2,279
Views
39
CrossRef citations to date
0
Altmetric
Commentary

Concomitant medications and immune checkpoint inhibitor therapy for cancer: causation or association?

, & ORCID Icon
Pages 55-61 | Received 24 Apr 2020, Accepted 10 May 2020, Published online: 23 Jun 2020

ABSTRACT

The majority of cancer patients assume concomitant medications for the treatment of cancer-related symptoms or co-morbidities. As immune checkpoint inhibitors expand in the treatment of a widening range of malignancies, drug–drug interactions have become an area of increasing interest due to the potential for some concomitant medications to exert immune-modulatory effects and influence outcomes from immunotherapy. Here, we review the evidence supporting this association across selected drug classes including antibiotics, proton pump inhibitors, metformin, and opioids.

Introduction

Immune checkpoint inhibitors (ICIs) have fast risen to the forefront in the therapeutic armamentarium against cancer. After initial evidence of activity in previously untreatable malignancies such as melanoma,Citation1 monoclonal antibodies inhibiting cytotoxic T-cell lymphocyte-associated antigen-4 (CTLA-4) and the programmed cell death-1 receptor/ligand (PD-1/PD-L1) interaction have swiftly expanded as a therapeutic option to other tumor sites including lung, bladder, renal, head and neck cancer and many others.Citation2 As the number of actionable drivers of anti-cancer immunity continues to expand, ICIs are set to play an increasingly relevant role in the management of cancer either as monotherapy or in combination with chemotherapy and targeted agents.Citation3 While capable of inducing cancer immune rejection primarily through the reversal of T-cell exhaustion,Citation4 ICIs are not universally effective and response rates are variable. Phenotypic characteristics of the tumor or the host response against malignancy have been shown to enrich for responses to immunotherapy. A number of traits including tumoral PD-L1 expression,Citation5 number of non-synonymous somatic mutations,Citation6 diversity in the host’s microbiome,Citation7 and many others have been evaluated as predictive correlates of response to ICI in clinical trials and in routine practice. However, mechanisms of primary refractoriness and acquired resistance to immunotherapy are still incompletely understood, a factor that limits the use and clinical effectiveness of ICI in the clinic.Citation8

Drug–drug interactions (DDI) represent a key area of interest in the context of systemic anti-cancer treatment. Traditionally, desired pharmacodynamic (PD) interactions between different classes of cytotoxic compounds have guided the development of polychemotherapy regimens leading to incremental oncological benefit by overcoming resistance to each chemotherapy class. On the other hand, unwanted DDIs at the pharmacokinetic (PK) level are potentially responsible for toxicity to cytotoxic and targeted anti-cancer therapeutics.Citation9 As most monoclonal antibodies, currently available ICIs have a reproducible PK profileCitation10 that is minimally influenced by concomitant therapies.Citation11

However, in view of the immune-mediated mechanism of action of ICI, considerable interest has been devoted to the investigation of how concomitant therapies with a potential immune-modulatory effect might interact with the PD of ICI, potentially modifying their efficacy or enhancing their toxicity in cancer patients.

In this commentary, we have evaluated five selected drug classes that are commonly administered alongside ICIs whose immune-modulatory effects have been postulated to interact with ICI and influence efficacy and/or toxicity outcomes, namely: antibiotics, corticosteroids, proton pump inhibitors, metformin and opioids.

Antibiotic therapy

The human body and the gut mucosal surfaces, in particular, are inhabited by over 100 trillion commensal bacteria which live in a symbiotic relationship with the host. Taxonomic features of the gut microbiota including diversity and enrichment of Faecalibacterium, Bifidobacterium and Bacteroides spp.Citation12,Citation13 have been shown to associate with the probability of response to ICI.Citation13

Broad-spectrum antibiotic therapy (ATB) can cause long-standing perturbation to the gut microbiota,Citation14 and in view of the increasing relevance of this domain in cancer immune tolerogenesis ATB has been one of the most exhaustively explored class of concomitant medications in the context of ICI therapy.Citation15

A vast number of retrospective as well as prospective studies has shown consistent and reproducible evidence of worse outcome in ATB-exposed patients across a range of malignancies and windows of exposure (). In general, the majority of studies suggest that patients treated with ATB have shorter overall (OS), progression-free survival (PFS) as well as reduced overall response rates (ORR) compared to ATB-unexposed patients.

Table 1. The relationship between selected classes of concomitant medications and immune checkpoint inhibitors for cancer therapy

A key challenge in interpreting these data is to disentangle the mechanistic role of ATB exposure from the potential confounding effect of patient’s fitness, co-morbid burden and underlying biology of cancer in dictating outcomes. Patients receiving ATB often have poorer performance status than those who do not,Citation17 suggesting that ATB use might mask features of more aggressive disease that are likely to influence outcome independent of the underlying microbiome-modulating effect of ATBs. Patients with poorer baseline reserve may be more likely to acquire life-shortening infectious complications.

Timing of ATB therapy appears crucial with regards to their influence on outcomes from ICI therapy. In a previous study by our group,Citation16 we showed that ATB therapy had a significantly detrimental impact on OS (2 vs 26 months, HR = 7.4; 95% CI: 4.2–12.9; p < .001) only if given within 30 d from ICI commencement, with no significant difference in OS seen in patients who received ATB concurrently with ICI. Importantly, prior ATB therapy was associated with an increased proportion of ICI-refractoriness evaluated radiologically, lending further credence to the hypothesis that ATB might exert a “priming” effect on cancer-specific immunity by disrupting the gut microbiome.Citation33 To further substantiate the hypothesis of a causative rather than associative link,Citation34 broad-spectrum ATB covering both Gram-positive and negative species have been associated with significantly lower response rates (25% vs 61%, HR = 2.34; 95% CI: 1.5–3.65; p = .02) and shorter PFS (HR = 1.8; 95% CI: 0.86–3.89; p = .012), with no effect seen in patients treated with, narrow-spectrum ATB.Citation20

Interestingly, the duration of ATB appears equally important in influencing patients’ prognosis. Patients receiving multiple ATB courses or a single-prolonged course lasting >7 d had the worst overall PFS (median PFS, 2.8 months; HR, 2.625; p =.026) and OS (median OS, 6.3 months; HR, 1.904; p = .009) compared to ATB-unexposed controls (median PFS, 6.3 months; median OS, 21.7 months). Single, shorter courses of ATB had no significant effect on survival.Citation24

While the current body of evidence uniformly points toward a detrimental effect of ATB in influencing oncological outcomes from ICI, there is virtually no mechanistic evidence to delineate the immune-biologic foundations of such a profound shift in responsiveness and survival following ATB exposure. In absence of more solid evidence, clinicians should promote antimicrobial stewardship in ICI recipients and ensure that ATB prescribing is guided by best evidence, clinical judgment and precautionary principles to avoid potentially detrimental effect on response and survival from immunotherapy.Citation19 Prospective research addressing the causal roots of this association is key to promote the development of approaches that may favorably manipulate diversity and composition of gut bacteria to synergize with ICIs; for instance, pro-biotics, pre-biotics and fecal microbiota transplantation.Citation7

Corticosteroid therapy

ICIs are known to cause a wide range of immune-related adverse events (irAEs) affecting skin, endocrine, pulmonary and gastrointestinal systems through nonspecific dysregulation of self-tolerance leading to immune pathology.Citation35 Immunosuppressive treatment with corticosteroids is the first-line therapy for the majority of irAEs,Citation36 a point that has led to growing concern over the potentially detrimental effect of corticosteroids on outcomes from ICI. In addition to irAEs, indications for corticosteroid use including preexisting co-morbidities such as auto-immune conditions, as well as for alleviation of cancer-related symptoms such as anorexia/cachexia, pain or treatment of central nervous system metastases to reduce peritumoral edema.Citation27 These conditions themselves are often associated with a poorer prognosis, and thus can make it difficult to meaningfully evaluate the effects of steroid use in combination with ICIs due to potentially independent factors contributing to reduced prognosis in patients taking corticosteroids.

Interpreting this association is made even more difficult by the fact that patients assuming ≥10 mg of prednisolone equivalent daily are generally excluded from clinical trials,Citation37 and that the occurrence of irAE per se seems to predict for improved efficacy of ICIs, adding a further layer of complexity to the study of preexisting versus concomitant corticosteroid therapy.Citation37,Citation38 While it is accepted that corticosteroids act through a wide variety of mechanisms ranging from activation of glucocorticoid response elements (GREs) resulting in inhibition of IL-1 and IL-6 transcription,Citation39,Citation40 and impairment of the CD28 costimulatory pathway leading to diminished T-cell function,Citation41 it is not yet fully known the extent to which these mechanisms might affect cancer immune rejection in the context of ICI therapy.

Unsurprisingly, clinical studies in this area have yielded contradicting results ().

In one of the initial studies, Arbor et al.Citation25 looked at 640 patients with non-small cell lung cancer (NSCLC) and found that baseline use of steroids, defined as ≥10 mg prednisone equivalent, was associated with a reduced overall response rate (6% vs 19%; p = .02), median PFS (1.9 vs 2.6 months; HR = 1.7, p = .001) and OS (5.4 vs 12.1 months; HR = 2.1; p < .001) in patients treated with PD-(L)1 blocking agents.

However, a follow-on study by Ricciuti et al.Citation27 focusing on the indication of corticosteroid therapy found no significant difference in OS between patients receiving <10 mg vs. ≥10 mg prednisone equivalent for cancer-unrelated indications (10.7 v 11.2 months, respectively, p = .77). Confirmation of worse outcome in patients receiving corticosteroids for palliative indications suggests a predominant role of adverse disease features in dictating poorer survival outcomes, as opposed to immune-modulatory effects of corticosteroids.

As clinical evidence continues to evolve, particularly in oncological indications other than NSCLC, the provision of immunotherapy in patients receiving chronic corticosteroid therapy should be the focus of individualized decision-making, taking into account the primary indication for steroid therapy. While corticosteroids do not seem to influence outcome from ICI if prescribed in response to irAEs,Citation42 initiation of immunotherapy in immunosuppressed patients yields the potential for underlying immune pathologies to exacerbate, leading to potentially life-threatening complications.Citation43

Proton pump inhibitors

Proton pump inhibitors (PPI) are selective inhibitors of the H+/K+ ATPase widely used prophylactically and for the treatment of gastrointestinal conditions like peptic ulcers or gastroesophageal reflux disease (GORD).

Their potential to act at the cancer/immunity interface is multifaceted and so is the potential direction of the association with outcomes. It is thought they can cause immune-suppression by way of their ability to reduce the expression of adhesion molecules by inflammatory cells or alter the secretion of pro-inflammatory cytokines amongst other mechanisms.Citation44

Alongside direct immune-modulatory consequences thought to negatively influence outcomes to ICIs,Citation45 PPI therapy exerts important consequences on the gut microbiomeCitation46 due to its direct changes to gastric pH and delayed gastric emptying.Citation47 PPI use has been shown to significantly decrease the diversity of the gut microbiota and induce both positive and negative selection of specific bacterial species within the gut.Citation48

The effects of PPI, however, may not be exclusively detrimental: pre-clinical evidence has highlighted the anti-tumor potential of this drug class as a result of their capacity to neutralize the acidic tumor microenvironment, a concept that lacks full validation in patients.Citation49

Retrospective analyses on two randomized control trials of atezolizumab in NSCLC (POPLAR and OAK) assessed the impact of ATB and PPI had on survival outcomes for patients who were randomly assigned between ICI and chemotherapy. For the ICI patients, OS survival was significantly shorter in the PPI receivers (9.6 versus 14.5 months, HR 1.45, 95% CI 1.20–1.75, p = .0001) as was PFS (1.9 versus 2.8 months, HR 1.30, 95% CI 1.10–1.53, p = .001). Interestingly, PPI use was associated with a greater risk of progression or death in the immunotherapy but not in the chemotherapy control arms, corroborating a specific immune-modulatory action of PPI in the context of ICI.Citation18

Similar results were observed in patients with melanoma treated with PD-1-targeted therapies, where PPI use was associated with reduced PFS, OS and response rates. Analysis of pre-treatment samples showed PPI treated patients to have increased neutrophil and lymphocyte counts as well as higher NCAM1 and CSF3-R levels, suggesting a prognostically adverse pro-inflammatory status in these patients which is postulated to link PPI use with reduced efficacy of ICI therapy.Citation30

Much more limited evidence exists in other cancer types: a smaller retrospective study of 95 patients of mixed histologies (24% head and neck, 18% gastric) showed tumor-specific differences in response but not in PFS and OS.Citation45

Robust recommendations for PPI use cannot be inferred given the retrospective nature of currently available evidence. Heterogeneity in dosing or PPI type prescribed has often been disregarded.Citation29 Some studies were underpowered to look at differences in outcomes across different cancer types and many studies did not look at the effects of other influencing factors, for instance, co-prescription of corticosteroids or antibiotics. The association between PPI use and the risk of nephrotoxicity from ICI therapy is of interest and warrants further evaluation in prospective studies.Citation50

Metformin

Metformin is the drug of choice for patients with type 2 diabetes and is a complex regulator of multiple signaling pathways which result in increased insulin sensitivity and reduce liver gluconeogenesis.Citation31 Its role as an immune-modulatory agent capable of reprogramming the metabolism of the tumor microenvironment has been of great interest. Metformin inhibits complex 1 of the mitochondrial electron transport chain, an essential metabolic source for cancer proliferation.Citation51 Via activation of the LKB1/AMPK pathway, metformin inhibits mTOR signaling and down-regulates pro-inflammatory pathways, making the tumor microenvironment less tolerogenic via downregulation of PD-L1,Citation52 and over-expression of major histocompatibility complex I.Citation53 Metformin can also reduce immune exhaustion by increasing IL-10 production to enhance CD8 + T-cell function,Citation52 and concurrently inhibit CD39/CD73 on myeloid-derived suppressor cells.Citation32 Other favorable properties include shifts in the gut microbiota composition through alteration of microbial folate and serine/methionine metabolism,Citation52 and reduction of intra-tumoral hypoxia.Citation54

Despite the multi-faceted anti-tumor properties of metformin, clinical studies have been disappointing in confirming a beneficial effect. In a retrospective study of 50 NSCLC patients metformin use correlated with numerically longer median OS (11.5 vs 7.6 months, p = .5), PFS (4 vs 3 months, p = .6) values and higher response rates (41.1 vs 30.7%, p = .4) in patients administered both metformin and an anti-PD-1 inhibitor in combination vs those only receiving ICIs, although these did not meet statistical significance.Citation32 A larger study of 224 patients confirmed the lack of association.Citation55 The ongoing open-label phase Ib trial by Kubo et al.Citation56 is evaluating metformin concomitantly with nivolumab as a strategy to overcome immunotherapy-refractoriness.

Opioids

Opioid use can influence many physiological functions because of its varied receptor locations; its primary analgesic effect is achieved by targeting μ receptors within the central nervous system. Another target is on the receptors found in the enteric nervous system which is what coordinates action within the gut. This influence on gastrointestinal function has many effects, one such example that is widely observed being opioid-induced constipation and nausea.Citation57

Opioids may induce significant changes to the gut microbiota,Citation58 an observation supported by clinical and pre-clinical evidence.Citation59–61 Opioid use can also impair the gut barrier function and induce gut bacterial translocation, triggering downstream inflammatory responses.Citation62

Evidence around opioid use is limited, with only one study evaluating the effects of opioids on the clinical outcome of ICIs, albeit as a secondary aim. It was found that its use was associated with a significantly lower PFS and OS (median PFS 4.5 vs. 8.1 months, HR 1.79, 95% CI 1.12–2.85, p = .010; median OS 8.6 vs. 26.3 months, HR 3.08, 95% CI 1.71–5.55, p < .001) compared to those patients who did not use opioids.63

While interesting, the immune-mediated contribution to the adverse prognosis of ICI recipients who are concurrently on opioids must be weighed against the prognostically adverse value of a symptomatic oncological diagnosis, which might have triggered opiates use in the first place and confounded the association.

Conclusion

Concomitant medication use is common in cancer patients and the postulated immune-modulatory effects of certain drugs have led to an increased interest in DDIs in the context of ICI therapy. While biologic plausibility behind an immune-mediated influence on outcomes from ICI can be inferred for the majority of drug classes described here, the strength and robustness of the association vary across DDI and tumor type considered. Even in the case of antibiotics, where evidence of an adverse interaction is stronger, the debate between causal versus associative relationship has not been fully resolved. As the knowledge around DDI in the context of ICI therapy expands, clinicians should use this information to carefully review the appropriateness of prescribing on a case by case basis to ensure optimal efficacy of ICI is maintained without detriment to the treatment of cancer-related symptoms or co-morbidities.

Disclosure of potential conflict of interest

DJP received lecture fees from ViiV Healthcare, Bayer Healthcare and travel expenses from BMS and Bayer Healthcare; consulting fees for Mina Therapeutics, EISAI, Roche, Astra Zeneca; received research funding (to institution) from MSD, BMS. There are no other personal or financial conflicts of interest to disclose.

Authors’ contributions

Study concept and design: DJP

Acquisition of data: NH, MN

Analysis and interpretation of data: NH, MN, DJP

Drafting of the manuscript: NH, MN, DJP

Critical revision of the manuscript for important intellectual content: All the authors.

Statistical analysis: N/A

Obtained funding: DJP

Study supervision: DJP

Acknowledgments

The authors would like to acknowledge the infrastructure support provided by Imperial Experimental Cancer Medicine Centre, Cancer Research UK Imperial Centre and the Imperial College Healthcare NHS Trust Tissue Bank. DJP is supported by grant funding from the NIHR Imperial Biomedical Research Centre (BRC), ITMAT Push for Impact Grant Scheme 2019. The views expressed are those of the authors and not necessarily those of the NIHR or the Department of Health and Social Care.

Additional information

Funding

DJP is supported by grant funding from the Wellcome Trust Strategic Fund [PS3416].

References

  • Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23. doi:10.1056/NEJMoa1003466.
  • Kruger S, Ilmer M, Kobold S, Cadilha BL, Endres S, Ormanns S, Schuebbe G, Renz BW, D’Haese JG, Schloesser H, et al. Advances in cancer immunotherapy 2019 – latest trends. J Exp Clin Cancer Res. 2019;38(1):268. doi:10.1186/s13046-019-1266-0.
  • Schmidt C. The benefits of immunotherapy combinations. Nature. 2017;552(7685):S67–S69. doi:10.1038/d41586-017-08702-7.
  • Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10. doi:10.1016/j.immuni.2013.07.012.
  • Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847–56. doi:10.1158/1535-7163.MCT-14-0983.
  • High TMB predicts immunotherapy benefit. Cancer Discov. 2018;8(6):668. doi:10.1158/2159-8290.CD-NB2018-048.
  • Routy B, Gopalakrishnan V, Daillère R, Zitvogel L, Wargo JA, Kroemer G. The gut microbiota influences anticancer immunosurveillance and general health. Nat Rev Clin Oncol. 2018;15(6):382–96. doi:10.1038/s41571-018-0006-2.
  • Kluger HM, Tawbi HA, Ascierto ML, Bowden M, Callahan MK, Cha E, Chen HX, Drake CG, Feltquate DM, Ferris RL, et al. Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC immunotherapy resistance taskforce. J Immunother Cancer. 2020;8:1. doi:10.1136/jitc-2019-000398.
  • Beijnen JH, Schellens JHM. Drug interactions in oncology. Lancet Oncol. 2004;5(8):489–96. doi:10.1016/S1470-2045(04)01528-1.
  • Kamath AV. Translational pharmacokinetics and pharmacodynamics of monoclonal antibodies. Drug Discov Today Technol. 2016;21-22:75–83. doi:10.1016/j.ddtec.2016.09.004.
  • Zhou H, Mascelli MA. Mechanisms of monoclonal antibody-drug interactions. Annu Rev Pharmacol Toxicol. 2011;51(1):359–72. doi:10.1146/annurev-pharmtox-010510-100510.
  • Gong J, Chehrazi-Raffle A, Placencio-Hickok V, Guan M, Hendifar A, Salgia R. The gut microbiome and response to immune checkpoint inhibitors: preclinical and clinical strategies. Clin Transl Med. 2019:8. doi:10.1186/s40169-019-0225-x.
  • Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. doi:10.1126/science.aan4236.
  • Francino MP. Antibiotics and the human gut microbiome: dysbioses and accumulation of resistances. Front Microbiol. 2016:6. doi:10.3389/fmicb.2015.01543.
  • Pinato DJ, Gramenitskaya D, Altmann DM, Boyton RJ, Mullish BH, Marchesi JR, Bower M. Antibiotic therapy and outcome from immune-checkpoint inhibitors. J Immunother Cancer. 2019:7. doi:10.1186/s40425-019-0775-x.
  • Pinato DJ, Howlett S, Ottaviani D, Urus H, Patel A, Mineo T, Brock C, Power D, Hatcher O, Falconer A, et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol. 2019. doi:10.1001/jamaoncol.2019.2785.
  • Kim H, Lee JE, Hong SH, Lee MA, Kang JH, Kim I. The effect of antibiotics on the clinical outcomes of patients with solid cancers undergoing immune checkpoint inhibitor treatment: A retrospective study. BMC Cancer. 2019;19(1):1100. doi:10.1186/s12885-019-6267-z.
  • Chalabi M, Cardona A, Nagarkar DR, Dhawahir Scala A, Gandara DR, Rittmeyer A, Albert ML, Powles T, Kok M, Herrera FG. 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(4):525–31. doi:10.1016/j.annonc.2020.01.006.
  • Zhao S, Gao G, Li W, Li X, Zhao C, Jiang T, Jia Y, He Y, Li A, Su C, et al. Antibiotics are associated with attenuated efficacy of anti-PD-1/PD-L1 therapies in chinese patients with advanced non-small cell lung cancer. Lung Cancer. 2019;130:10–17. doi:10.1016/j.lungcan.2019.01.017.
  • Ahmed J, Kumar A, Parikh K, Anwar A, Knoll BM, Puccio C, Chun H, Fanucchi M, Lim SH. Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors. Oncoimmunology. 2018;7:11. doi:10.1080/2162402X.2018.1507670.
  • Derosa L, Hellmann MD, Spaziano M, Halpenny D, Fidelle M, Rizvi H, Long N, Plodkowski AJ, Arbour KC, Chaft JE, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Annals of Oncology. 2018;29(6):1437–44. doi:10.1093/annonc/mdy103.
  • Schett A, Rothschild SI, Curioni-Fontecedro A, Krähenbühl S, Früh M, Schmid S, Driessen C, Joerger M. Predictive impact of antibiotics in patients with advanced non small-cell lung cancer receiving immune checkpoint inhibitors. Cancer Chemother Pharmacol. 2020;85(1):121–31. doi:10.1007/s00280-019-03993-1.
  • Elkrief A, El Raichani L, Richard C, Messaoudene M, Belkaid W, Malo J, Belanger K, Miller W, Jamal R, Letarte N, et al. Antibiotics are associated with decreased progression-free survival of advanced melanoma patients treated with immune checkpoint inhibitors. OncoImmunology. 2019;8(4):e1568812. doi:10.1080/2162402X.2019.1568812.
  • Tinsley N, Zhou C, Tan G, Rack S, Lorigan P, Blackhall F, Krebs M, Carter L, Thistlethwaite F, Graham D, et al. Cumulative antibiotic use significantly decreases efficacy of checkpoint inhibitors in patients with advanced cancer. Oncologist. 2020;25(1):55–63. doi:10.1634/theoncologist.2019-0160.
  • Arbour KC, Mezquita L, Long N, Rizvi H, Auclin E, Ni A, Martínez-Bernal G, Ferrara R, Lai WV, Hendriks LEL, et al. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol. 2018;36(28):2872–78. doi:10.1200/JCO.2018.79.0006.
  • Scott SC, Pennell NA. Early use of systemic corticosteroids in patients with advanced NSCLC treated with nivolumab. Journal of Thoracic Oncology. 2018;13(11):1771–75. doi:10.1016/j.jtho.2018.06.004.
  • Ricciuti B, Dahlberg SE, Adeni A, Sholl LM, Nishino M, Awad MM. Immune checkpoint inhibitor outcomes for patients with non-small-cell lung cancer receiving baseline corticosteroids for palliative versus nonpalliative indications. J Clin Oncol. 2019;37(22):1927–34. doi:10.1200/JCO.19.00189.
  • Fucà G, Galli G, Poggi M, Lo Russo G, Proto C, Imbimbo M, Ferrara R, Zilembo N, Ganzinelli M, Sica A, et al. Modulation of peripheral blood immune cells by early use of steroids and its association with clinical outcomes in patients with metastatic non-small cell lung cancer treated with immune checkpoint inhibitors. ESMO Open. 2019;4(1):e000457. doi:10.1136/esmoopen-2018-000457.
  • Mukherjee S, Ibrahimi S, Khalid B, Roman D, Zhao D, Aljumaily R. Do proton pump inhibitors modulate the efficacy of anti-PD-1/PD-L1 therapy? A retrospective study. J Oncol Pharm Pract. 2019;25(3):762–64. doi:10.1177/1078155218771152.
  • Homicsko K, Richtig G, Tuchmann F, Tsourti Z, Hanahan D, Coukos G, Wind-Rotolo M, Richtig E, Zygoura P, Holler C, et al. Proton pump inhibitors negatively impact survival of PD-1 inhibitor based therapies in metastatic melanoma patients. Annals of Oncology. 2018;29:40–40. doi:10.1093/annonc/mdy511.001.
  • Afzal MZ, Mercado RR, Shirai K. Efficacy of metformin in combination with immune checkpoint inhibitors (anti-PD-1/anti-CTLA-4) in metastatic malignant melanoma. J Immunother Cancer. 2018;6(1):64. doi:10.1186/s40425-018-0375-1.
  • Afzal MZ, Dragnev K, Sarwar T, Shirai K. Clinical outcomes in non-small-cell lung cancer patients receiving concurrent metformin and immune checkpoint inhibitors. Lung Cancer Manag. 2019;8:2. doi:10.2217/lmt-2018-0016.
  • Abid MB. Could the menagerie of the gut microbiome really cure cancer? hope or hype. J Immunother Cancer. 2019;7(1):92. doi:10.1186/s40425-019-0561-9.
  • Lagier J, Million M, Hugon P, Armougom F, Raoult D. Human gut microbiota: repertoire and variations. Front Cell Infect Microbiol. 2012;2:136. doi:10.3389/fcimb.2012.00136.
  • Fessas P, Possamai LA, Clark J, Daniels E, Gudd C, Mullish BH, Alexander JL, Pinato DJ. Immunotoxicity from checkpoint inhibitor therapy: clinical features and underlying mechanisms. Immunology. 2020;159(2):167–77. doi:10.1111/imm.13141.
  • Myers G. Immune-related adverse events of immune checkpoint inhibitors: A brief review. Curr Oncol. 2018;25(5):342–47. doi:10.3747/co.25.4235.
  • Johnson DB, Sullivan RJ, Menzies AM. Immune checkpoint inhibitors in challenging populations. Cancer. 2017;123(11):1904–11. doi:10.1002/cncr.30642.
  • Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J ImmunoTher Cancer. 2019;7(1):306. doi:10.1186/s40425-019-0805-8.
  • Coutinho AE, Chapman KE. The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol Cell Endocrinol. 2011;335(1):2–13. doi:10.1016/j.mce.2010.04.005.
  • Luo Y, Zheng SG. Hall of fame among pro-inflammatory cytokines: interleukin-6 gene and its transcriptional regulation mechanisms. Front Immunol. 2016;7:604. doi:10.3389/fimmu.2016.00604.
  • Giles AJ, Hutchinson MND, Sonnemann HM, Jung J, Fecci PE, Ratnam NM, Zhang W, Song H, Bailey R, Davis D, et al. Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer. 2018;6(1):51. doi:10.1186/s40425-018-0371-5.
  • Medina P, Jeffers KD, Trinh VA, Harvey RD. The role of pharmacists in managing adverse events related to immune checkpoint inhibitor therapy. J Pharm Prac. 2019:0897190019885230. doi:10.1177/0897190019885230.
  • Abu-Sbeih H, Faleck DM, Ricciuti B, Mendelsohn RB, Naqash AR, Cohen JV, Sellers MC, Balaji A, Ben-Betzalel G, Hajir I, et al. Immune checkpoint inhibitor therapy in patients with preexisting inflammatory bowel disease. J Clin Oncol. 2020;38(6):576–83. doi:10.1200/JCO.19.01674.
  • Biswas S, Benedict SH, Lynch SG, LeVine SM. Potential immunological consequences of pharmacological suppression of gastric acid production in patients with multiple sclerosis. BMC Med. 2012;10:57. doi:10.1186/1741-7015-10-57.
  • Nguyen QP, Nomura M, Matsumoto S, Muto M. MO3-10-1 - the effect of proton pump inhibitors on the efficacy of nivolumab monotherapy in different types of cancer. Ann Oncol. 2019;30:vi115. doi:10.1093/annonc/mdz338.107.
  • Trabolsi A, Winter M, Rodriguez E. Proton pump inhibitors and response to immune check-point inhibitors: single center study. JCO. 2019;37(15):e14092. doi:10.1200/JCO.2019.37.15_suppl.e14092.
  • Bruno G, Zaccari P, Rocco G, Scalese G, Panetta C, Porowska B, Pontone S, Severi C. Proton pump inhibitors and dysbiosis: current knowledge and aspects to be clarified. World J Gastroenterol. 2019;25(22):2706–19. doi:10.3748/wjg.v25.i22.2706.
  • Jackson MA, Goodrich JK, Maxan M, Freedberg DE, Abrams JA, Poole AC, Sutter JL, Welter D, Ley RE, Bell JT, et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut. 2016;65(5):749–56. doi:10.1136/gutjnl-2015-310861.
  • Lugini L, Federici C, Borghi M, Azzarito T, Marino ML, Cesolini A, Spugnini EP, Fais S. Proton pump inhibitors while belonging to the same family of generic drugs show different anti-tumor effect. J Enzyme Inhibit Med Chem. 2016;31(4):538–45. doi:10.3109/14756366.2015.1046062.
  • Seethapathy H, Zhao S, Chute DF, Zubiri L, Oppong Y, Strohbehn I, Cortazar FB, Leaf DE, Mooradian MJ, Villani A, et al. The incidence, causes, and risk factors of acute kidney injury in patients receiving immune checkpoint inhibitors. Clin J Am Soc Nephrol. 2019;14(12):1692–700. doi:10.2215/CJN.00990119.
  • Wheaton WW, Weinberg SE, Hamanaka RB, Soberanes S, Sullivan LB, Anso E, Glasauer A, Dufour E, Mutlu GM, Budigner GS, et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. eLife. 2014;3. doi:10.7554/eLife.02242.
  • Verdura S, Cuyàs E, Martin-Castillo B, Menendez JA. Metformin as an archetype immuno-metabolic adjuvant for cancer immunotherapy. Oncoimmunology. 2019;8:10. doi:10.1080/2162402X.2019.1633235.
  • Oliveras-Ferraros C, Cufí S, Vazquez-Martin A, Menendez OJ, Bosch-Barrera J, Martin-Castillo B, Joven J, Menendez JA. Metformin rescues cell surface major histocompatibility complex class I (MHC-I) deficiency caused by oncogenic transformation. Cell Cycle. 2012;11(5):865–70. doi:10.4161/cc.11.5.19252.
  • Ne S, Av M, Rd WXZ. Gm D. Efficacy of PD-1 blockade is potentiated by metformin-induced reduction of tumor hypoxia. Cancer Immunol Res. 2016;5(1):9–16. doi:10.1158/2326-6066.CIR-16-0103.
  • Svaton M, Zemanova M, Zemanova P, Kultan J, Fischer O, Skrickova J, Jakubikova L, Cernovska M, Hrnciarik M, Jirousek M, et al. Impact of concomitant medication administered at the time of initiation of nivolumab therapy on outcome in non-small cell lung cancer. Anticancer Res. 2020;40(4):2209–17. doi:10.21873/anticanres.14182.
  • Kubo T, Ninomiya T, Hotta K, Kozuki T, Toyooka S, Okada H, Fujiwara T, Udono H, Kiura K. Study protocol: phase-ib trial of nivolumab combined with metformin for refractory/recurrent solid tumors. Clin Lung Cancer. 2018;19(6):e861–e864. doi:10.1016/j.cllc.2018.07.010.
  • Ketwaroo GA, Cheng V, Lembo A. Opioid-induced bowel dysfunction. Curr Gastroenterol Rep. 2013;15(9):344. doi:10.1007/s11894-013-0344-2.
  • Taylor A. Revealing a brain-gut microbiome connection following chronic opioid treatment. The Journal of Pain. 2018;19(3):S1. doi:10.1016/j.jpain.2017.12.012.
  • Acharya C, Betrapally NS, Gillevet PM, Sterling RK, Akbarali H, White MB, Ganapathy D, Fagan A, Sikaroodi M, Bajaj JS. Chronic opioid use is associated with altered gut microbiota and predicts readmissions in patients with cirrhosis. Aliment Pharmacol Ther. 2017;45(2):319–31. doi:10.1111/apt.13858.
  • Banerjee S, Sindberg G, Wang F, Meng J, Sharma U, Zhang L, Dauer P, Chen C, Dalluge J, Johnson T, et al. Opioid-induced gut microbial disruption and bile dysregulation leads to gut barrier compromise and sustained systemic inflammation. Mucosal Immunol. 2016;9(6):1418–28. doi:10.1038/mi.2016.9.
  • Ren M, Lotfipour S. The role of the gut microbiome in opioid use. Behav Pharmacol. 2020;31:2. doi:10.1097/FBP.0000000000000538.
  • Mora AL, Salazar M, Pablo-Caeiro J, Frost CP, Yadav Y, DuPont HL, Garey KW. Moderate to high use of opioid analgesics are associated with an increased risk of clostridium difficile infection. Am J Med Sci. 2012;343(4):277–80. doi:10.1097/MAJ.0b013e31822f42eb.
  • Iglesias-Santamaría A. Impact of antibiotic use and other concomitant medications on the efficacy of immune checkpoint inhibitors in patients with advanced cancer. Clin Transl Oncol. 2020. doi:10.1007/s12094-019-02282-w.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.