Skip to main content
Asian Pacific Journal of Cancer Prevention : APJCP logoLink to Asian Pacific Journal of Cancer Prevention : APJCP
. 2024;25(7):2291–2295. doi: 10.31557/APJCP.2024.25.7.2291

Immune-Related Adverse Events due to Concomitant Use of Immune Checkpoint Inhibitors and Chinese Herbal Medicines: A Study Based on a Japanese Adverse Event Database

Toru Koshiishi 1, Nanako Nishioka 1, Koichi Yoshimoto 1
PMCID: PMC11480620  PMID: 39068560

Abstract

Background:

Fatigue is an immune-related adverse event (irAE) associated with immune checkpoint inhibitors (ICIs) used for cancer treatment. Chinese herbal medicines (Ho-zai) are used to treat cancer-related fatigue. However, no interactions between ICIs and Ho-zai have been reported. Herein, we investigated the risk of irAEs associated with the concomitant use of ICIs and Ho-zai.

Methods:

We extracted data of patients who used ICI and Ho-zai from the Japanese Adverse Event Reporting Database. The proportional reporting ratio (PRR) was calculated for patients using ICI, Ho-zai, or both. We focused on cases of interstitial lung disease (ILD) and colitis, which were among the most severe cases of irAEs among these patients. The shrinkage method used by the World Health Organization-Uppsala Monitoring Center was used to detect the interactions.

Results:

Of the 799,670 patients in the database, 77,219, 2060, and 92 were using ICIs, Ho-zai, and combination treatment, respectively. The ILD and colitis groups included 39,388 and 17,522 patients, respectively. ILD signals were detected for both ICIs and Ho-zai. There were 24 cases of patients treated with concomitant ICIs and Ho-zai who developed ILD. For all combinations of all ICIs and all Ho-zai, Ω025 was negative, which suggested no ILD-related interactions. Colitis signals were detected for ICIs except for atezolizumab, avelumab, and durvalumab. There were eight patients treated with concomitant ICI and Ho-zai who developed colitis. For all combinations of all ICIs and all Ho-zai, Ω025 was negative, which suggested no colitis-related interactions.

Conclusion:

To our knowledge, this is the first study to investigate interactions between ICIs and Ho-zai. Signals were detected for ILD in both ICI and Ho-zai groups, and colitis in the ICI group. However, the combined use of these treatments did not increase the risk of irAEs.

Key Words: immune-related adverse events, immune checkpoint inhibitors, Chinese, herbal medicines

Introduction

Fatigue commonly occurs in patients with cancer and impairs their quality of life [1]. There is no standardized treatment for fatigue; however, various intervention approaches have been attempted, including physical activity, psychosocial, psychosomatic, and pharmacological treatments [2]. In recent years, immune checkpoint inhibitors (ICIs) have been used in chemotherapy for all types of cancer beginning with the first use of nivolumab for malignant melanoma. Immune-related adverse events (irAEs) associated with the use of ICIs are known, and the most severe of these AEs include interstitial lung disease (ILD), colitis, thyroid dysfunction, and scratchiness. Furthermore, one of the most frequent irAEs is fatigue [3, 4].

The use of Chinese herbal medicines, such as Hochuikitou Hochuekkito (HET), Juzen Daihoto (JJT), and Ginsen Yoeito (NYT), for the treatment of fatigue in patients with tumors has been reported [5, 6]. These herbal medicines are called “Ho-zai” because they are effective in replenishing the deficiency in the physical strength and energy of the body and restoring the condition of the patient. Reportedly, one mechanism of action of Ho-zai involves T cells. HET has been reported to improve an inhibition of tumor-specific Th1-type cytokine production [7], JTT increases the regulatory activity of T cells by reducing the Foxp3(+) Treg population in patients with pancreatic cancer [8], and NYT has been reported to potentiate the effects of tumor vaccines via CD8+ T cells [9]. Furthermore, a study examining the T cell and cytokine-inducing effects of HET, JTT, and NYT showed that neither HET, JTT, nor NYT induced CD4+ T cells but tended to increase CD8+ T cells in a dose-dependent manner. Ho-zai has also been reported to decrease regulatory T cells in a dose-dependent manner [10]. Thus, the use of these herbal medicines for treating fatigue in patients receiving ICIs may exacerbate irAEs, as well as their therapeutic effects. However, to date, there are no reports on the interactions between ICIs and Ho-zai.

The Pharmaceuticals and Medical Devices Agency collects post-marketing spontaneous AE reports and makes them publicly available via the Japanese Adverse Drug Event Report (JADER) database. JADER is a database that contains a case list table with basic patient information such as sex and age; a drug information table with details of the generic name and start and end date of administration; an AE table with information regarding the name, outcome, and onset date of the AE; and an underlying disease table with the underlying disease and other information. Recently, type 1 diabetes has been reported to be associated with ICI administration [11] and irAEs [12] in the JADER database. Herein, we used JADER to investigate the possible effects of Ho-zai used for fatigue on irAEs.

Materials and Methods

Analysis tables and extraction of target patient data

Data was extracted from the JADER database from April 2004 to November 2022 [13]. The drug information table classifies administered drugs as suspected drugs, concomitant drugs, and interactions according to their involvement in AEs. In this study, suspected drugs to be responsible for AEs were included in the analysis. The ICIs included in this study were nivolumab, pembrolizumab, durvalumab, atezolizumab, avelumab, and ipilimumab, all of which are approved in Japan. Because ipilimumab is sometimes used in combination with nivolumab, the combination group was defined as the nivolumab plus ipilimumab combination group. The herbal medicines used in this study were HET, JJT, and NYT. Cases in which the drug name was not included in the drug information table were excluded.

The irAEs considered relevant for patients who received ICIs and were admitted to the intensive care unit included respiratory diseases, colitis, and metabolic diseases [14]. Metabolic diseases included renal failure, adrenocortical insufficiency, hyponatremia, and thrombotic microangiopathies. Because all these diseases were present in a small number of cases, we chose to focus on interstitial lung disease (ILD) and colitis in this study.

The JADER AE terms are based on the basic definitions in the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use International Dictionary for Regulatory Activities/Japanese version (Medical Dictionary for Regulatory Activities/Japanese version, hereafter MedDRA). In this study, ILD and colitis of the MedDRA Standardized MedDRA Queries (SMQ) of MedDRA Ver. 25.1 were used. The case lists, drug information, and AE tables were combined into a single file and used as tables for analysis.

Signal analysis for ILD and colitis

The proportional reporting ratio (PRR) [15] was used to detect signals related to ILD and colitis. The following equations were used for calculating PRR and χ2. In this study, PRR≥2, χ2≥4, and N11≥3 were considered as a signal.

PRR=(N11/N1+)(N01/NO+)      (1)

x2=n+++×N11×N00-N10×N01-n+++/2)2N1+×N+1×N0×N+0           (2)

Drug D1: N11 = n111 + n101, N00 = n000 + n010, N10 = n110+ n100, N01 = n001 + n011, N1+ = n11++ n10+, N+1 = n++1, N0+ = n00+ + n01+, N+0 = n++0.

Drug D2: N11 = n111 + n011, N00 = n000 + n100, N10 = n110+ n010, N01 = n001 + n101, N1+ = n11++ n01+, N+1 = n++1, N0+ = n00+ + n10+, N+0 = n++0.

Signal analysis of drug-drug interactions

We used the Ω shrinkage measure model [16] for signal detection of drug–drug interactions. This method is based on a measure calculated as the ratio of the observed reporting ratio of the adverse event (AE) associated with the combination of two drugs and its expected value. Further, this model is used by the Uppsala Monitoring Center (UMC) and the World Health Organization (WHO) Collaborating Centre for International Drug Monitoring for signal analysis of drug–drug interactions. The Ω shrinkage measure prepares a 4 × 2 contingency table when AEs are considered in combination, when each AE is used alone, and when all other drugs are used; and the signal is obtained by dividing the observed value by the expected value. The detailed method of calculating the signal is shown in Equation 4. Ω025 > 0 was used as the threshold to screen for signals associated with a two-drug combination.

Ω=log2n111+0.5E111+0.5          (3)

Ω025=Ω-(0.975)log2n1112           (4)         

n111: reported number of AEs associated with a targeted two-drug combination.

E111: expected number of AEs associated with the targeted two-drug combination.

Φ (0.975) was 97.5% of the standard normal distribution.

Statistical analysis

JMP13.0 (SAS Institute Inc., Cary, NC, USA) was used for the all statistical analyses.

Results

Of the 799,670 patients registered in the JADER database, 77,219 were in the ICI group, 2060 in the Ho-zai group, and 92 in the combination group. The ILD group consisted of 39,388 patients and the colitis group consisted of 17,522 patients, respectively.

Colitis

Colitis signals were detected with ICI, except for atezolizumab, avelumab, and durvalumab (Table 1). There were eight cases of patients treated with ICIs and Ho-zai who developed colitis. The combinations included one case each for nivolumab+ipilimumab+JJY, nivolumab+JJY, pembrolizumab+JJY, and pembrolizumab+NYT, and nivolumab+HET in four cases. Ω025 was not detectable for nivolumab+ipilimumab+JJY, nivolumab+JJY, pembrolizumab+JJY, and pembrolizumab+NYT. For each nivolumab+HET, the combination of all ICIs and all Ho-zai, Ω025 was negative (Table 2).

Table 1.

ILD and Colitis Signals were Detected with each Drug, all ICIs, and all Ho-zai

Drug Name ILD Other PRR χ2 Colitis Other PRR χ2
Atezolizumab 673 4062 2.92 875.42 200 4,535 1.94 90.88
Avelumab 20 424 0.91 0.09 29 415 2.98 37.05
Durvalumab 1440 1180 11.54 14042.63 30 2,590 0.52 12.94
Ipilimumab 34 364 1.74 10.37 79 319 9.1 571.13
Nivolumab 2270 7391 5 7198.01 636 9,025 3.08 878.12
Pembrolizumab 2194 8909 4.19 5288.11 563 10,540 2.36 434.26
Nivolumab+ipilimumab 1250 7008 3.14 1855.72 810 7,448 4.64 2255.63
All ICIs 7881 29338 5.12 22005.71 2347 34,872 3.17 3081.9
HET 102 931 2.01 53.04 35 998 1.55 6.37
JJT 90 748 2.18 59.32 21 817 1.14 0.25
NYT 24 165 2.58 22.76 6 183 1.45 0.46
All Ho-zai 216 1844 2.14 135.15 62 1,998 1.37 6.08

ICI, Immune checkpoint inhibitors; JJT, Juzen Daihoto; NYT, Ginsen Yoeito; HET, Hochuekkito; Ho-zai, JJT+NYT+HET

Table 2.

Ω025 on Colitis with ICIs and Ho-zai

Drug Name Colitis Other Ω025
Nivolumab+HET 4 23 -0.52
Nivolumab+JJY 1 8 ND
Nivolumab+ipilimumab+JJY 1 5 ND
Pembrolizumab+JJY 1 16 ND
Pembrolizumab+NYT 1 5 ND
All ICIs+All Ho-zai 8 84 -0.26

ND, not detected; ICI, Immune checkpoint inhibitors; JJT, Juzen Daihoto; NYT, Ginsen Yoeito; HET, Hochuikitou Hochuekkito; Ho-zai, JJT+NYT+HET

ILD

ILD signals were detected for each drug, ICI, and Ho-zai (Table 1). There were 24 cases of patients treated with ICIs and Ho-zai who developed ILD. The combinations were atezolizumab+JJT in one case, atezolizumab+NYT in one case, atezolizumab+HET in one case, nivolumab+JJT in two cases, nivolumab+NYT in two cases, nivolumab+HET in nine cases, pembrolizumab+JJT in three cases, pembrolizumab+NYT three cases, and pembrolizumab+HET in two cases. Ω025 was not detectable for atezolizumab with each Ho-zai. For each of the other drug combinations, the combination of all ICIs and all Ho-zai, Ω025 was negative (Table 3).

Table 3.

Ω025 on ILD with ICIs and Ho-zai

Drug Name ILD Other Ω025
Atezolizumab+HET 1 2 ND
Atezolizumab+JJT 1 0 ND
Atezolizumab+NYT 1 0 ND
Nivolumab+HET 9 18 -0.21
Nivolumab+JJT 2 7 -1.91
Nivolumab+NYT 2 3 -1.28
Pembrolizumab +HET 2 3 -1.08
Pembrolizumab+JJT 3 14 -1.44
Pembrolizumab+NYT 3 3 -0.23
All ICIs+All Ho-zai 24 68 -0.3

ND, not detected; ICI, Immune checkpoint inhibitors; JJT, Juzen Daihoto; NYT, Ginsen Yoeito; HET, Hochuekkito; Ho-zai, Standardize the order to HET→JJT→NYT.

Discussion

To our knowledge, this is the first study to investigate the interaction between ICIs and Ho-zai. Signals were detected for ILD in both the ICI and Ho-zai groups and for colitis in the ICI group. However, the combined use of these treatments did not increase the risk of irAEs.

Fatigue commonly occurs in patients with cancer and those undergoing cancer treatment. Ho-zai has been shown to effectively alleviate this fatigue [5, 6]. In patients receiving ICIs, fatigue is a symptom of serious AEs, such as hypoadrenalism, hypothyroidism, and myasthenia gravis. This study suggests that Ho-zai may not pose an additional risk when used concurrently with ICIs, which can be valuable for managing such events.

Ho-zai is known for activating cytotoxic T cells and reducing regulatory T cells. Additionally, the mechanism of action of CTLA-4 inhibitors is also understood to involve the activation of cytotoxic T cells and the reduction of regulatory T cells [17]. Furthermore, the combination of CTLA-4 inhibitors with PD-1 antibodies is expected to enhance response rates and is currently used in clinical practice [18]. The combination of HET and PD-1 inhibitors was found to suppress tumor growth and increase cytotoxic T lymphocytes and natural killer cells in MC38 colon cancer model mice. The combination of Ho-zai with ICIs suggests the potential enhancement of anti-cancer immune activity [19]. Conversely, it has been reported that the incidence of irAEs, including ILD and colitis, is higher when CTLA-4 inhibitors and PD-1/PD-L1 antibodies are used in combination than when they are used individually [20, 21, 22].

However, in this study, the combination of ICI and Ho-zai did not increase the risk of ILD or colitis. When ipilimumab was administered at 10 mg/kg or 3 mg/kg for unresectable stage III or IV melanoma, it was reported that colitis had a higher incidence when administered at 10 mg/kg, suggesting a dose-dependent relationship [23]. Therefore, it is suggested that the clinical dose of Ho-zai may not be sufficient to induce colitis. Additionally the incidence of irAE-related lung disorders and colitis varies among different tumor types, with non-small cell lung cancer (NSCLC) and renal cell carcinoma (RCC) having a higher incidence of lung disorders compared to melanoma, and melanoma having a higher incidence of colitis compared to the other two diseases [24, 25]. In this study, the information in the JADER database about the underlying diseases was limited; hence, the patient backgrounds were not considered, and it is possible that the bias in underlying diseases may have affected the results.

Although there have been reports of ILD caused by traditional Chinese medicine (TCM), all of them contained ou-gon, which is considered to be the cause of allergy [26, 27]. Regarding traditional Chinese medicine-induced colitis, phlebosclerotic colitis has been reported, which is considered to be caused by geniposide found in the gardenia fruit [28, 29]. However, Ho-zai used in this study did not contain ou-gon or the gardenia fruit, and it was considered that Ho-zai did not increase the risk of ILD or colitis in this study.

The present study has several limitations. First, it only considered two particularly urgent irAEs, and there may be interactions for other irAEs. Second, spontaneous reporting databases are generally associated with reporting biases, such as underreporting [30, 31], data loss [30], increased reporting rates for topical AEs (notoriety and ripple effects) [30], and duplication of reports [31]. Therefore, caution is warranted when interpreting the results obtained from the JADER database.

The interactions between ICIs and herbal medicines are not only limited to irAEs but may also have an impact on the therapeutic efficacy of the drugs. In the future, we will measure the treatment effect and include other irAEs using real-world data.

Acknowledgements

If it was approved by any scientific Body/ if it is part of an approved student thesis

This study did not require the approval of an ethics committee, as it involved the secondary use of existing, anonymized data from publicly accessible databases. No specific scientific body oversaw the conduct of this research; however, all procedures were carried out in accordance with relevant guidelines and regulations.

How the ethical issue was handled (name the ethical committee that approved the research)

This study does not fall under the ‘Code of Ethics for Medical Research Involving Human Subjects’ as it exclusively uses publicly available, anonymized data obtained from JADER. Therefore, it was determined that Institutional Review Board approval was not required.

Availability of data (if apply to your research)

The datasets generated and analyzed during the current study are available from the corresponding author on reasonable request.

Any conflict of interest

The authors declare no conflict of interest related to the study.

Author Contribution Statement

Toru Koshiishi and Koichi Yoshimoto contributed to the study conceptualization. Toru Koshiishi contributed to data analysis and interpretation, and also wrote the manuscript. Koichi Yoshimoto contributed to the manuscript drafting. Nanao Nishioka contributed to revising the manuscript. All authors critically reviewed and revised the manuscript draft and approved the final version for submission.

References

  • 1.Portenoy R, Irti LM. Cancer-related fatigue: guidelines for evaluation and management. Oncologist. 1999;4(1):1–10. [PubMed] [Google Scholar]
  • 2.Bower JE. Cancer-related fatigue: Mechanisms, risk factors, and treatments. Nat Rev Clin Oncol. 2014;11(10):597–609. doi: 10.1038/nrclinonc.2014.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Naidoo J, Page DB, Li BT, Connell LC, Schindler K, Lacouture ME, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol. 2015;26(12):2375–91. doi: 10.1093/annonc/mdv383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kanji S, Morin S, Agtarap K, Purkayastha D, Thabet P, Bosse D, et al. Adverse events associated with immune checkpoint inhibitors: Overview of systematic reviews. Drugs. 2022;82:793–809. doi: 10.1007/s40265-022-01707-1. [DOI] [PubMed] [Google Scholar]
  • 5.Inoue M, Hoshino E. Symptoms of cancer patients and Kampo formulas effective for them. Jpn J Cancer Chemether. 2015;42(13):2418–22. [Google Scholar]
  • 6.Qi F, Zhao L, Zhou A, Zhang B, Li A, Wang Z, et al. The advantages of using traditional Chinese medicine as an adjunctive therapy in the whole course of cancer treatment instead of only terminal stage of cancer. BioScience Trends. 2015;9(1):16–34. doi: 10.5582/bst.2015.01019. [DOI] [PubMed] [Google Scholar]
  • 7.Li T, Tamada K, Abe K, Tada H, Onoe Y, Tatsugami K, et al. The restoration of the antitumor T cell response from stress-induced suppression using a traditional Chinese herbal medicine Hochu-ekki-to (TJ-41: Bu-Zhong-Yi-Qi-Tang) Immunopharmacology. 1999;43(1):11–21. doi: 10.1016/s0162-3109(99)00034-x. [DOI] [PubMed] [Google Scholar]
  • 8.Ikemoto T, Shimada M, Iwahashi S, Saito Y, Kanamoto M, Mori H, et al. Changes of immunological parameters with administration of Japanese Kampo medicine (Juzen-Taihoto/TJ-48) in patients with advanced pancreatic cancer. Int J Clin Oncol. 2014;19(1):81–6. doi: 10.1007/s10147-013-0529-6. [DOI] [PubMed] [Google Scholar]
  • 9.Takaku S, Shimizu M, Takahashi H. Japanese Kampo medicine ninjin’yoeito synergistically enhances tumor vaccine effects mediated by CD8+ T cells. Oncol Lett. 2017;13(5):3471–3478. doi: 10.3892/ol.2017.5937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kiyomi A, Matsuda A, Nara M, Yamazaki K, Imai S, Sugiura M. Immunological differences in human peripheral blood mononuclear cells treated with Traditional Japanese Herbal Medicines Hochuekkito, Juzentaihoto, and Ninjin’yoeito from different pharmaceutical companies. Evid Based Complement Alternat Med. 2021;2021:7605057. doi: 10.1155/2021/7605057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Takada S, Hashishita H, Yamagishi K, Hideki S, Masayuki E. Predictors of the onset of type 1 diabetes obtained from real-world data analysis in cancer patients treated with immune checkpoint inhibitors. Asian Pac J Cancer Prev. 2020;21(6):1697–1699. doi: 10.31557/APJCP.2020.21.6.1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hasegawa S, Ikesue H, Nakao S, Shimada K, Mukai R, Tanaka M, et al. Analysis of immune-related adverse events caused by immune checkpoint inhibitors using the Japanese Adverse Drug Event Report database. Pharmacoepidemiol Drug Saf. 2020;29(10):1279–1294. doi: 10.1002/pds.5108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pharmaceuticals and Medical Devices Agency. Japanese adverse drug event report database [internet] Available from: https://www.info.pmda.go.jp/fukusayoudb/CsvDownload.jsp.
  • 14.Toffart AC, Meert AP, Wallet F, Gibelin A, Guisset O, Gonzalez F, et al. ICU admission for solid cancer patients treated with immune checkpoint inhibitors. Ann Intensive Care. 2023;13(1):29 . doi: 10.1186/s13613-023-01122-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Evans SJ, Waller PC, Davis S. Use of proportional reporting ratios (PRRs) for signal generation from spontaneous adverse drug reaction reports. Pharmacoepidemiol Drug Saf. 2001;10(6):483–6. doi: 10.1002/pds.677. [DOI] [PubMed] [Google Scholar]
  • 16.Norén GN, Sundberg R, Bate A, Edwards IR. A statistical methodology for drug-drug interaction surveillance. Stat Med. 2008;27(16):3057–70. doi: 10.1002/sim.3247. [DOI] [PubMed] [Google Scholar]
  • 17.Graziani G, Tentori L, Navarra P. Ipilimumab: a novel immunostimulatory monoclonal antibody for the treatment of cancer. Pharmacol Res. 2012;65(1):9–22. doi: 10.1016/j.phrs.2011.09.002. [DOI] [PubMed] [Google Scholar]
  • 18.Rotte A, Jin JY, Lemaire V. Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy. Ann Oncol. 2018;29(1):71–83. doi: 10.1093/annonc/mdx686. [DOI] [PubMed] [Google Scholar]
  • 19.Chun J, Park SM, Yi JM, Ha IJ, Kang HN, Jeong MK. Bojungikki-Tang improves response to PD-L1 immunotherapy by regulating the tumor microenvironment in MC38 tumor-bearing mice. Front Pharmacol. . 2022;13:901563. doi: 10.3389/fphar.2022.901563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–17. doi: 10.1056/NEJMoa1414428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34. doi: 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Naidoo J, Wang X, Woo KM, Iyriboz T, Halpenny D, Cunningham J, et al. Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy. J Clin Oncol. 2017;35(7):709–717. doi: 10.1200/JCO.2016.68.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Paolo A, Michele DV, Caroline R, Andrzej M, Vanna CS, Ana A, et al. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2017;18(5):611–622. doi: 10.1016/S1470-2045(17)30231-0. [DOI] [PubMed] [Google Scholar]
  • 24.Wang DY, Ye F, Zhao S, Johnson DB. Incidence of immune checkpoint inhibitor-related colitis in solid tumor patients: A systematic review and meta-analysis. Oncoimmunology. 2017;6(10):e1344805. doi: 10.1080/2162402X.2017.1344805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Khoja L, Day D, Wei-Wu Chen T, Siu LL, Hansen AR. Tumour- and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: a systematic review. Ann Oncol. 2017;28(10):2377–2385. doi: 10.1093/annonc/mdx286. [DOI] [PubMed] [Google Scholar]
  • 26.Enomoto Y, Nakamura Y, Enomoto N, Fujisawa T, Inui N, Suda T. Japanese herbal medicine-induced pneumonitis: A review of 73 patients. Respir Investig. 2017;55(2):138–144. [Google Scholar]
  • 27.Mochizuki N, Ano S, Kikuchi N, Sakai C, Masuda M, Kondo Y, et al. Pneumonitis due to Oren-gedoku-to (Coptis Detoxifying Decoction) Intern Med. 2019;58(20):3019–3023. doi: 10.2169/internalmedicine.2586-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hiramatsu K, Sakata H, Horita Y, Orita N, Kida A, Mizukami A, et al. Mesenteric phlebosclerosis associated with long-term oral intake of geniposide, an ingredient of herbal medicine. Aliment Pharmacol Ther. 2012;36(6):575–86. doi: 10.1111/j.1365-2036.2012.05221.x. [DOI] [PubMed] [Google Scholar]
  • 29.Wen Y, Chen YW, Meng AH, Zhao M, Fang SH, Ma YQ. Idiopathic mesenteric phlebosclerosis associated with long-term oral intake of geniposide. World J Gastroenterol. 2021;27(22):3097–3108. doi: 10.3748/wjg.v27.i22.3097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sakaeda T, Tamon A, Kadoyama K, Okuno Y. Data mining of the public version of the FDA Adverse Event Reporting System. Int J Med Sci. 2013;10(7):796–803. doi: 10.7150/ijms.6048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bate A, Evans SJ. Quantitative signal detection using spontaneous ADR reporting. Pharmacoepidemiol Drug Saf. 2009;18(6):427–36. doi: 10.1002/pds.1742. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The datasets generated and analyzed during the current study are available from the corresponding author on reasonable request.


Articles from Asian Pacific Journal of Cancer Prevention : APJCP are provided here courtesy of West Asia Organization for Cancer Prevention

RESOURCES