Next Article in Journal
Quantitative Spatial Characterization of Lymph Node Tumor for N Stage Improvement of Nasopharyngeal Carcinoma Patients
Next Article in Special Issue
Evaluation of 8% Capsaicin Patches in Chemotherapy-Induced Peripheral Neuropathy: A Retrospective Study in a Comprehensive Cancer Center
Previous Article in Journal
Dynamic Prediction of Resectability for Patients with Advanced Ovarian Cancer Undergoing Neo-Adjuvant Chemotherapy: Application of Joint Model for Longitudinal CA-125 Levels
Previous Article in Special Issue
BRAF and MEK Inhibitors and Their Toxicities: A Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Association of Pulmonary Sepsis and Immune Checkpoint Inhibitors: A Pharmacovigilance Study

1
Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China
2
International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha 410011, China
3
Toxicology Counseling Center of Hunan Province (TCCH), Changsha 410011, China
4
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
5
Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
6
Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
7
Laboratory of Clinical Pharmacy, Gifu Pharmaceutical University, Gifu 501-1196, Japan
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(1), 240; https://doi.org/10.3390/cancers15010240
Submission received: 28 November 2022 / Revised: 23 December 2022 / Accepted: 26 December 2022 / Published: 30 December 2022
(This article belongs to the Special Issue Side Effects of Anticancer Therapy: Prevention and Management)

Abstract

:

Simple Summary

This study investigates the association of pulmonary sepsis with immune checkpoint inhibitors by conducting an analysis using data from the Food and Drug Administration pharmacovigilance database. Compared to chemotherapy or targeted therapy, a robust signal emerged for nivolumab and atezolizumab. Co-administration of immune checkpoint inhibitors and glucocorticoids or proton pump inhibitors synergistically increased the risk of pulmonary sepsis. These signals should promote both prospective research and multidisciplinary proactive monitoring by healthcare professionals.

Abstract

Background: Although some sepsis cases were reported with immune checkpoint inhibitors (ICIs) in clinical trials, the link between pulmonary sepsis and ICIs remains mostly unknown. We aim to investigate the association between pulmonary sepsis and ICIs, and to describe the clinical features. Methods: A disproportionality analysis was performed using FAERS data and compared rates of pulmonary sepsis in cancer patients receiving ICIs vs. other drug regimens (such as chemotherapy and targeted therapy). Associations between ICIs and sepsis were assessed using reporting odds ratios (ROR) and information component (IC). We also detected drug interaction signals based on the Ω shrinkage measure. Age and gender distribution were compared between pulmonary sepsis and all adverse events associated with ICIs. Results: We identified 120 reports of pulmonary sepsis associated with ICIs between Q1, 2011 to Q3, 2021. A total of 82 of 120 (68.3%) patients on ICIs suffered from pulmonary sepsis and progressed to death. In addition, there is no significant difference in age and gender in the occurrence of pulmonary sepsis in cancer patients on ICIs. Overall ICIs, nivolumab, and atezolizumab still have a significant signal of pulmonary sepsis (ROR025 > 1, IC025 > 0, p < 0.001) compared with targeted therapy (such as tyrosine kinase inhibitors) or chemotherapy. Co-administration of ICIs and glucocorticoids or proton pump inhibitors synergistically increased the risk of pulmonary sepsis (Ω025 > 0). Conclusions: Our study suggested ICIs, especially nivolumab and atezolizumab, tended to increase the risk of pulmonary sepsis more than other anticancer regimens. Clinicians should be vigilant in the prevention and management of pulmonary sepsis during ICIs therapy.

1. Introduction

Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape of numerous cancers, generating durable responses in many patients [1]. Programmed cell death 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) are co-inhibitory receptors expressed on the surface of T cells to negatively regulate T cell-mediated immune responses; however, tumor cells exploit these inhibitory molecules to induce tumor tolerance and T cell exhaustion. Accordingly, ICIs such as anti-CTLA-4, anti-PD-1, and anti-PD-L1 can attach to these co-inhibitory receptors, thereby reactivating the immune response against tumor cells [2]. LAG-3 is a transmembrane protein involved in cytokine release and inhibitory signaling in T cells. Preclinical data showed that LAG-3 is a negative regulator of both the CD4 T cell and CD8 T cell and the activity on the CD8 T cell is independent of CD4 activation. On the CD8 T cell, LAG-3 activation abrogates the antigen presentation, whereas arrests the S phase of the cell cycle on the CD4 T cell. Based on that, the inhibition of LAG-3 is relevant and could have promising clinical benefits in treating several solid tumors [3]. The FDA approved ICI regimens including PD-1 inhibitors: nivolumab, pembrolizumab, cemiplimab, dostarlimab; PD-L1 inhibitors: atezolizumab, avelumab, durvalumab; CTLA-4 inhibitors: ipilimumab, tremelimumab; LAG-3 inhibitors: relatlimab; and combination therapy of ICIs (ipilimumab and nivolumab). Toxic effects from these ICIs agents are related to removing nodes of self-tolerance and unleashing autoimmune-like phenomena [4]. Although usually manageable with corticosteroid and immunosuppressants administration, clinically severe events leading to morbidity and even mortality may complicate ICIs treatment [5].
Sepsis is a life-threatening organ dysfunction resulting from dysregulated host responses to infection [6]. The Sequential Organ Failure Assessment (SOFA) score is used to codify the degree of organ dysfunction [7]. Sepsis is a common condition that is associated with disproportionately high mortality and, for many of those who survive, long-term morbidity. The World Health Organization (WHO) made sepsis a global health priority in 2017 and has adopted a resolution to improve the prevention, diagnosis, and management of sepsis [8]. A recent study [9] showed that cancer patients with sepsis have a higher mortality rate than non-cancer patients. During sepsis, the primary site of infection is the lung (67.4%), followed by the abdomen (20%) [10]. Pulmonary sepsis is usually characterized by hypoxemia and impaired gas exchange. It is generally referred to as acute lung injury (ALI), which further results in acute respiratory distress syndrome (ARDS), the severe form [11]. A recent meta-analysis [12] identified five cases of sepsis, ranked the second reason for fatal adverse events associated with PD-L1 inhibitors. Another case [13] reported grade 5 sepsis occurred with pembrolizumab (PD-1 inhibitor) and caused death in the KEYNOTE-028 study. The data from clinical trials with strict inclusion criteria and cohorts with limited sample sizes may not sufficiently represent the real clinical setting. In addition, there is no study that analyzed the link between pulmonary sepsis and immune checkpoint inhibitors.
Given the widespread use of ICIs in clinical practice and the potentially life-threatening nature of sepsis, it is critical for clinicians to realize the safety concern and clinical manifestations of sepsis correlated with ICIs. This pharmacovigilance study aims to investigate the potential association between pulmonary sepsis and ICIs and characterize the main features of pulmonary sepsis with ICIs in the FAERS database.

2. Methods

2.1. Study Design and Data Sources

This retrospective pharmacovigilance study is a disproportionality analysis based on deidentified individual case safety reports (ICSRs) in FAERS, the FDA’s Adverse Events Reporting System, which allows for the signal detection and quantification of the association between drugs and reporting of AEs (adverse effects). We used AERSMine [14], a validated web-based platform that analyzes FAERS reports for AEs association with drugs, indications, demographics (age and gender), and reporters. Several studies [15,16] have used AERSMine to analyze FAERS data, including a recent study that combined clinical cardiotoxicity of kinase inhibitors with cell line-derived transcriptomic datasets to identify a gene signature that can predict the risk of cardiotoxicity [17]. Ethical approval was not required because this study was conducted by using deidentified data.

2.2. Procedures

A pharmacovigilance study was conducted from 2011 Q1 (because ipilimumab was the first ICI approved by FDA on 25 March 2011) to 2021 Q3 with the FAERS data in AERSMine to evaluate the risk of pulmonary sepsis correlated with ICIs in a large-scale population. We included eight FDA-approved ICI regents (nivolumab, pembrolizumab, ipilimumab, atezolizumab, avelumab, durvalumab, dostarlimab, cemiplimab, because tremelimumab and relatlimab were recently approved by the FDA and cases were scarce) and one ICI combination therapy (nivolumab plus ipilimumab). Firstly, we analyzed the signals of all PT (preferred terms) under sepsis (SMQ, Standardized MedDRA Query, narrow) according to the Medical Dictionary for Regulatory Activities (MedDRA 25.0). Detailed PT terms could be found in supplementary file S1. Only case numbers of more than five were included in this study. We used case/non-case analysis to analyze if sepsis was differentially reported with ICIs as compared to other drugs in the full database. Then, we selected the preferred term under sepsis (SMQ) with the strongest signal and displayed detailed clinical characteristics, which included age, gender, indication, outcome and co-treatment drugs.
To assess the robustness of disproportionality signals between immune checkpoint inhibition and sepsis and account for underlying confounders of the drug-event association, we selected the preferred term under sepsis (SMQ) with the strongest signal, and then compared the safety signal among ICIs and other traditional cancer regimens, such as chemotherapy and targeted therapy, as a comparator (to reduce confounding by indication and provide a clinical perspective). First, we identified relevant NCCN (National Comprehensive Cancer Network) guidelines (list can be found in supplementary file S1) according to FDA-approved indications of ICIs. Then we extracted chemotherapy and targeted therapy from those selected NCCN guidelines. AERSMine was used to analyze safety signal variation among different regimens. Previous studies [18,19] showed that concomitant medications, such as steroids, proton pump inhibitors (PPI), and antibiotics, might affect clinical outcomes with immune checkpoint inhibitors. We inferred that the drug–drug interaction (DDI) between ICIs and these aforementioned drugs may also affect sepsis safety signals. Therefore, we conducted signal detection for drug–drug interaction between ICIs and other adjuvant drugs.

2.3. Statistical Analysis

In this study, safety signals were used as indicators of disproportionality in the reporting odds ratio (ROR) [20] based on the frequentist statistical method; and the information component (IC) [21] based on the Bayesian statistical method used at the Uppsala Monitoring Centre (UMC). The detection criterion was the lower limit of the 95% confidence interval (CI) of ROR(ROR025) > 1 [20], and the lower limit of the 95% confidence interval of IC (IC025) > 0 [21]. Norén et al. [22] put forth shrinkage observed-to-expected ratios, which added effective protection against spurious associations in signal detection. This IC and ROR approach has recently been proven effective to characterize the spectrum and characteristics of neurologic toxicity of checkpoint inhibitors [23]. Many algorithms [24] have also been reported to search for drug–drug interaction (DDI) signals. Among them, the Ω shrinkage [25] measure used by the UMC [26] has shown that it has the most conservative detection trend in the previous study [27]. The detection criterion is the lower limit of the 95% confidence interval of the Ω (Ω025) > 0. The detailed calculation process of ROR, IC, and Ω can be found in supplementary file S1. Differences in categorical variables were assessed using a chi-squared test of independence performed on a 2 × 2 contingency table with Yates’ continuity correction or Fisher’s exact test. Significance was assumed when the p value was less than 0.05. All data analyses were performed independently by two authors and statistical analyses and calculations were performed with IBM SPSS Statistics for Windows, Version 26.0. Armonk, NY: (IBM, Chicago, IL, USA), and Microsoft Excel 2021 (Microsoft Corporation, Redmond, WA, USA).

3. Results

3.1. Sepsis Signal Detected Using FAERS Database

From a total number of 215,363 case reports of patients on ICIs in the full FAERS between 2011 (Q1) and 2021 (Q3), we detected 7535 cases of PT terms under sepsis (SMQ). Compared with all other drugs in the FAERS database, ICIs have a significant safety signal of sepsis (SMQ) (ROR025 2.72, IC025 1.37). Moreover, the pulmonary sepsis attributes the strongest signal (ROR025 6.43, IC025 2.48) among all detected PT terms under sepsis (SMQ). Sepsis toxicities associated with immune checkpoint inhibitors are detailed in Table 1.
Then, we assessed the robustness of the association of pulmonary sepsis with ICIs (nivolumab, pembrolizumab, ipilimumab, atezolizumab, durvalumab, nivolumab plus ipilimumab were included in the subsequent analysis because the case number of dostarlimab, avelumab, and cemiplimab is less than 5). When compared with other anti-cancer drugs, overall ICIs (ROR025 2.50, IC025 1.03, p < 0.001), nivolumab (ROR025 3.59, IC025 1.60, p < 0.001), atezolizumab (ROR025 1.80, IC025 0.57, p < 0.001) showed a significant safety concern of pulmonary sepsis. When compared with targeted therapy (such as tyrosine kinase inhibitors) or chemotherapy extracted from NCCN’s guideline, overall ICIs, nivolumab, and atezolizumab still have a significant signal of pulmonary sepsis (ROR025 > 1, IC025 > 0, p < 0.001) (Figure 1).

3.2. Clinical Features of Pulmonary Sepsis

120 cases of pulmonary sepsis were detected in patients on ICIs. A total of 73 of 120 (60.8%) cases were from patients on nivolumab. A total of 85.8% of cases were reported during 2018–2021. A total of 83 of 120 (69.2%) pulmonary sepsis cases were reported by health professionals such as doctors or pharmacists. Regarding indications, 40 of 120 (33.3%) cases were reported as lung cancer. A total of 48 of 120 (40.0%) pulmonary sepsis cases received ICIs and glucocorticoids/corticosteroids, and 44 of 120 (36.7%) cases received proton pump inhibitors during ICIs therapies (Table 2). After comparing the age of pulmonary sepsis (age of more than 65) associated with ICIs against the same age range of any reported adverse events of this drug class, we did not detect any difference of signals among various age periods. Although, the overall ICIs (IC025 0.06, p < 0.001) showed that males have a higher safety concern of pulmonary sepsis than females. However, we did not find any difference of pulmonary sepsis signals between males and females for any specific kind of ICIs (Figure 2). A total of 82 of 120 (68.3%) cases suffering from pulmonary sepsis progressed to death for patients on ICIs. A total of 31 of 120 (25.8%) cases experienced a life-threatening situation.

3.3. Drug-Drug Interaction Signal Detection

We identified that co-administration of nivolumab (Ω025 = 0.91), ipilimumab (Ω025 = 0.77), nivolumab plus ipilimumab (Ω025 = 1.04) with glucocorticoids or corticosteroids have an elevated safety concern of pulmonary sepsis. In addition, the co-treatment of nivolumab (Ω025 = 1.00), nivolumab plus ipilimumab (Ω025 = 0.17) with proton pump inhibitors may also synergistically increase the risk of pulmonary sepsis (Table 3).
When Ω025 > 0, a significant drug–drug interaction signal was detected. The detailed calculation process of Ω025 can be found in supplementary file S1 and raw data could be found in supplementary file S2.

4. Discussion

To the best of our knowledge, this is the first large-scale pharmacovigilance study on pulmonary sepsis associated with ICIs leveraging the FAERS database. In general, there were three key findings in our study. Firstly, our disproportionality analyses suggested ICIs may increase the risk of pulmonary sepsis compared with other anti-cancer agents such as chemotherapy or targeted therapy. Secondly, we investigated the detailed safety profile and clinical features of pulmonary sepsis. Finally, we identified the potential medications which would increase the risk of pulmonary sepsis when co-administrated with ICIs through drug–drug interaction signals detection.
In this study, our analysis showed a significant incidence of pulmonary sepsis associated with ICIs, suggesting that pulmonary sepsis may be underrepresented in the published literature. As of 28 November 2022, no peer-reviewed observational studies, meta-analysis, or reviews were published related to the underlying association between ICIs and pulmonary sepsis. Although, previous clinical trial data [12] showed that sepsis is the second main reason leading to death in patients who received PD-L1 inhibitors, sepsis is not on the current NCCN guidelines for ICIs-related toxicity management [28]. By conducting a retrospective large-scale pharmacovigilance analysis, we detected that ICIs (case), especially nivolumab and atezolizumab, tended to increase the risk of pulmonary sepsis compared with non-case (other anticancer regimens included chemotherapy and targeted therapy).
Few pieces of literature demonstrated the clinical features of pulmonary sepsis associated with ICIs. We found 85.8% of pulmonary sepsis cases were reported since 2018, which reflected the accompanying safety concern of the increasing application of immune checkpoint inhibitors in cancers. A total of 25.8% of cases experience a life-threatening situation when pulmonary sepsis occurred and 68.3% of cases are finally deceased, indicating the severe outcome of this type of toxicity. A total of 60.8% of pulmonary sepsis cases were from patients on nivolumab. In addition, a study [29] showed that 62.5% of tuberculosis and 69.3% of atypical mycobacterial infections were induced by nivolumab. Our post-marketing large-scale pharmacovigilance analysis supports that PD-1 inhibitors nivolumab increase the incidence of pulmonary sepsis. We did not detect a significant difference in age and gender in the occurrence of pulmonary sepsis in cancer patients on ICIs. Further research is warranted to investigate the influence of age and gender on the occurrence of pulmonary sepsis associated with ICIs.
We identified 40.0% and 36.7% of patients suffered from pulmonary sepsis received glucocorticoids/corticosteroids or proton pump inhibitors when they were on ICIs, respectively, indicating the underlying influence of polypharmacy on the reported frequency of pulmonary sepsis. A large multicenter integrated analysis [18] showed that baseline steroids, systemic antibiotics, and proton pump inhibitors were associated with worse clinical outcomes in patients receiving ICIs. We further detected drug–drug interaction signals between ICIs and the aforementioned adjuvant medications. Our results indicated that co-administration of ICIs and glucocorticoids or corticosteroids increased the pulmonary sepsis report frequency in patients on nivolumab, ipilimumab, or nivolumab plus ipilimumab, which was in line with previous studies [30,31]. It is interesting that interaction signals of nivolumab, nivolumab plus ipilimumab, and proton pump inhibitors were detected (Table 3). Several previous studies [32,33] suggest that proton pump inhibitors negatively influence the magnitude of ICI efficacy and may increase the risk of death, which maybe results from severe alterations to the gut microbiome because of long-term use of PPIs. Regarding to the influence of proton pump inhibitors on the occurrence of pulmonary sepsis, we believe there are some possible reasons. First, proton pump inhibitors changes gut microbiota and subsequently increases sepsis susceptibility by following underlying mechanism: allowing for expansion of pathogenic intestinal bacteria, priming the immune system for a robust pro-inflammatory response, and decreasing production of beneficial microbial products such as short-chain fatty acids [34]. Second, proton pump inhibitors change upper gastrointestinal environment allows colonization of the oropharynx by gastrointestinal bacteria, which could increase the risk of pneumonia and even progress to sepsis [35]. Our study is the first to highlight that the combination of PPIs and ICIs not only negatively affects efficacy but also can increase rates of pulmonary sepsis. Therefore, physicians need to use PPIs carefully when patients are receiving ICIs therapy.
Regarding the potential mechanism of pulmonary sepsis associated with ICIs, bacteria are the pathogens most associated with pulmonary infection and sepsis; however, fungi viruses can also act as a source of infection [11]. A previous review [36] summarized that there are two main reasons for infection events associated with immune checkpoint inhibitors: one is opportunistic infections associated with immune-related adverse events treatment (such as glucocorticoids, corticosteroids or immunosuppressants); the other one is infections due to dysregulated immunity, such as cases of atypical mycobacterium infection following PD-1/PD-L1 immunotherapy in the absence of immunosuppression [37]. In addition, those infection events that occurred resulted from the aforementioned two pathways, which may progress to pulmonary sepsis without appropriate management and treatment.
Our pharmacovigilance analysis showed an increased reporting frequency of pulmonary sepsis associated with immune checkpoint inhibitors. Further preclinical and clinical studies are warranted to validate our results and confirm the link between pulmonary sepsis and ICIs.

5. Limitations

There are several limitations of the study that are intrinsic to FAERS [38]. First, adverse event reporting is voluntary and comes from heterogeneous sources, thus raising the possibility of incomplete information or underreporting. However, cases in the FAERS database cover many countries in the world, thus ensuring an unparalleled global assessment in diverse clinical settings. Second, detailed clinical information and diagnostic criteria are unavailable, thus limiting our assessment to those reports. Third, we are unable to definitively determine the incidence of each event using FAERS and only generate hypotheses. As with other pharmacovigilance studies, this study allows for signal detection and generation in a large population, which will need prospective and long-term validation of findings.

6. Conclusions

This real-world pharmacovigilance analysis of the FAERS database first identified that pulmonary sepsis was significantly associated with nivolumab and atezolizumab. In addition, when ICIs were co-administrated with glucocorticoids or proton pump inhibitors, the safety concern of pulmonary sepsis increased. Further studies need to be conducted to confirm the association, explore the underlying mechanisms, and address management strategies for pulmonary sepsis.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers15010240/s1, supplementary file S1: Calculation formula, preferred terms and NCCN guidelines; supplementary file S2: drug interaction ICIs and other drugs; supplementary file S3: AERSMine load files.

Author Contributions

Formal analysis, S.X.; data curation, S.X.; writing—original draft, S.X.; review and editing, S.X., M.S. and Y.N.; visualization S.X.; software, M.S. and Y.N.; methodology, M.Y., M.S., and Y.N.; resources, L.G. and Y.Z.; software, H.G. and Y.W.; supervision, B.Z.; conceptualization, M.Y. All authors participated in the interpretation of the results. The final manuscript was read, checked, and approved by all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Health Commission of Hunan Province with fund number (202113012480).

Institutional Review Board Statement

All clinical data were de-identified and are in a publicly available database (FAERS). Ethics approval and consent are not needed.

Informed Consent Statement

Patient consent was waived because the study is based on anonymous data that can be downloaded from a publicly available source.

Data Availability Statement

The datasets analyzed during the current study are available in the following resource, which is available in the public domain: https://research.cchmc.org/aers/ (accessed on 26th November 2022) (AERSMine, a multi-cohort analyzing application designed to mine data across millions of patient reports (currently 16,849,672) from the FDA’s Adverse Event Reporting System).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef] [PubMed]
  2. Shiravand, Y.; Khodadadi, F.; Kashani, S.M.A.; Hosseini-Fard, S.R.; Hosseini, S.; Sadeghirad, H.; Ladwa, R.; O’Byrne, K.; Kulasinghe, A. Immune Checkpoint Inhibitors in Cancer Therapy. Curr. Oncol. 2022, 29, 3044–3060. [Google Scholar] [CrossRef] [PubMed]
  3. Aroldi, F.; Saleh, R.; Jafferji, I.; Barreto, C.; Saberian, C.; Middleton, M.R. Lag3: From Bench to Bedside. Cancer Treat. Res. 2022, 183, 185–199. [Google Scholar] [CrossRef]
  4. Postow, M.A.; Sidlow, R.; Hellmann, M.D. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N. Engl. J. Med. 2018, 378, 158–168. [Google Scholar] [CrossRef]
  5. Wang, D.Y.; Salem, J.E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol 2018, 4, 1721–1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef] [PubMed]
  7. Vincent, J.L.; Moreno, R.; Takala, J.; Willatts, S.; De Mendonca, A.; Bruining, H.; Reinhart, C.K.; Suter, P.M.; Thijs, L.G. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. On behalf of the Working Group on Sepsis-Related Problems of the European Society of Intensive Care Medicine. Intensive Care Med. 1996, 22, 707–710. [Google Scholar] [CrossRef]
  8. Cecconi, M.; Evans, L.; Levy, M.; Rhodes, A. Sepsis and septic shock. Lancet 2018, 392, 75–87. [Google Scholar] [CrossRef]
  9. Hensley, M.K.; Donnelly, J.P.; Carlton, E.F.; Prescott, H.C. Epidemiology and Outcomes of Cancer-Related Versus Non-Cancer-Related Sepsis Hospitalizations. Crit. Care Med. 2019, 47, 1310–1316. [Google Scholar] [CrossRef]
  10. Gotts, J.E.; Matthay, M.A. Sepsis: Pathophysiology and clinical management. BMJ 2016, 353, i1585. [Google Scholar] [CrossRef]
  11. Mohsin, M.; Tabassum, G.; Ahmad, S.; Ali, S.; Ali Syed, M. The role of mitophagy in pulmonary sepsis. Mitochondrion 2021, 59, 63–75. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, X.; Wu, S.; Chen, Y.; Shao, E.; Zhuang, T.; Lu, L.; Chen, X. Fatal Adverse Events Associated With Programmed Cell Death Ligand 1 Inhibitors: A Systematic Review and Meta-Analysis. Front. Pharmacol. 2020, 11, 5. [Google Scholar] [CrossRef] [Green Version]
  13. Hsu, C.; Lee, S.H.; Ejadi, S.; Even, C.; Cohen, R.B.; Le Tourneau, C.; Mehnert, J.M.; Algazi, A.; van Brummelen, E.M.J.; Saraf, S.; et al. Safety and Antitumor Activity of Pembrolizumab in Patients With Programmed Death-Ligand 1-Positive Nasopharyngeal Carcinoma: Results of the KEYNOTE-028 Study. J. Clin. Oncol. 2017, 35, 4050–4056. [Google Scholar] [CrossRef]
  14. Sarangdhar, M.; Tabar, S.; Schmidt, C.; Kushwaha, A.; Shah, K.; Dahlquist, J.E.; Jegga, A.G.; Aronow, B.J. Data mining differential clinical outcomes associated with drug regimens using adverse event reporting data. Nat. Biotechnol. 2016, 34, 697–700. [Google Scholar] [CrossRef]
  15. Fadini, G.P.; Bonora, B.M.; Mayur, S.; Rigato, M.; Avogaro, A. Dipeptidyl peptidase-4 inhibitors moderate the risk of genitourinary tract infections associated with sodium-glucose co-transporter-2 inhibitors. Diabetes Obes. Metab. 2018, 20, 740–744. [Google Scholar] [CrossRef] [PubMed]
  16. Xia, S.; Zhao, Y.C.; Guo, L.; Gong, H.; Wang, Y.K.; Ma, R.; Zhang, B.K.; Sheng, Y.; Sarangdhar, M.; Noguchi, Y.; et al. Do antibody-drug conjugates increase the risk of sepsis in cancer patients? A pharmacovigilance study. Front. Pharmacol. 2022, 13, 967017. [Google Scholar] [CrossRef] [PubMed]
  17. van Hasselt, J.G.C.; Rahman, R.; Hansen, J.; Stern, A.; Shim, J.V.; Xiong, Y.; Pickard, A.; Jayaraman, G.; Hu, B.; Mahajan, M.; et al. Transcriptomic profiling of human cardiac cells predicts protein kinase inhibitor-associated cardiotoxicity. Nat. Commun. 2020, 11, 4809. [Google Scholar] [CrossRef] [PubMed]
  18. Cortellini, A.; Tucci, M.; Adamo, V.; Stucci, L.S.; Russo, A.; Tanda, E.T.; Spagnolo, F.; Rastelli, F.; Bisonni, R.; Santini, D.; et al. Integrated analysis of concomitant medications and oncological outcomes from PD-1/PD-L1 checkpoint inhibitors in clinical practice. J. Immunother. Cancer 2020, 8, e001361. [Google Scholar] [CrossRef]
  19. Kostine, M.; Mauric, E.; Tison, A.; Barnetche, T.; Barre, A.; Nikolski, M.; Rouxel, L.; Dutriaux, C.; Dousset, L.; Prey, S.; et al. Baseline co-medications may alter the anti-tumoural effect of checkpoint inhibitors as well as the risk of immune-related adverse events. Eur. J. Cancer 2021, 157, 474–484. [Google Scholar] [CrossRef]
  20. Rothman, K.J.; Lanes, S.; Sacks, S.T. The reporting odds ratio and its advantages over the proportional reporting ratio. Pharmacoepidemiol. Drug Saf. 2004, 13, 519–523. [Google Scholar] [CrossRef]
  21. Bate, A.; Lindquist, M.; Edwards, I.R.; Olsson, S.; Orre, R.; Lansner, A.; De Freitas, R.M. A Bayesian neural network method for adverse drug reaction signal generation. Eur. J. Clin. Pharmacol. 1998, 54, 315–321. [Google Scholar] [CrossRef] [PubMed]
  22. Noren, G.N.; Hopstadius, J.; Bate, A. Shrinkage observed-to-expected ratios for robust and transparent large-scale pattern discovery. Stat. Methods Med. Res. 2013, 22, 57–69. [Google Scholar] [CrossRef]
  23. Johnson, D.B.; Manouchehri, A.; Haugh, A.M.; Quach, H.T.; Balko, J.M.; Lebrun-Vignes, B.; Mammen, A.; Moslehi, J.J.; Salem, J.E. Neurologic toxicity associated with immune checkpoint inhibitors: A pharmacovigilance study. J. Immunother. Cancer 2019, 7, 134. [Google Scholar] [CrossRef] [Green Version]
  24. Noguchi, Y.; Tachi, T.; Teramachi, H. Review of Statistical Methodologies for Detecting Drug-Drug Interactions Using Spontaneous Reporting Systems. Front. Pharmacol. 2019, 10, 1319. [Google Scholar] [CrossRef]
  25. Noren, G.N.; Sundberg, R.; Bate, A.; Edwards, I.R. A statistical methodology for drug-drug interaction surveillance. Stat. Med. 2008, 27, 3057–3070. [Google Scholar] [CrossRef]
  26. UMC. The UMC Measures of Disproportionate Reporting. 2016. Available online: https://www.who-umc.org/media/164041/measures-of-disproportionate-reporting_2016.pdf (accessed on 10 April 2022).
  27. Noguchi, Y.; Tachi, T.; Teramachi, H. Comparison of Signal Detection Algorithms Based on Frequency Statistical Model for Drug-Drug Interaction Using Spontaneous Reporting Systems. Pharm. Res. 2020, 37, 86. [Google Scholar] [CrossRef]
  28. Thompson, J.A.; Schneider, B.J.; Brahmer, J.; Achufusi, A.; Armand, P.; Berkenstock, M.K.; Bhatia, S.; Budde, L.E.; Chokshi, S.; Davies, M.; et al. Management of Immunotherapy-Related Toxicities, Version 1.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2022, 20, 387–405. [Google Scholar] [CrossRef]
  29. Anand, K.; Sahu, G.; Burns, E.; Ensor, A.; Ensor, J.; Pingali, S.R.; Subbiah, V.; Iyer, S.P. Mycobacterial infections due to PD-1 and PD-L1 checkpoint inhibitors. ESMO Open 2020, 5, e000866. [Google Scholar] [CrossRef]
  30. Sorup, S.; Darvalics, B.; Russo, L.; Oksen, D.; Lamy, F.X.; Verpillat, P.; Aa, K.; Ht, S.; Cronin-Fenton, D. High-dose corticosteroid use and risk of hospitalization for infection in patients treated with immune checkpoint inhibitors--A nationwide register-based cohort study. Cancer Med. 2021, 10, 4957–4963. [Google Scholar] [CrossRef]
  31. Karam, J.D.; Noel, N.; Voisin, A.L.; Lanoy, E.; Michot, J.M.; Lambotte, O. Infectious complications in patients treated with immune checkpoint inhibitors. Eur. J. Cancer 2020, 141, 137–142. [Google Scholar] [CrossRef]
  32. Qin, B.D.; Jiao, X.D.; Zhou, X.C.; Shi, B.; Wang, J.; Liu, K.; Wu, Y.; Ling, Y.; Zang, Y.S. Effects of concomitant proton pump inhibitor use on immune checkpoint inhibitor efficacy among patients with advanced cancer. Oncoimmunology 2021, 10, 1929727. [Google Scholar] [CrossRef]
  33. Chalabi, M.; Cardona, A.; Nagarkar, D.R.; Dhawahir Scala, A.; Gandara, D.R.; Rittmeyer, A.; Albert, M.L.; Powles, T.; Kok, M.; Herrera, F.G. Efficacy of chemotherapy and atezolizumab in patients with non-small-cell lung cancer receiving antibiotics and proton pump inhibitors: Pooled post hoc analyses of the OAK and POPLAR trials. Ann. Oncol. 2020, 31, 525–531. [Google Scholar] [CrossRef] [Green Version]
  34. Adelman, M.W.; Woodworth, M.H.; Langelier, C.; Busch, L.M.; Kempker, J.A.; Kraft, C.S.; Martin, G.S. The gut microbiome’s role in the development, maintenance, and outcomes of sepsis. Crit. Care. 2020, 24, 278. [Google Scholar] [CrossRef]
  35. Meijvis, S.C.; Cornips, M.C.; Voorn, G.P.; Souverein, P.C.; Endeman, H.; Biesma, D.H.; Leufkens, H.G.; van de Garde, E.M. Microbial evaluation of proton-pump inhibitors and the risk of pneumonia. Eur. Respir. J. 2011, 38, 1165–1172. [Google Scholar] [CrossRef] [Green Version]
  36. Morelli, T.; Fujita, K.; Redelman-Sidi, G.; Elkington, P.T. Infections due to dysregulated immunity: An emerging complication of cancer immunotherapy. Thorax 2022, 77, 304–311. [Google Scholar] [CrossRef]
  37. Fujita, K.; Yamamoto, Y.; Kanai, O.; Okamura, M.; Nakatani, K.; Mio, T. Development of Mycobacterium avium Complex Lung Disease in Patients With Lung Cancer on Immune Checkpoint Inhibitors. Open Forum Infect. Dis. 2020, 7, ofaa067. [Google Scholar] [CrossRef] [Green Version]
  38. Noguchi, Y.; Tachi, T.; Teramachi, H. Detection algorithms and attentive points of safety signal using spontaneous reporting systems as a clinical data source. Brief. Bioinform. 2021, 22, bbab347. [Google Scholar] [CrossRef]
Figure 1. The comparison of pulmonary sepsis signal between ICIs and controls (other anticancer drugs, targeted therapy, chemotherapy) in FAERS database. The list of the control group (targeted therapy and chemotherapy) was extracted from NCCN guidelines for ICIs’ indication. More details could be found in the AERSMine load files in the supplementary file S3. Abbreviation: ROR, reporting odds ratio. IC, information component. 95%CI, 95% confidence interval. N, number. AEs, adverse events. ICIs, immune checkpoint inhibitors. NCCN, National Comprehensive Cancer Network.
Figure 1. The comparison of pulmonary sepsis signal between ICIs and controls (other anticancer drugs, targeted therapy, chemotherapy) in FAERS database. The list of the control group (targeted therapy and chemotherapy) was extracted from NCCN guidelines for ICIs’ indication. More details could be found in the AERSMine load files in the supplementary file S3. Abbreviation: ROR, reporting odds ratio. IC, information component. 95%CI, 95% confidence interval. N, number. AEs, adverse events. ICIs, immune checkpoint inhibitors. NCCN, National Comprehensive Cancer Network.
Cancers 15 00240 g001
Figure 2. Age and gender distribution in pulmonary sepsis cases of ICIs. ROR, reporting odds ratio. IC, information component. 95%CI, 95% confidence interval. AEs, adverse events. ICIs, immune checkpoint inhibitors. N/A, not applicable. (Because the case number < 3, ROR could not be calculated).
Figure 2. Age and gender distribution in pulmonary sepsis cases of ICIs. ROR, reporting odds ratio. IC, information component. 95%CI, 95% confidence interval. AEs, adverse events. ICIs, immune checkpoint inhibitors. N/A, not applicable. (Because the case number < 3, ROR could not be calculated).
Cancers 15 00240 g002
Table 1. Checkpoint inhibitor-related sepsis reported with ICIs versus those reported in the full database from FAERS, from Q1, 2011 to Q3, 2021.
Table 1. Checkpoint inhibitor-related sepsis reported with ICIs versus those reported in the full database from FAERS, from Q1, 2011 to Q3, 2021.
CategoriesOverall ICIsFull Database
(Starting Q1,2011)
ROR (95%CI)IC (95%CI)
Total number of ICSRs available215,36313,943,677
Number of ICSRs by Sepsis subgroups
Sepsis (SMQ)7535184,0802.78 (2.72–2.85)1.41 (1.37–1.43)
Sepsis359982,046 2.96 (2.86–3.06)1.51 (1.45–1.55)
Multiple organ dysfunction syndrome109431,5102.30 (2.16–2.44)1.17 (1.07–1.24)
Bacteraemia31278502.64 (2.36–2.96)1.36 (1.17–1.50)
Systemic inflammatory response syndrome2523189 5.47 (4.81–6.23)2.34 (2.14–2.49)
Urosepsis2777014 2.62 (2.33–2.96)1.35 (1.15–1.49)
Neutropenic sepsis24254472.97 (2.61–3.37)1.52 (1.31–1.67)
Pulmonary sepsis12011057.77 (6.43–9.39)2.78 (2.48–3.00)
Escherichia sepsis7421232.30 (1.83–2.90)1.16 (0.78–1.44)
Escherichia bacteraemia5914562.69 (2.08–3.49)1.37 (0.94–1.68)
Systemic candida5514662.49 (1.90–3.25)1.26 (0.81–1.58)
Device related sepsis4614672.06 (1.54–2.77)1.01 (0.52–1.36)
Staphylococcal sepsis9634151.84 (1.51–2.26)0.86 (0.52–1.10)
Abdominal sepsis296373.04 (2.09–4.41)1.51 (0.89–1.95)
Post-procedural sepsis134641.84 (1.06–3.19)0.82 (−0.12–1.46)
Streptococcal sepsis267912.17 (1.47–3.20)1.06 (0.40–1.52)
Procalcitonin increased397533.48 (2.52–4.81)1.70 (1.17–2.08)
Cytomegalovirus viraemia2624480.68 (0.46–1.01)−0.53 (−1.19−(−0.07))
Blood culture positive1213120.59 (0.33–1.04)−0.73 (−1.71−(−0.06))
ICIs refer to any ICSRs reported for treatment with nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, cemiplimab, dostarlimab, ipilimumab, and their combination. Sepsis subgroups were extracted from MedDRA 25.0, sepsis (SMQ). When IC025 value (>0) or ROR025 value (>1), a significant signal of drug-AE was detected. ICSRs, individual case safety reports. ICIs, immune checkpoint inhibitors. IC, information component. IC025 = lower end of a 95% confidence interval for the IC.
Table 2. Patient characteristics of pulmonary sepsis reports with immune checkpoint inhibitors in FAERS database.
Table 2. Patient characteristics of pulmonary sepsis reports with immune checkpoint inhibitors in FAERS database.
CategoriesNivolumabPembrolizumabAtezolizumabDurvalumabIpilimumabNivolumab + IpilimumabAll ICIs
Reports of Pulmonary sepsis7391321310120
Report Year
2011–20179 (12.3%)2 (22.2%)2 (15.4%)2 (100.0%)2 (15.4%)017 (14.2%)
2018–2021 (Q3)64 (87.7%)7 (77.8%)11 (84.6%)011 (84.6%)10 (100.0%)103 (85.8%)
Reporter
Healthcare professionals51 (69.9%)
22 (31.1%)
5 (55.6%)
4 (44.4%)
13 (100.0%)
0
2 (100.0%)
0
7 (53.8%)
6 (46.2%)
9 (90.0%)
1 (10.0%)
83 (69.2%)
37 (30.8%)
Other
Age Category
0–140000000
15–242 (2.8%)000002 (1.7%)
25–6535 (49.3%)5 (55.6%)5 (38.5%)07 (53.8%)5 (50.0%)57 (48.3%)
>6534 (47.9%)4 (44.4%)8 (61.5%)2 (100.0%)6 (46.2%)5 (50.0%)59 (50.0%)
Data available71 (97.3%)9 (100.0%)13 (100.0%)2 (100.0%)13 (100.0%)10 (100.0%)118 (98.3%)
Gender
Male51 (70.8%)7 (77.8%)8 (61.5%)2 (100.0%)12 (92.3%)9 (90.0%)89 (74.8%)
Female21 (29.2%)2 (22.2%)5 (38.5)01 (7.7%)1 (10.0%)30 (25.2%)
Data available72 (98.6%)9 (100.0%)13 (100.0%)2 (100.0%)13 (100.0%)10 (100.0%)119 (99.2%)
Indication
Non-small Lung cancer17 (23.3%)5 (55.6%)2 (15.4%)1 (50.0%)0025 (19.2%)
Lung neoplasm malignant14 (19.2%)1 (11.1%)000015 (12.5%)
Other42 (57.5%)3 (33.3%)11 (84.6%)1 (50.0%)13 (100.0%)10 (100.0%)80 (66.7%)
Co-administration drugs
Glucocorticoids or corticosteroids
28 (38.4%)01 (7.7%)2 (100.0%)9 (69.2%)8 (80.0%)48 (40.0%)
proton pump inhibitors26 (35.6%)2 (22.2%)4 (92.3%)2 (100.0%)5 (38.5%)5 (50.0%)44 (36.7%)
Outcome
Death49 (67.1%)6 (66.7%)10 (76.9%)2 (100.0%)9 (69.2%)6 (60.0%)82 (68.3%)
Life-threatening15 (20.5%)2 (22.2%)2 (15.4%)1 (50.0%)6 (46.2%)5 (50.0%)31 (25.8%)
Disability1 (1.4%)000001 (0.8%)
Hospitalization64 (87.7%)9 (100.0%)13 (100.0%)2 (100.0%)10 (76.9%)8 (80.0%)106 (88.3%)
Other Serious71 (97.3%)6 (66.7%)1 (7.7%)011 (84.6%)10 (100.0%)99 (82.5%)
We included N > 5 case reports of pulmonary sepsis related to immune checkpoint inhibitors. ICIs, immune checkpoint inhibitors. Q3, quarter 3. Glucocorticoids or corticosteroids including “dexamethasone” “prednisone” “hydrocortisone” etc.Proton pump inhibitors including omeprazole, lansoprazole, esomeprazole etc. A detailed list of included drugs can be found in the AERS load files in the supplementary file S3. Note regarding patient counts—for example, the total number of outcomes for nivolumab is not equal to total drug events (73) since patients have reported more than one outcome.
Table 3. Drug–Drug interaction between ICIs and other drugs.
Table 3. Drug–Drug interaction between ICIs and other drugs.
Drug 1Drug 2AEsΩ (95%CI)
NivolumabGlucocorticoids or corticosteroidsPulmonary sepsis1.45 (0.91–1.98)
IpilimumabGlucocorticoids or corticosteroidsPulmonary sepsis1.72 (0.77–2.66)
Nivolumab plus ipilimumabGlucocorticoids or corticosteroidsPulmonary sepsis2.04 (1.04–3.04)
NivolumabProton pump inhibitorsPulmonary sepsis1.55 (1.00–2.11)
Nivolumab plus ipilimumabProton pump inhibitorsPulmonary sepsis1.44 (0.17–2.70)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xia, S.; Gong, H.; Zhao, Y.; Guo, L.; Wang, Y.; Zhang, B.; Sarangdhar, M.; Noguchi, Y.; Yan, M. Association of Pulmonary Sepsis and Immune Checkpoint Inhibitors: A Pharmacovigilance Study. Cancers 2023, 15, 240. https://doi.org/10.3390/cancers15010240

AMA Style

Xia S, Gong H, Zhao Y, Guo L, Wang Y, Zhang B, Sarangdhar M, Noguchi Y, Yan M. Association of Pulmonary Sepsis and Immune Checkpoint Inhibitors: A Pharmacovigilance Study. Cancers. 2023; 15(1):240. https://doi.org/10.3390/cancers15010240

Chicago/Turabian Style

Xia, Shuang, Hui Gong, Yichang Zhao, Lin Guo, Yikun Wang, Bikui Zhang, Mayur Sarangdhar, Yoshihiro Noguchi, and Miao Yan. 2023. "Association of Pulmonary Sepsis and Immune Checkpoint Inhibitors: A Pharmacovigilance Study" Cancers 15, no. 1: 240. https://doi.org/10.3390/cancers15010240

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop