Skip to main content

Antibiotic therapy and outcome from immune-checkpoint inhibitors

Abstract

Sensitivity to immune checkpoint inhibitor (ICPI) therapy is governed by a complex interplay of tumor and host-related determinants. Epidemiological studies have highlighted that exposure to antibiotic therapy influences the probability of response to ICPI and predict for shorter patient survival across malignancies. Whilst a number of studies have reproducibly documented the detrimental effect of broad-spectrum antibiotics, the immune-biologic mechanisms underlying the association with outcome are poorly understood. Perturbation of the gut microbiota, an increasingly well-characterized factor capable of influencing ICPI-mediated immune reconstitution, has been indicated as a putative mechanism to explain the adverse effects attributed to antibiotic exposure in the context of ICPI therapy. Prospective studies are required to validate antibiotic-mediated gut perturbations as a mechanism of ICPI refractoriness and guide the development of strategies to overcome this barrier to an effective delivery of anti-cancer immunotherapy.

Introduction

Antibiotic therapy has produced unquestionable advances in the management of patients with cancer, a population with intrinsically higher risk of bacterial infection as a result of malignancy or treatment-related immune suppression.

While antimicrobial therapy has markedly reduced morbidity and mortality stemming from infection, the effects of broad-spectrum antibiotics on commensal, non-pathogenic bacterial species have remained for a long time an under-appreciated effect of this therapeutic class of drugs.

The gut microbiota, source of over 100 trillion bacteria, exists in a condition of mutually beneficial relationship with the host. Commensal bacteria are provided with a niche to colonise the host in return for their participation in the digestion of nutrients and xenobiotics, protection from pathogens and shaping of the host’s immune system subsets. Derangement of this delicate relationship has been increasingly well-characterised in the context of tumour-specific immune tolerogenesis [1].

Multiple levels of evidence now support the link between sensitivity to immunotherapy, taxonomic diversity and enrichment in specific gut bacterial taxa, suggesting that some species or species consortia provide intrinsic immune-modulating properties. The landmark study by Gopalakrishnan [2] demonstrated how broader stool bacterial diversity and higher representation of Ruminococcaceae communities including Faecalibacterium positively influences patients’ survival following ICPI by promoting a strongly immune-reactive microenvironment and lower systemic release of pro-inflammatory cytokines [3]. Many other commensal bacteria have subsequently been recognised to play a similar role including Bifidobacteria spp., a saccarolytic Gram-positive genus highly represented within the gut that facilitates dendritic cell maturation and increased accumulation of antigen-specific T-cells within the tumour microenvironment [4]. Similarly, the presence of the anaerobic commensal Akkermansia Muciniphila is more common in responders to ICPI, who display higher peripheral CD4 and CD8 memory T-cell responses to this bacterium [5].

Antibiotic (ATB) therapy imposes profound and protracted changes to the taxonomic diversity of the host microbial ecosystem, affecting the composition of up to 30% of the bacterial species in the gut microbiome [6], consequently leading to loss of microbial functions that are protective for the host. Such changes in gut microbial communities are rapid and pervasive, occurring within days from the first antibiotic dose [7] and persisting for up to several months after completion of therapy [8].

Mounting evidence from epidemiological studies has underscored the detrimental role of antibiotics in ICPI outcome, with exposure to antibiotics having been linked to shortened progression-free, overall survival and reduced response rates in patients receiving ICPI as part of clinical trials and in routine practice (Table 1). In a previous study, we demonstrated time-dependence of antibiotics exposure as a strong, tumour-agnostic determinant of outcome in ICPI recipients, confirming prior, but not concurrent antibiotic therapy as doubling the risk of primary progression to immunotherapy and leading to a > 20-months shortening in patients’ survival independent of established prognostic factors and corticosteroid use [10]. Whilst mirroring pre-clinical evidence, where antibiotic pre-conditioning ahead of tumour implantation leads to impaired responses to ICPI in mice [26, 27], the expanding body of clinical studies has so far painted an incomplete picture as to the mechanistic foundations underlying the relationship between ATB and immunotherapy, a point of greater consequence given the potential practice-influencing implications of ATB prescribing in the clinic.

Table 1 The relationship between antibiotic exposure and outcomes from immune checkpoint inhibitor therapy

Most of the studies highlighting the importance of a healthy gut microbial environment as a pre-requisite for ICPI response were unfortunately characterised by insufficient data on preceding or concomitant antibiotic exposure, making it impossible to disentangle the role of antibiotic-induced perturbation of the gut ecosystem in influencing clinically meaningful outcomes in these patients [3].

Mechanistically, the breadth and depth of downstream effects produced by antibiotics within the cancer-immune synapse are an important challenge in studying this prognostically adverse relationship. On one hand, the direct bacteriostatic/bactericidal effect of antibiotics can cause selective pressure within the host microbial ecosystem and instigate an alternative microbiota state characterised, amongst other traits, by downregulation of major histocompatibility complex (MHC) class I/II genes and impaired effector T-cell responses, immunologic traits implicated in reduced responsiveness to ICPI [28].

ATB-induced depletion of gut bacteria can also shift the repertoire of microbial-associated molecular patterns (MAMPS). These molecules signal through mucosal innate immune cells primarily via toll-like receptors (TLRs) and NOD1 [29] to influence neutrophil priming, reduce local cytokine release and prime adaptive immunity by influencing the expression of MHC genes within the intestinal mucosa and reduce immunoglobulin secretion [30]. Antibiotic treatment impairs TH1/TH17 responses in tumour-bearing mice through direct pre-conditioning of the gut microbiota, reducing the efficacy of cyclophosphamide-mediate immune-rejection of the tumour [31]. In addition, antibiotics can also reduce the capacity of adoptively transferred CD8+ T-cells to mediate a tumour-specific response through altered LPS/TLR4 signaling in lymphodepleted mice [32].

By disrupting the gut ecosystem, antibiotics instigate downstream metabolic alterations within the microenvironment with complex repercussions to the tumour-host-microbe interface. Amongst them, changes in the availability of short-chain fatty acids produced by Akkermansia, Faecalibacteria and Enterococcus from the catabolism of non-digestible carbohydrates and the conversion of primary bile acids to secondary bile acids (including deoxycholate) mediated by Clostridiales can significantly alter gut homeostasis and lead to profound and clinically meaningful immune-modulatory consequences [33]. The immune-metabolic repercussions secondary to gut dysbiosis, potentially reversible by oral Akkermansia supplementation [34], might explain the influence of body mass index in determining response to ICPI [35, 36].

With improved characterization of immune-microbiologic underpinning of the relationship between antibiotics and ICPI outcome, a key question now is whether disruption of a well-equilibrated gut bacterial ecosystem is truly causal in this relationship, and thus whether reversal of antibiotic-mediated gut dysbiosis might prove beneficial in restoring full sensitivity to ICPI. Whether a favourable gut microbiota is a reflection of an otherwise healthy host rather than the primum movens of clinically meaningful anti-cancer immune responses is still the subject of intense debate [13]. To this end, appreciating how antibiotics might dynamically affect such a strong immune-microbiologic correlate of response to checkpoint inhibition is of key importance to pave the way for strategies that could restore or protect the integrity of this important phenotypic correlate of response. To address the multiplicity of mechanisms that are likely to underscore this complex and bi-directional relationship, the coordinated study of a number of fundamental pathophysiologic processes including bacterial translocation, immune-modulation, an altered metabolome, enzymatic degradation and reduced diversity of the gut microbiome has been proposed as an overarching framework [37].

Gaining sufficient insight as to the mode of action by which bacteria might work as biotherapeutic agents is not just important for patient prognostication, but is in fact key to a successful, rational development of microbiome-modulating therapies which improve patient’s outcome with ICPI. With antibiotic use now having been validated as an important and dynamic factor influencing outcome from immunotherapy, concerted efforts should be aimed at characterizing the candidate taxonomic features in the gut microbiota that are associated with worse outcome from ICPI in the context of preceding and concomitant antibiotic exposure and evaluate them in conjunction with the concomitant prescription of proton pump inhibitors, corticosteroids and vaccines, all of which have been postulated to influence ICPI response [38].

Recognising these changes is expected to facilitate the clinical development of diverse biotherapeutic approaches to induce microbiome reprogramming including dietary interventions with pre-biotics, therapeutic administration of single or multiple types of bacterial species or their metabolites, selective antibiotic therapy or faecal microbial transplantation, all of which are currently at the focus of intense clinical research efforts [26].

Availability of data and materials

n/a.

Abbreviations

ATB:

Antibiotic

cATB:

Concurrent antibiotic treatment

CD:

Cluster of Differentiation

CTLA-4:

Cyotoxic T-Lymphocyte Associated Protein 4

DCR:

Disease control rate

EIOP:

Early Immunotherapy Period

GI:

Gastrointestinal

HR:

Hazard Ratio

ICPI:

Immune-checkpoint inhibitors

LPS:

Lipopolysaccharide

MHC:

Major Histocompatibility Complex

NOD1:

Nucleotide-binding oligomerization domain-containing protein 1

NSCLC:

Non-small Cell Lung Cancer

ORR:

Overall response rate

OS:

Overall Survival

pATB:

Prior antibiotic treatment

PD:

Progressive Disease

PD-1:

Programmed Cell-Death 1

PD-L1:

Programmed Cell-Death Ligand 1

PFS:

Progression-free survival

RCC:

Renal Cell Carcinoma

RR:

Response rate

TH:

T-Helper cell

TLR:

Toll-like receptors

WIOP:

Whole immunotherapy Period

References

  1. Zitvogel L, Galluzzi L, Viaud S, Vetizou M, Daillere R, Merad M, et al. Cancer and the gut microbiota: an unexpected link. Sci Transl Med. 2015;7(271):271ps1.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.

    Article  CAS  PubMed  Google Scholar 

  3. Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368–79.

    Article  CAS  PubMed  Google Scholar 

  4. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.

    Article  CAS  PubMed  Google Scholar 

  6. Francino MP. Antibiotics and the human gut microbiome: Dysbioses and accumulation of resistances. Front Microbiol. 2015;6:1543.

    CAS  PubMed  Google Scholar 

  7. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4554–61.

    Article  CAS  PubMed  Google Scholar 

  8. Yoon MY, Yoon SS. Disruption of the gut ecosystem by antibiotics. Yonsei Med J. 2018;59(1):4–12.

    Article  CAS  PubMed  Google Scholar 

  9. Derosa L, Hellmann M, Spaziano M, Halpenny D, Fidelle M, Rizvi H 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. Ann Oncol. 2018;29(6):1437-1444.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pinato DJ, Howlett S, Ottaviani D, Urus H, Patel A, Mineo T, et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol. 2019. https://doi.org/10.1001/jamaoncol.2019.2785.

  11. Hakozaki T, Okuma Y, Omori M, Hosomi Y. Impact of prior antibiotic use on the efficacy of nivolumab for non‑small cell lung cancer. Oncol Lett. 2019;17(3):2946-2952. https://doi.org/10.3892/ol.2019.9899. Epub 2019 Jan 8.

  12. Galli G, Triulzi T, Proto C, Signorelli D, Imbimbo M, Poggi M et al. Association between antibiotic-immunotherapy exposure ratio and outcome in metastatic non small cell lung cancer. Lung Cancer. 2019;132:72-78. https://doi.org/10.1016/j.lungcan.2019.04.008.

    Article  PubMed  Google Scholar 

  13. Ahmed J, Kumar A, Parikh K, Anwar A, Knoll B, Puccio C et al. Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors. OncoImmunology. 2018;7(11):e1507670. https://doi.org/10.1080/2162402X.2018.1507670

    Article  PubMed  PubMed Central  Google Scholar 

  14. Tinsley N, Zhou C, Villa S, Tan G, Lorigan P, Blackhall F et al. Cumulative antibiotic use and efficacy of immune checkpoint inhibitors in patients with advanced cancer. J Clin Oncol. 2018;36(15_suppl):3010-3010.

    Article  Google Scholar 

  15. Khan U, Peña C, Brouwer J, Hoffman K, Choudhury A, Zhang C et al. Impact of antibiotic use on response to treatment with immune checkpoint inhibitors. J Clin Oncol. 2019;37(4_suppl):143-143.

    Article  Google Scholar 

  16. Mielgo-Rubio X, Chara L, Sotelo-Lezama M, Lopez Castro R, Rubio-Martínez J, Velastegui A et al. MA10.01 Antibiotic Use and PD-1 Inhibitors: Shorter Survival in Lung Cancer, Especially When Given Intravenously. Type of Infection Also Matters. J Thorac Oncol. 2018;13(10):S389.

    Article  Google Scholar 

  17. Ouaknine Krief J, Helly de Tauriers P, Dumenil C, Neveux N, Dumoulin J, Giraud V et al. Role of antibiotic use, plasma citrulline and blood microbiome in advanced non-small cell lung cancer patients treated with nivolumab. J Immunother Cancer. 2019;7(1):176. https://doi.org/10.1186/s40425-019-0658-1.

  18. Kaderbhai C, Richard C, Fumet JD, Aarnink A, Foucher P, Coudert B, Favier L, Lagrange A, L E, Boidot R, Ghiringhelli F. Antibiotic Use Does Not Appear to Influence Response to Nivolumab. Anticancer Res. 2017;37(6):3195-3200.

  19. Zhao S, Gao G, Li W, Li X, Zhao C, Jiang T 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. https://doi.org/10.1016/j.lungcan.2019.01.017. Epub 2019 Jan 31.

    Article  PubMed  Google Scholar 

  20. Thompson J, Szabo A, Arce-Lara C, Menon S. P1.07-008 Microbiome & Immunotherapy: Antibiotic Use Is Associated with Inferior Survival for Lung Cancer Patients Receiving PD-1 Inhibitors. J Thorac Oncol. 2017;12(11):S1998.

  21. Derosa L, Routy B, Enot D, Baciarello G, Massard C, Loriot Y et al. Impact of antibiotics on outcome in patients with metastatic renal cell carcinoma treated with immune checkpoint inhibitors. J Thorac Oncol. 2017;35(6_suppl):462-462.

    Article  Google Scholar 

  22. Do T, Hegde A, Cherry C, Stroud C, Sharma N, Cherukuri S et al. Antibiotic use and overall survival in lung cancer patients receiving nivolumab. J Clin Oncol. 2018;36(15_suppl):e15109-e15109.

    Article  Google Scholar 

  23. Elkrief A, El Raichani L, Richard C, Messaoudene M, Belkaid W, Malo J et al. Antibiotics are associated with decreased progression-free survival of advanced melanoma patients treated with immune checkpoint inhibitors. OncoImmunology. 2019 Feb 18;8(4):e1568812. https://doi.org/10.1080/2162402X.2019.1568812.

    Article  PubMed  Google Scholar 

  24. Huemer F, Rinnerthaler G, Westphal T, Hackl H, Hutarew G, Gampenrieder S et al. Impact of antibiotic treatment on immune-checkpoint blockade efficacy in advanced non-squamous non-small cell lung cancer. Oncotarget. 2018;9(23):16512-16520. https://doi.org/10.18632/oncotarget.24751.

  25. Lalani A, Xie W, Lin X, Steinharter J, Martini D, Duquette A et al. Antibiotic use and outcomes with systemic therapy in metastatic renal cell carcinoma (mRCC). J Clin Oncol. 2018;36(6_suppl):607-607.

    Article  Google Scholar 

  26. Routy B, Gopalakrishnan V, Daillere 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.

    Article  CAS  PubMed  Google Scholar 

  27. Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cianci R, Franza L, Schinzari G, Rossi E, Ianiro G, Tortora G, et al. The interplay between immunity and microbiota at intestinal immunological niche: The case of cancer. Int J Mol Sci. 2019;20(3). https://doi.org/10.3390/ijms20030501.

    Article  CAS  PubMed Central  Google Scholar 

  29. Ubeda C, Pamer EG. Antibiotics, microbiota, and immune defense. Trends Immunol. 2012;33(9):459–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Dufour V, Millon L, Faucher JF, Bard E, Robinet E, Piarroux R, et al. Effects of a short-course of amoxicillin/clavulanic acid on systemic and mucosal immunity in healthy adult humans. Int Immunopharmacol. 2005;5(5):917–28.

    Article  CAS  PubMed  Google Scholar 

  31. Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Paulos CM, Wrzesinski C, Kaiser A, Hinrichs CS, Chieppa M, Cassard L, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest. 2007;117(8):2197–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Maslowski KM. Metabolism at the Centre of the host-microbe relationship. Clin Exp Immunol. 2019;197(2):193–204. https://doi.org/10.1111/cei.13329.

    Article  CAS  Google Scholar 

  34. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med. 2019;25(7):1096–103.

    Article  CAS  PubMed  Google Scholar 

  35. Cortellini A, Bersanelli M, Buti S, Cannita K, Santini D, Perrone F, et al. A multicenter study of body mass index in cancer patients treated with anti-PD-1/PD-L1 immune checkpoint inhibitors: when overweight becomes favorable. J Immunother Cancer. 2019;7(1):57.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Xu H, Cao D, He A, Ge W. The prognostic role of obesity is independent of sex in cancer patients treated with immune checkpoint inhibitors: a pooled analysis of 4090 cancer patients. Int Immunopharmacol. 2019;74:105745.

    Article  CAS  PubMed  Google Scholar 

  37. Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356–65.

    Article  CAS  PubMed  Google Scholar 

  38. Rossi G, Pezzuto A, Sini C, Tuzi A, Citarella F, McCusker MG, et al. Concomitant medications during immune checkpoint blockage in cancer patients: novel insights in this emerging clinical scenario. Crit Rev Oncol Hematol. 2019;142:26–34.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge the Imperial College National Institute for Health Research Biomedical Research Centre and Imperial Cancer Research UK centre for infrastructural and grant funding support. DJP is supported by grant funding from the Wellcome Trust Strategic Fund (PS3416). BHM is the recipient of a Medical Research Council (MRC) Clinical Research Training Fellowship (grant MR/R000875/1).

Funding

DJP, RJB, BHM, JRM, DMA have received direct project funding by 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.

Author information

Authors and Affiliations

Authors

Contributions

DJP, DG, DMA, RJB, BHM, JRM and MB contributed to the writing and editing of this manuscript. All authors read and approved the final version of the manuscript.

Corresponding author

Correspondence to David J. Pinato.

Ethics declarations

Ethics approval and consent to participate

n/a.

Consent for publication

n/a.

Competing interests

Dr. Pinato reports receiving grant funding in support of clinical trials from Merck Sharpe and Dohme and Bristol Myers Squibb and having received speaker/consultancy fees from ViiV Healthcare, Bayer and MiNa therapeutics outside the submitted work. There are no other conflicts of interest to report.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pinato, D.J., Gramenitskaya, D., Altmann, D.M. et al. Antibiotic therapy and outcome from immune-checkpoint inhibitors. j. immunotherapy cancer 7, 287 (2019). https://doi.org/10.1186/s40425-019-0775-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40425-019-0775-x

Keywords