabigatran 2466 92 2374 0.8 (0.7−1.0)
Direct factor Xa inhibitors B01AF01 Rivaroxaban 4691 165 4526 0.8 (0.7–0.9)
B01AF02 Apixaban 4800 133 4667 0.6 (0.5–0.7)
Antiarrhythmics, class III C01BD01 Amiodarone 1993 665 1328 10.9 (9.9–11.9)
Dihydropyridine derivatives C08CA01 Amlodipine 3672 100 3572 0.6 (0.5−0.7)
Phenylalkylamine derivatives C08EA02 Bepridil 734 133 601 4.7 (3.9–5.7)
Angiotensin II receptor blockers (ARBs), plain C09CA03 Valsartan 3548 131 3417 0.8 (0.7–1.0)
C09CA06 Candesartan 1925 121 1804 1.4 (1.2–1.7)
HMG CoA reductase inhibitors C10AA01 Simvastatin 318 14 304 1.0 (0.6–1.7)
C10AA03 Pravastatin 922 40 882 1.0 (0.7–1.3)
C10AA04 Fluvastatin 638 19 619 0.6 (0.4–1.0)
C10AA05 Atorvastatin 2748 104 2644 0.8 (0.7–1.0)
C10AA07 Rosuvastatin 1517 71 1446 1.0 (0.8–1.3)
C10AA08 Pitavastatin 813 41 772 1.1 (0.8–1.5)
Glucocorticoids H02AB02 Dexamethasone 5952 144 5808 0.5 (0.4–0.6)
Tetracyclines J01AA08 Minocycline 1637 152 1485 2.2 (1.8–2.6)
Carbapenems J01DH02 Meropenem 1940 103 1837 1.2 (0.97−1.4)
Combinations of sulfonamides and trimethoprim, incl. derivatives J01EE01 Sulfamethoxazole • Trimethoprim 2737 104 2633 0.8 (0.7−1.0)
Macrolides J01FA09 Clarithromycin 4066 101 3965 0.5 (0.4−0.7)
Fluoroquinolones J01MA12 Levofloxacin 4187 196 3991 1.0 (0.9–1.2)
Other antibacterials J01XX09 Daptomycin 353 20 333 1.3 (0.8–2.0)
Antibiotics J04AB02 Rifampicin 1600 76 1524 1.1 (0.8−1.3)
Other drugs for treatment of tuberculosis J04AK02 Ethambutol 1253 50 1203 0.9 (0.7−1.2)
Antivirals for treatment of HCV infections J05AP01 Ribavirin 10394 319 10075 0.7 (0.6–0.7)
Nitrogen mustard analogues L01AA01 Cyclophosphamide 5129 390 4739 1.8 (1.6–1.9)
Folic acid analogues L01BA01 Methotrexate 18336 1899 16437 2.6 (2.4–2.7)
L01BA04 Pemetrexed 2431 347 2084 3.6 (3.2–4.0)
Pyrimidine analogues L01BC02 Fluorouracil 7796 801 6995 2.5 (2.3–2.7)
L01BC05 Gemcitabine 4454 1161 3293 7.8 (7.3–8.3)
L01BC06 Capecitabine 3561 209 3352 1.3 (1.1-1.5)
L01BC53 Tegafur U+002E; Uracil 1635 108 1527 1.5 (1.2–1.8)
L01BC53 Tegafur U+002E; Gimeracil U+002E; Oteracil 6618 639 5979 2.3 (2.1–2.5)
Vinca alkaloids and analogues L01CA02 Vincristine 2939 145 2794 1.1 (0.9–1.3)
L01CA04 Vinorelbine 758 175 583 6.4 (5.4–7.6)
Podophyllotoxin derivatives L01CB01 Etoposide 3017 123 2894 0.9 (0.7–1.1)
Taxanes L01CD01 Paclitaxel 6900 944 5956 3.5 (3.2–3.7)
L01CD02 Docetaxel 6403 1066 5337 4.4 (4.1–4.7)
Anthracyclines and related substances L01DB01 Doxorubicin 3804 186 3618 1.1 (0.9–1.3)
L01DB03 Epirubicin 1547 138 1409 2.1 (1.7–2.5)
L01DB10 Amrubicin 1245 116 1129 2.2 (1.8–2.6)
Other cytotoxic antibiotics L01DC01 Bleomycin 418 104 314 7.0 (5.6–8.8)
Platinum compounds L01XA01 Cisplatin 8673 260 8413 0.7 (0.6–0.7)
L01XA02 Carboplatin 5281 332 4949 1.4 (1.3–1.6)
L01XA03 Oxaliplatin 8001 682 7319 2.0 (1.8–2.2)
Monoclonal antibodies L01XC02 Rituximab 3979 209 3770 1.2 (1.0–1.4)
L01XC03 Trastuzumab 2469 380 2089 3.9 (3.5–4.3)
L01XC06 Cetuximab 2746 451 2295 4.2 (3.8–4.7)
L01XC07 Bevacizumab 9440 505 8935 1.2 (1.1–1.3)
L01XC08 Panitumumab 1393 302 1091 5.9 (5.2–6.7)
L01XC11 Ipilimumab 545 41 504 1.7 (1.3–2.4)
L01XC13 Pertuzumab 683 122 561 4.6 (3.8–5.6)
L01XC17 Nivolumab 4419 991 3428 6.3 (5.9–6.8)
L01XC18 Pembrolizumab 2148 622 1526 8.8 (8.0–9.7)
L01XC21 Ramucirumab 1570 91 1479 1.3 (1.1−1.6)
Protein kinase inhibitors L01XE01 Imatinib 4399 348 4051 1.8 (1.6–2.0)
L01XE02 Gefitinib 2736 1217 1519 17.8 (16.5–19.2)
L01XE03 Erlotinib 2748 836 1912 9.6 (8.8–10.4)
L01XE04 Sunitinib 3320 106 3214 0.7 (0.6–0.8)
L01XE05 Sorafenib 4922 136 4786 0.6 (0.5–0.7)
L01XE06 Dasatinib 1256 85 1171 1.5 (1.2–1.9)
L01XE07 Lapatinib 731 36 695 1.1 (0.8–1.5)
L01XE09 Temsirolimus 652 300 352 18.3 (15.6–21.3)
L01XE10 Everolimus 3671 1093 2578 9.4 (8.7–10.0)
L01XE13 Afatinib 786 178 608 6.2 (5.3–7.4)
L01XE16 Crizotinib 1027 147 880 3.6 (3.0–4.2)
L01XE35 Osimertinib 653 241 412 12.5 (10.7-14.7)
L01XE36 Alectinib 243 81 162 10.6 (8.1–13.9)
Other antineoplastic agents L01XX19 Irinotecan 5545 650 4895 2.9 (2.6–3.1)
L01XX32 Bortezomib 2219 153 2066 1.6 (1.3–1.9)
L01XX33 Celecoxib 3222 110 3112 0.7 (0.6–0.9)
L01XX41 Eribulin 840 104 736 3.0 (2.4−3.7)
Gonadotropin releasing hormone analogues L02AE02 Leuprorelin 1436 229 1207 4.0 (3.5–4.7)
Anti-androgens L02BB03 Bicalutamide 849 255 594 9.2 (7.9–10.6)
Colony stimulating factors L03AA02 Filgrastim 635 117 518 4.8 (3.9−5.9)
Interferons L03AB02 Interferon beta 1555 38 1517 0.5 (0.4–0.7)
L03AB03 Interferon gamma 32 6 26 4.9 (2.0–11.9)
L03AB11 PEG INF-2α 3386 305 3081 2.1 (1.9–2.4)
Selective immunosuppressants L04AA10 Sirolimus 44 5 39 2.7 (1.1–6.9)
L04AA13 Leflunomide 630 55 575 2.0 (1.5–2.7)
L04AA24 Abatacept 1186 84 1102 1.6 (1.3−2.0)
L04AA29 Tofacitinib 981 74 907 1.7 (1.4–2.2)
Tumor necrosis factor alpha (TNF-α) inhibitors L04AB01 Etanercept 4050 402 3648 2.4 (2.1–2.6)
L04AB02 Infliximab 4605 347 4258 1.7 (1.6–1.9)
L04AB04 Adalimumab 2452 227 2225 2.2 (1.9–2.5)
L04AB05 Certolizumab Pegol 863 66 797 1.8 (1.4−2.3)
L04AB06 Golimumab 1047 86 961 1.9 (1.5−2.4)
Interleukin inhibitors L04AC07 Tocilizumab 4187 209 3978 1.1 (0.97–1.3)
Calcineurin inhibitors L04AD01 Ciclosporin 6602 121 6481 0.4 (0.3–0.5)
L04AD02 Tacrolimus 10478 268 10210 0.6 (0.5–0.6)
Other immunosuppressants L04AX04 Lenalidomide 4247 99 4148 0.5 (0.4−0.6)
Acetic acid derivatives and related substances M01AB05 Diclofenac 3552 106 3446 0.6 (0.5−0.7)
Propionic acid derivatives M01AE Loxoprofen 6372 304 6068 1.1 (0.9–1.2)
Penicillamine and similar agents M01CC02 Bucillamine 1095 251 844 6.4 (5.5–7.3)
Preparations inhibiting uric acid production M04AA01 Allopurinol 3202 142 3060 1.0 (0.8–1.2)
Salicylic acid and derivatives N02BA01 Aspirin (acetylsalicylic acid) 7477 109 7368 0.3 (0.3−0.4)
Carboxamide derivatives N03AF01 Carbamazepine 5568 76 5492 0.3 (0.2−0.4)
Other antiepileptics N03AX16 Pregabalin 4659 158 4501 0.7 (0.6–0.9)
Detoxifying agents for antineoplastic treatment V03AF04 Levofolinate 3989 410 3579 2.4 (2.2–2.7)
Herbal Medicines Sai-rei-to 325 132 193 14.5 (11.7–18.2)
Sho-saiko-to 152 66 86 16.3 (11.8–22.4)
Others Iguratimod 487 86 401 4.6 (3.6−5.7)

ROR: reporting odds ratio; CI: confidence interval; HCV: Hepatitis C Virus.

a

Anatomical therapeutic classification.

b

Reporting odds ratio.

Statistics

Reporting odds ratio

The reporting odds ratio (ROR) is the authorized pharmacovigilance index and was calculated using two-by-two contingency tables of the presence or absence of a particular drug and a particular AE in the case reports.9 An association was considered disproportionate when the lower limit of the 95% confidence interval (CI) was >1 (Figure 1).9,10 Two or more cases were required to define the signal.11

Figure 1.

Figure 1.

Two-by-two contingency table for analysis.

Time to onset

Time-to-onset duration was calculated from the time of the patient’s first prescription to the occurrence of the AEs.7,12 It is necessary to take the correct truncation into account when estimating the time to onset of AEs from SRS data. We chose an analysis period of 730 days after the start date of administration to focus attention on the onset of AEs within 2 years. The median duration, quartiles, and the Weibull shape parameters (WSPs) were used to evaluate the time-to-onset data.7,12 The scale parameter, α, of the Weibull distribution determines the scale of the distribution function. A larger scale value stretches the distribution, whereas a smaller scale value shrinks the data distribution. The shape parameter, β, of the Weibull distribution determines the shape of the distribution function. A larger shape value produces a left-skewed curve, whereas a smaller shape value produces a right-skewed curve. In the analysis of the SRS, the shape parameter of the Weibull distribution was used to indicate hazards without a reference population as follows: when β was equal to 1, the hazard was estimated to be constant over time; if β was greater than 1 and the 95% confidence interval (CI) of β excluded the value 1, the hazard was considered to increase over time (wear-out failure type); finally, if β was less than 1 and the 95% CI of β excluded the value 1, the hazard was considered to decrease over time (initial-failure type).7,1317 Data analyses were performed by using JMP, version 12.0.1 (SAS Institute Inc., Cary, NC, USA).

Association rule mining

Association rule mining has been proposed as an analytical approach for discovering interesting relationships among the possible risk factors and variables in the SRS database. The method is focused on finding frequent co-existing associations among a collection of items.6,7 Given a set of transactions T (each transaction is a set of items), an association rule can be expressed as X (the antecedent (left-hand-side, lhs) of the rule) → Y (the consequent (right-hand-side, rhs) of the rule), where X and Y are mutually exclusive sets of items.6,7 The Apriori algorithm was applied to find association rules. Support, confidence, and lift were used as indicators to decide the relative strength of the rules. These indices were calculated as follows:

Support=NumberoftransactionswithbothXandY/Totalnumberoftransactions={XY}/{D}=P(XY)
Confidence=NumberoftransactionswithbothXandY/TotalnumberoftransactionswithX=P(XY)/P(X)

Confidence corresponds to the conditional probability P (Y|X) and Confidence measures the reliability of the interf Confidence erence made by a rule.

Expectedconfidence=NumberoftransactionswithY/Totalnumberoftransactions=P(B)
Lift=Confidence/ExpectedConfidence=P(XY)/P(X)P(Y)

Lift is the factor by which the co-occurrence of X and Y exceeds the expected probability of X and Y co-occurring, had they been independent. Lift is the ratio between the confidence of the rule and the support of the itemset as a consequence of the rule. The lift can be expressed as the confidence divided by P (Y). The lift can be evaluated as follows: lift = 1, if X and Y are independent; lift > 1, if X and Y are positively correlated; lift < 1, if X and Y are negatively correlated. Furthermore, we calculated the chi-square values to evaluate the association rules18

Chi-squared=D(lift1)2Support*Confidence(ConfidenceSupport)*(LiftConfidence)

Association rule mining was performed using the apriori function of the arules library in the arules package of the R software (version 3.3.3). Support and lift were visualized using the R-extension package arulesViz which implements novel visualization techniques to explore association rules.

Results

The JADER database contained 534,688 reports. The number of AE reports corresponding to DIILD was 24,123 reports (Table 1). The number of AEs associated with the top 10 reported drugs, methotrexate, gefitinib, gemcitabine, everolimus, docetaxel, nivolumab, paclitaxel, erlotinib, fluorouracil, and oxaliplatin was 1899, 1217, 1161, 1093, 1066, 991, 944, 836, 801, and 682, respectively. The top 10 RORs (95% CIs) with drugs, 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. In contrast, the ROR signals of HMG CoA reductase and antithrombotic agents such as platelet aggregation inhibitors, direct thrombin inhibitors, and direct factor Xa inhibitors were not detected.

For the time-to-onset analysis, we extracted combinations that had complete information for the date of treatment initiation and the date of AE onset. The median durations (day) (interquartile range) for DIILD 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)), PEG IFN-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, respectively (Figure 2). Among the drugs which demonstrated the lower limit of the 95% CI of the ROR was >1, >50% of the DIILD cases associated with minocycline, amrubicin, carboplatin, gefitinib, erlotinib, dasatinib, afatinib, crizotinib, bortezomib, filgrastim, or certolizumab pegol were observed within 4 weeks. >50% of the reports of DIILD following administration of amiodarone, methotrexate, PEG IFN-2α, leflunomide, or etanercept were recorded more than 4 months of treatment initiation. The WSP β (95% CI) of amiodarone, nivolumab, gefitinib, and sho-saiko-to was 0.77 (0.70–0.84), 0.90 (0.85–0.95), 0.78 (0.74–0.82), and 0.76 (0.59–0.95), respectively. The lower limits of the 95% CI of the WSP β value for daptomycin, vinorelbine, paclitaxel, amrubicin, bevacizumab, everolimus, and PEG INF-2α were greater than 1.

Figure 2.

Figure 2.

A box plot of drug-induced interstitial lung disease. The bottom end is minimum value. The top end is maximum value. The bottom of black box is 25th percentile. The top of white box is 75th percentile. The line joining the white and black is median. Panel A contains the drugs from ATC code A02BA03 to ATC code L01XA03 in the Table 1. Panel B contains the drugs from ATC code L01XC02 to ATC code V03AF04 in the Table 1.

To evaluate the risk factors for DIILD by using demographic data, such as age, patient history, and administered drugs, we applied the Apriori algorithm (minimum support and minimum confidence threshold, 0.00001 and 0.01, respectively) and maxlen was restricted to 3. The result of the mining algorithm for DIILD was a set of 11 rules, respectively (Table 2). {sho-saiko-to, 50–59 years}, {sho-saiko-to, 60–69 years}, {sho-saiko-to, 70–79 years}  ⇒ {DIILD}, {sho-saiko-to-ka-kikyo-sekko, 70–79 years}  ⇒ {DIILD} demonstrated high lift scores (Table 2, id(8–11) and Figure 3). The association rules of the combination of {amiodarone, 50–59 years}, {amiodarone, 60–69 years}, {amiodarone, 70–79 years}, {amiodarone, 80–89 years}, {amiodarone, ⩾ 90 years}  ⇒ {DIILD} demonstrated high support and lift scores (Table 2, id(3–7) and Figure 3).

Table 2.

Association parameters of rules of Drug-Induced Interstitial Lung Disease (DIILD) based on the administered drug and the stratified age group (sort by lift).

Id lhsa rhsb Support Confidence Lift χ2
[1] {amiodarone, 40–49 years} {DIILD} 0.00015 0.52288 1.17 3.52
[2] {amiodarone, 30–39 years} {DIILD} 0.00001 0.06667 1.49 0.90
[3] {amiodarone, 50–59 years} {DIILD} 0.00011 0.18182 4.06 142.24c
[4] {amiodarone, ⩾ 90 years} {DIILD} 0.00019 0.21277 4.75 315.56c
[5] {amiodarone, 60–69 years} {DIILD} 0.00034 0.27492 6.14 820.47c
[6] {amiodarone, 70–79 years} {DIILD} 0.00048 0.29702 6.64 1288.35c
[7] {amiodarone, 80–89 years} {DIILD} 0.00025 0.29797 6.66 673.40c
[8] {sho-saiko-to, 50–59 years} {DIILD} 0.00019 0.03030 6.77 505.12c
[9] {sho-saiko-to-ka-kikyo-sekko, 70–79 years} {DIILD} 0.00011 0.31579 7.06 320.15c
[10] {sho-saiko-to, 70–79 years} {DIILD} 0.00049 0.38806 8.67 1863.67c
[11] {sho-saiko-to, 60–69 years} {DIILD} 0.00036 0.48718 10.89 1810.41c
a

lhs: left-hand-side (antecedents).

b

rhs: right-hand-side (consequents).

c

Statistical significance: χ2 value ⩾ 4.

Figure 3.

Figure 3.

Association rules for drug-induced interstitial lung disease based on the JADER database between April 2004 and June 2018. The arguments of plot in the arulesViz were set as follows: method = “graph,” measure = “support,” shading = “lift.” The measures of support were used in visualization as area of circle. The measures of lift were used for the shading of color of the circle. Support and lift were visualized using the R-extension package arulesViz which implements novel visualization techniques to explore association rules.

Discussion

In this study, we evaluated the relationship between the drug and DIILD by using data from the SRS database. The exact frequency of drug-induced pulmonary toxicity is unknown.3 Although global incidence of DIILD is not clearly known, at least 2.5%–3.0% of cases are drug induced.19,20 Several studies have indicated that drug-induced pulmonary toxicity is underdiagnosed worldwide.3 We summarized the incidence of DIILD, the ROR values, and time-to-onset profile from the SRS database. It is considered to be more comprehensive information indicating the occurrence of DIILD reflecting the actual clinical use than has been published previously.

DIILD can occur at any time during treatment.21 We applied time-to-onset analysis to validate the results, and found that >50% of the DIILD cases associated with carboplatin, gefitinib, erlotinib, dasatinib, afatinib, crizotinib, bortezomib, and so on were observed within 4 weeks in the real-world data set. DIILD occurring after 4 months of amiodarone, methotrexate, PEG IFN-2α, leflunomide, or etanercept administration should not be overlooked.

It is suggested that risk factors for amiodarone-related DIILD were cumulative dose, and a combination of high doses over longer periods.22 The cumulative incidence of amiodarone-related DIILD was 4.2%, 7.8%, and 10.6% after 1, 3, and 5 years, respectively, during 48-month follow-up periods in a retrospective study.23 The time-to-onset duration of amiodarone was 123.0 days in our study using the JADER data set. Amiodarone-related DIILD was likely to be initial-failure type. For methotrexate, Kremer et al.24 reported a mean time to DIILD onset of 23 days (range = 3–112 days). In other studies, time to DIILD onset has been as long as 4 years.25 The onset of DIILD due to methotrexate was 145.5 days in our study. A nationwide Japanese study of gemcitabine determined a median time of onset of 65 days.2 The onset of DIILD due to gemcitabine was 53.0 days in our study. The median DIILD initiation time in patients with germ cell tumors receiving high-dose bleomycin was 4.2 months (126 days).26 The median DIILD initiation time of bleomycin was 92.0 days in our study. DIILD onset of epidermal growth factor receptor (EGFR)-directed monoclonal antibodies such as cetuximab and panitumumab demonstrated a broad range of times (median = 101 days, range = 17–431 days).27 The time-to-onset durations of cetuximab and panitumumab were 45.0 and 55.0 days in our study, respectively. For immune checkpoint inhibitors such as programmed cell death 1 (PD-1) inhibitors (nivolumab (DIILD onset in the JADER data set: 56.0 days), pembrolizumab (DIILD onset in the JADER data set: 40.0 days)), time to onset ranged from 0.2 to 27.4 months, with DIILD occurring within 2 months of treatment initiation in 42% of patients.2 No clear relationship has been observed between DIILD onset and dose or duration of treatment.28 Gefitinib (DIILD onset in the JADER data set: 24.0 days) and erlotinib (DIILD onset in the JADER data set: 21.0 days) are EGFR-targeting agents. The incidence of DIILD associated with gefitinib and erlotinib was highest within 4 weeks (28 days) of the initiation of treatment.29,30 DIILD induced by gefitinib was likely to be initial-failure type. Crizotinib, an oral tyrosine kinase inhibitor, induced DIILD several months after the initiation of treatment (median, 8.5 (6.5–11.5) months (255 days)).31 In contrast, the onset of DIILD due to crizotinib was 17.0 days in our study. A distinct discrepancy in crizotinib was observed in the time-to-onset duration between the literature data and our result; however, we do not have a plausible explanation for this discrepancy. For leflunomide (DIILD onset in the JADER data set: 131.5 days), DIILD was reported in most patients within 20 weeks (140 days) in a study in Japan.32 Our findings for the time to onset were not clearly linked to the literature data. However, we could demonstrate similar trends in most of the drugs considered in this study. Information from the SRS database and the literature data might be considered complementary.

There are many unclear points about the causative substances and underlying mechanisms of DIILD, which is diagnosed on the basis of clinical, physiological, and radiological findings consistent with interstitial lung disease.2 Some of the known risk factors of DIILD include follows: age, drug interaction, genetic variations, ethnicity, dose, sex, radiation-induced lung injury, pulmonary edema, smoking, progression of the underlying disease, and use or non-use of corticosteroid therapy.3,4

In general, old age is associated with an increased risk of drug toxicity.3 In a retrospective review of the pulmonary toxicity of bleomycin, Simpson et al.33 showed that for cases in which pulmonary toxicity was fatal, the patients were older than the remaining patients, and in patients aged over 40 years, especially those with renal function in the lower range of normal, the risk of developing fatal toxicity might exceed 10%.3 We detected the possible association rule related to DIILD for the combination of sho-saiko-to or amiodarone and aging (⩾50 years). Furthermore, the other rule of association {sho-saiko-to-ka-kikyo-sekko, 70–79 years} was observed in the antecedent (lhs). Thus, elderly patients receiving sho-saiko-to or amiodarone should be advised to adhere to appropriate treatment plan.

Sho-saiko-to contains seven crude drugs.34 Among them, Bupleurum root and Scutellaria root are thought to be the potential causes of lung injury.34 Many Chinese herbal medicines contain Bupleurum root and Scutellaria root, and herbal medicines such as saiko-ka-ryukotsu-borei-to and sai-rei-to can induce DIILD in a manner similar to that associated with sho-saiko-to.35,36 It remains to be elucidated whether one or both drugs affect the lungs. Until then, it is a reasonable assumption that DIILD associated with sho-saiko-to was caused by Bupleurum root and Scutellaria root.34

Drug interaction by concomitant drug use is a risk factor of AEs. As people age, they develop more chronic diseases and, accordingly, use more drugs. It is reported that amiodarone inhibits CYP1A2, CYP3A4, CYP2C9, and CYP2D6.3739 As the medication that is metabolized by any of these enzymes will be affected by plasma levels, it is likely that patients using amiodarone use other drugs which might increase the risk to DIILD occurrence. We evaluated the dose dependency of amiodarone on DIILD. The average dose of amiodarone for cases with DIILD (n = 778) and without DIILD (n = 1351) was 211.2 ± 154.3 (mean ± standard deviation) and 191.4 ± 174.9 mg/day, respectively. There were no statistically significant differences in our results. We did not evaluate the effects of concomitant drugs further.

Gefitinib plasma levels might be affected when using drugs that are metabolized by CYP2D6, such as metoprolol.37,38 In our study, the number of all AE reports related to gefitinib was 2736. The number of cases of DIILD related to gefitinib was 1217. The combination of gefitinib and metoprolol was 8, and 4 cases were related to DIILD among them (8 cases). We did not examine the potential drug-by-drug bias of gefitinib and metoprolol because there were too few cases for a robust analysis.

Erlotinib and smoking are also a bad combination because of the induction of CYP1A2 and the subsequent lower plasma levels.3840 Even doubling up the dose (300 mg instead of 150 mg) is not sufficient,41 but it can increase the incidence of DIILD, even without the presence of a polymorphism in one or several of these enzymes. As variability in drug response among patients is multifactorial, genetic variations in metabolizing enzymes may enhance the drivers of DIILD. Both clinical and genetic risk stratification (pharmacogenomics) may lead to a more accurate prevention of drug-induced lung damage in the future.

Our study has some limitations that should be considered. First, the JADER database does not contain detailed background information, such as genetic information, lifestyle habit (e.g. smoking), medical history (e.g. treatment regimen and pre-existing lung disease). For example, as detailed information is lacking from the studied population, factors affecting latency time (time to occurrence of the DIILD), such as concomitant infections that increase the degree of oxidative stress and cell injury or the occurrence of renal impairment, that influence pharmacokinetics and therefore serum drug levels,2,42 are not evaluated. Second, the SRS is subject to over-reporting, under-reporting, missing data, exclusion of data from healthy individuals, lack of a denominator, and presence of confounding factors.9 Therefore, ROR is not applicable to inferences of comparative degrees of causality. ROR only offers a rough indication of signal strength. Several approaches can be used to control for covariates, such as multiple-logistic regression,43 Bayesian logistic regression,44 and propensity score.45 These approaches may be useful for further analysis of SRS. Third, in the association rule mining method, the researcher determined the parameters (support, confidence, and maxlen) according to the data set and purpose of the research. Therefore, further epidemiological studies may be required to confirm the results of this study.

Conclusion

Despite the limitations inherent to the SRS, we showed the potential risk of DIILD in a real-life setting. The present analysis showed that patients receiving gefitinib, erlotinib, afatinib, or crizotinib should be closely monitored for the development of DIILD within a short duration (4 weeks). In contrast, patients receiving methotrexate, leflunomide, etanercept, amiodarone, or PEG INF-2α should be carefully monitored for the development of DIILD over a longer duration (more than 4 months). Patients who are co-administered amiodarone, sho-saiko-to, and sho-saiko-to-ka-kikyo-sekko should also be carefully monitored for the development of DIILD.

Footnotes

Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was partially supported by Japan Society for the Promotion of Science KAKENHI grant number, 17K08452. The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the article.

Ethical approval: Ethical approval was not sought for this study because the study was an observational study without any research subjects. All results were obtained from data openly available online from the PMDA website. All data from the JADER database were fully anonymized by the regulatory authority before we accessed them.

Informed consent: Informed consent was not sought for the present study because the study was an observational study without any research subjects. All results were obtained from data openly available online from the Pharmaceuticals and Medical Devices Agency (PMDA) website (www.pmda.go.jp). All data from the JADER database were fully anonymized by the regulatory authority before we accessed them.

Trial registration: This clinical trial was not registered because the study was an observational study without any research subjects. All results were obtained from data openly available online from the Pharmaceuticals and Medical Devices Agency (PMDA) website (www.pmda.go.jp). All data from the JADER database were fully anonymized by the regulatory authority before we accessed them.

ORCID iD: Mitsuhiro Nakamura Inline graphic https://orcid.org/0000-0002-5062-5522

References

  • 1. Antoniou KM, Margaritopoulos GA, Tomassetti S, et al. Interstitial lung disease. Eur Respir Rev 2014; 23: 40–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Skeoch S, Weatherley N, Swift AJ, et al. Drug-induced interstitial lung disease: a systematic review. J Clin Med 2018; 7: 356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Schwaiblmair M, Behr W, Haeckel T, et al. Drug induced interstitial lung disease. Open Respir Med J 2012; 6: 63–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Roden AC, Camus P. Iatrogenic pulmonary lesions. Semin Diagn Pathol 2018; 35: 260–271. [DOI] [PubMed] [Google Scholar]
  • 5. Azuma A, Kudoh S. High prevalence of drug-induced pneumonia in Japan. Japan Med Assoc J 2007; 50: 405–411. [Google Scholar]
  • 6. Hatahira H, Hasegawa S, Sasaoka S, et al. Analysis of fall-related adverse events among older adults using the Japanese Adverse Drug Event Report (JADER) database. J Pharm Health Care Sci 2018; 4: 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Tanaka M, Hasegawa S, Nakao S, et al. Analysis of drug-induced hearing loss by using a spontaneous reporting system database. PLoS ONE 2019; 14(10): e0217951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Komada F. Analysis of time-to-onset of interstitial lung disease after the administration of small molecule molecularly-targeted drugs. Yakugaku Zasshi 2018; 138(2): 229–235. [DOI] [PubMed] [Google Scholar]
  • 9. Van Puijenbroek EP, Bate A, Leufkens HG, et al. A comparison of measures of disproportionality for signal detection in spontaneous reporting systems for adverse drug reactions. Pharmacoepidemiol Drug Saf 2002; 11(1): 3–10. [DOI] [PubMed] [Google Scholar]
  • 10. Van Puijenbroek EP, Egberts AC, Heerdink ER, et al. Detecting drug-drug interactions using a database for spontaneous adverse drug reactions: an example with diuretics and non-steroidal anti-inflammatory drugs. Eur J Clin Pharmacol 2000; 56(9–10): 733–738. [DOI] [PubMed] [Google Scholar]
  • 11. Poluzzi E, Raschi E, Piccinni C, et al. Data mining techniques in pharmacovigilance: analysis of the publicly accessible FDA Adverse Event Reporting System (AERS). In: Karahoca A. (ed.) Data mining applications in engineering and medicine. Rijeka: InTech, 2012, pp. 265–302. [Google Scholar]
  • 12. Hasegawa S, Matsui T, Hane Y, et al. Thromboembolic adverse event study of combined estrogen-progestin preparations using Japanese Adverse Drug Event Report database. PLoS ONE 2017; 12(7): e0182045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Nakao S, Hatahira H, Sasaoka S, et al. Evaluation of drug-induced photosensitivity using the Japanese Adverse Drug Event Report (JADER) Database. Biol Pharm Bull 2017; 40(12): 2158–2165. [DOI] [PubMed] [Google Scholar]
  • 14. Sauzet O, Carvajal A, Escudero A, et al. Illustration of the Weibull shape parameter signal detection tool using electronic healthcare record data. Drug Saf 2013; 36(10): 995–1006. [DOI] [PubMed] [Google Scholar]
  • 15. Shimada K, Hasegawa S, Nakao S, et al. Adverse event profiles of ifosfamide-induced encephalopathy analyzed using the Food and Drug Administration Adverse Event Reporting System and the Japanese Adverse Drug Event Report databases. Cancer Chemother Pharmacol 2019; 84(5): 1097–1105. [DOI] [PubMed] [Google Scholar]
  • 16. Mukai R, Hasegawa S, Umetsu R, et al. Evaluation of pregabalin-induced adverse events related to falls using the FDA Adverse Event Reporting System (FAERS) and Japanese Adverse Drug Event Report (JADER) database. J Clin Pharm Ther 2019; 44: 285–291. [DOI] [PubMed] [Google Scholar]
  • 17. Sasaoka S, Matsui T, Hane Y, et al. Time-to-onset analysis of drug-induced long QT syndrome based on a spontaneous reporting system for adverse drug events. PLoS ONE 2016; 11(10): e0164309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Yildirim P. Association patterns in open data to explore ciprofloxacin adverse events. Appl Clin Inform 2015; 6(4): 728–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Coultas DB, Zumwalt RE, Black WC, et al. The epidemiology of interstitial lung diseases. Am J Respir Crit Care Med 1994; 150: 967–972. [DOI] [PubMed] [Google Scholar]
  • 20. Thomeer MJ, Costabe U, Rizzato G, et al. Comparison of registries of interstitial lung diseases in three European countries. Eur Respir J Suppl 2001; 32: 114s–118s. [PubMed] [Google Scholar]
  • 21. Blum RH, Carter SK, Agre K. A clinical review of bleomycin—a new antineoplastic agent. Cancer 1973; 31(4): 903–914. [DOI] [PubMed] [Google Scholar]
  • 22. Kupferschmid JP, Rosengart TK, McIntosh CL, et al. Amiodarone-induced complications after cardiac operation for obstructive hypertrophic cardiomyopathy. Ann Thorac Surg 1989; 48(3): 359–364. [DOI] [PubMed] [Google Scholar]
  • 23. Yamada Y, Shiga T, Matsuda N, et al. Incidence and predictors of pulmonary toxicity in Japanese patients receiving low-dose amiodarone. Circ J 2007; 71(10): 1610–1616. [DOI] [PubMed] [Google Scholar]
  • 24. Kremer JM, Alarcon GS, Weinblatt ME, et al. Clinical, laboratory, radiographic, and histopathologic features of methotrexate-associated lung injury in patients with rheumatoid arthritis: a multicenter study with literature review. Arthritis Rheum 1997; 40(10): 1829–1837. [DOI] [PubMed] [Google Scholar]
  • 25. Imokawa S, Colby TV, Leslie KO, et al. Methotrexate pneumonitis: review of the literature and histopathological findings in nine patients. Eur Respir J 2000; 15: 373–381. [DOI] [PubMed] [Google Scholar]
  • 26. O’Sullivan JM, Huddart RA, Norman AR, et al. Predicting the risk of bleomycin lung toxicity in patients with germ-cell tumours. Ann Oncol 2003; 14(1): 91–96. [DOI] [PubMed] [Google Scholar]
  • 27. Ishiguro M, Watanabe T, Yamaguchi K, et al. A Japanese post-marketing surveillance of cetuximab (Erbix®) in patients with metastatic colorectal cancer. Jpn J Clin Oncol 2012; 42: 287–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Khunger M, Rakshit S, Pasupuleti V, et al. Incidence of pneumonitis with use of programmed death 1 and programmed death-ligand 1 inhibitors in non-small cell lung cancer: a systematic review and meta-analysis of trials. Chest 2017; 152(2): 271–281. [DOI] [PubMed] [Google Scholar]
  • 29. Gemma A, Kudoh S, Ando M, et al. Final safety and efficacy of erlotinib in the phase 4 POLARSTAR surveillance study of 10,708 Japanese patients with non-small-cell lung cancer. Cancer Sci 2014; 105(12): 1584–1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kudoh S, Kato H, Nishiwaki Y, et al. Interstitial lung disease in Japanese patients with lung cancer: a cohort and nested case-control study. Am J Respir Crit Care Med 2008; 177(12): 1348–1357. [DOI] [PubMed] [Google Scholar]
  • 31. Crequit P, Wislez M, Fleury Feith J, et al. Crizotinib associated with ground-glass opacity predominant pattern interstitial lung disease: a retrospective observational cohort study with a systematic literature review. J Thorac Oncol 2015; 10(8): 1148–1155. [DOI] [PubMed] [Google Scholar]
  • 32. Chikura B, Lane S, Dawson JK. Clinical expression of leflunomide-induced pneumonitis. Rheumatology (Oxford) 2009; 48(9): 1065–1068. [DOI] [PubMed] [Google Scholar]
  • 33. Simpson AB, Paul J, Graham J, et al. Fatal bleomycin pulmonary toxicity in the west of Scotland 1991–95: a review of patients with germ cell tumours. Br J Cancer 1998; 78(8): 1061–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Komiya K, Ishii H, Ohama M, et al. Sai-rei-to-induced lung injury: a case report and brief review of the literature. Intern Med 2012; 51(24): 3421–3425. [DOI] [PubMed] [Google Scholar]
  • 35. Tokunaga T. A case of drug-induced pneumonitis associated with Chinese herbal drugs and valsartan. Nihon Kokyuki Gakkai Zasshi 2005; 43(7): 406–411. [PubMed] [Google Scholar]
  • 36. Sekiyama T, Asai Y, Fujii T, et al. Case of drug-induced pneumonia due to Saiko-karyuukotsu-boreitou. Nihon Kokyuki Gakkai Zasshi 2009; 47(5): 362–366. [PubMed] [Google Scholar]
  • 37. Wijnen PA, Drent M, Nelemans PJ, et al. Role of cytochrome P450 polymorphisms in the development of pulmonary drug toxicity: a case-control study in the Netherlands. Drug Saf 2008; 31(12): 1125–1134. [DOI] [PubMed] [Google Scholar]
  • 38. Wijnen PA, Bekers O, Drent M. Relationship between drug-induced interstitial lung diseases and cytochrome P450 polymorphisms. Curr Opin Pulm Med 2010; 16(5): 496–502. [DOI] [PubMed] [Google Scholar]
  • 39. Jessurun NT, Drent M, Van Puijenbroek EP, et al. 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. [DOI] [PubMed] [Google Scholar]
  • 40. O’Malley M, King AN, Conte M, et al. Effects of cigarette smoking on metabolism and effectiveness of systemic therapy for lung cancer. J Thorac Oncol 2014; 9(7): 917–926. [DOI] [PubMed] [Google Scholar]
  • 41. Hamilton M, Wolf JL, Rusk J, et al. Effects of smoking on the pharmacokinetics of erlotinib. Clin Cancer Res 2006; 12(7 Pt 1): 2166–2171. [DOI] [PubMed] [Google Scholar]
  • 42. Kao MP, Ang DS, Pall A, et al. Oxidative stress in renal dysfunction: mechanisms, clinical sequelae and therapeutic options. J Hum Hypertens 2010; 24(1): 1–8. [DOI] [PubMed] [Google Scholar]
  • 43. Suzuki Y, Suzuki H, Umetsu R, et al. Analysis of the interaction between clopidogrel, aspirin, and proton pump inhibitors using the FDA adverse event reporting system database. Biol Pharm Bull 2015; 38(5): 680–686. [DOI] [PubMed] [Google Scholar]
  • 44. Szarfman A, Machado SG, O’Neill RT. Use of screening algorithms and computer systems to efficiently signal higher-than-expected combinations of drugs and events in the US FDA’s spontaneous reports database. Drug Saf 2002; 25(6): 381–392. [DOI] [PubMed] [Google Scholar]
  • 45. Akimoto H, Oshima S, Negishi A, et al. Assessment of the risk of suicide-related events induced by concomitant use of antidepressants in cases of smoking cessation treatment with varenicline and assessment of latent risk by the use of varenicline. PLoS ONE 2016; 11(9): e0163583. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from SAGE Open Medicine are provided here courtesy of SAGE Publications

RESOURCES