Skip to main content
Infection and Immunity logoLink to Infection and Immunity
. 2021 Aug 16;89(9):e00665-20. doi: 10.1128/IAI.00665-20

How Could Antibiotics, Probiotics, and Corticoids Modify Microbiota and Its Influence in Cancer Immune Checkpoint Inhibitors: A Review

Mara Cruellas a,b,*,, Alfonso Yubero a,b, María Zapata a,b, Eva M Galvez c, Marta Gascón a,b, Dolores Isla a,b, Rodrigo Lastra a,b, Luis Martínez-Lostao b,d,e,f,g, Maitane Ocariz a,b, Julián Pardo b,h,i,j, Ariel Ramírez k, Andrea Sesma a,b, Irene Torres-Ramón a,b, José Ramón Paño b,l
Editor: Anthony R Richardsonm
PMCID: PMC8370676  PMID: 33526567

ABSTRACT

Immunotherapy has become a new paradigm in oncology, improving outcomes for several types of cancer. However, there are some aspects about its management that remain uncertain. One of the key points that needs better understanding is the interaction between immunotherapy and gut microbiome and how modulation of the microbiome might modify the efficacy of immunotherapy. Consequently, the negative impact of systemic antibiotics and corticosteroids on the efficacy of immunotherapy needs to be clarified.

KEYWORDS: cancer immunotherapy, immune checkpoint inhibitors, antibiotics, immunotherapy, corticosteroids, gut microbiota

INTRODUCTION

Immune checkpoints (ICP) are modulators of the immune response and have an essential role in self-tolerance. ICP could inhibit T-cell function through several mechanisms, such as inhibition of dendritic cell-mediated T-cell activation (CTLA-4) or inducing T-cell exhaustion at sites where there is ongoing inflammation (PD-1/PD-L1) (1). Some neoplasms use ICP in favor of reducing the antineoplastic effect of the host immune system.

Immune checkpoint inhibitors (ICI) act against specific molecules of the immune system, which have a suppressor function. Furthermore, ICI increase the antineoplastic effect of the host immune system. Treatments with ICI have been shown to improve cancer prognosis in patients with several neoplasms, having become the standard of care in some instances (24). Nevertheless, the benefit of ICP is restricted to a limited number of patients, and only 25 to 30% of individuals who receive ICI achieve clinical benefit. Several tumor cells or oncogenic pathway-related factors are associated with favorable outcomes with ICI (26). Other host-related or extrinsic factors, such as those that modify the microbiome, might influence overall efficacy of ICI (7).

To establish the influence of the microbiome on the response to ICI, we will briefly review the relationship between the microbiome and ICI efficacy and how altering the microbiome might modulate the efficacy of immunotherapy in cancer patients.

MICROBIOME AND CANCER

The microbiome encompasses all host-associated microorganisms present on epithelial barriers, constituting a dynamic ecosystem (8, 9). It is formed by approximately 1014 microorganisms, such as bacteria, fungi, viruses, and archaea. It is located in different body surfaces and compartments, with the gastrointestinal (GI) tract being one of its main locations. Its composition is influenced by several factors, such as diet, drug therapies, age, and genetics (8, 9). The immune system at the GI tract works to achieve a mutually beneficial equilibrium between these microbes and the host, involving both the innate and adaptive immune systems. The mechanisms aiming to protect the host from bacterial damage or invasion are counterbalanced by processes that restrain the antibacterial response, preventing collateral damage from inflammation (10). Several studies on animal models found that immunological responses vary depending on the bacterial species present in the gut microbiota (10). In this way, the microbiome contributes to shape and modulate the immune system, exerting an overall profound impact on the peripheral immune system. The microbiome might have a direct role in local and distant carcinogenesis, providing toxic or oncogenic products, or an indirect role by promoting inflammation and/or immunosuppression. Conversely, some bacteria have been found to have distant antitumor activity by potentiating host immune responses (8).

THE MICROBIOME AND IMMUNOTHERAPY IN CANCER PATIENTS

The microbiome apparently influences the efficacy of several antineoplastic therapies, including immunomodulatory agents such as ICI. Recently, a review by Gong et al. reported that several interdependence mechanisms and pathways have been proposed to explain the effect of the microbiome on the efficacy of ICI (11).

Some experiments have found that in mice with altered gut microbiomes, whether by administrating antibiotics or in germfree mice, ICI treatment outcomes are poorer (12, 13). Routy et al. provided an example of the impact of the human microbiome on the effect of ICI. In their experiment, fecal microbiota transplantation (FMT) from cancer patients who responded to ICI, into germfree or antibiotic-treated mice, was found to improve the antitumor effects of PD-1 blockade, as opposed to FMT from nonresponding patients (14).

A relationship between Bacteroides fragilis and the efficacy of anti-CTLA-4 has also been postulated, as B. fragilis usually is overexpressed in ileum, stimulating Th1 cells, after CTLA-4 blockade (15). Similarly, other Bacteroides spp. might induce a Th1 response that contributes to an optimal effect therapeutic from anti-CTLA-4 (16, 17). In a recent review, Ma et al. identified that other species, such as Bifidobacterium longum, Bifidobacterium breve species, and Faecalibacterium spp., have been associated with increased anti-PD1 efficacy (15, 18). Bifidobacterium seems to increase gamma interferon (IFN-γ) and CD8+ T cells in tumors, stimulate the maturation of dendritic cells, and increase the amount of tumor-specific cytotoxic T lymphocytes (15, 16). Moreover, several other bacterial species and genera, such as Akkermansia muciniphila, Clostridiales, and Ruminococcaceae, if present in the microbiome, prevent leaky colon and systemic immunosuppression (14) and are related to better outcomes (15, 18).

IMMUNE-RELATED ADVERSE EVENTS, RESPONSE, AND THE MICROBIOME IN PATIENTS RECEIVING ICI

ICI-induced T-cell activation is not tumor specific. ICI-induced immune response may damage host tissues, causing immune-related adverse events (IrAE). These can present as colitis, endocrine dysfunction, skin disorders, pneumonitis, or myocarditis (among others). Most IrAEs are mild to moderate and tend to appear in the first 12 weeks after initiating treatment (although they may occur even 6 months after ICI discontinuation) (1). Some factors, such as therapeutic dosage, the use of combination checkpoint blockade, or microbiome-related factors, suggest an association with an increased risk of IrAE (19).

A correlation between IrAE and ICI efficacy has been postulated. In a secondary analysis of the CA209-003 study, a multicenter, phase 1 trial that included 270 patients with non-small-cell lung cancer (NSCLC), melanoma, or renal cancer, this association was assessed (20). Patients with treatment-related adverse effects had a significantly increased long-term overall survival (OS) compared with patients who did not experience these kind of adverse effects (Δ, 19.8 months; 95% confidence intervals [CI], 13.8 to 26.9 months). In some cases with early discontinuation due to grade 3 adverse events, tumor regression was persistent.

The relationship between IrAE and the microbiome has also been previously explored. Several changes in colonic mucosa after administration of anti-CTLA-4 have been observed (17). Bacteroides spp. are associated with favorable tumor responses to anti-CTLA-4 without significant intercurrent intestinal toxicity (17). Likewise, Dubin et al. proposed the Bacteroidetes phylum as a biomarker correlated with protection against colitis secondary to anti-CTLA-4. The Bacteroidetes phylum was found in colitis-resistant patients and could stimulate T-regulatory cell differentiation and limit inflammation (21). Similar results were proposed by Chaput et al., relating Bacteroidetes with colitis-free patients and low anticancer response (22). Conversely, microbiota with high levels of Firmicutes seems to predispose to a better response but with more risk of colitis (22).

MECHANISMS OF INFLUENCE ON MICROBIOME AND IMMUNOTHERAPY ANTIBIOTICS

Antibiotics.

Antibiotics modify the gastrointestinal microbiome by diminishing bacterial diversity (23). It may take longer than 6 weeks to restore the microbiome depending on the duration, use, and type of antibiotic (4, 22, 23). Antibiotics, as microbiome-modifying agents, are expected to influence the overall efficacy of this type of immunotherapy (7).

Several observational studies and recent reviews suggest that antibiotic use decreases the efficacy of ICI (4, 7, 14, 2327). For instance, Routy et al. addressed the impact of antibiotics within 2 months before, or 1 month after, the initiation of PD1-PD-L1 monoclonal antibodies. This was based on a cohort of 249 patients with non-small-cell lung cancer (NSCLC) (140 patients), renal cancer (67 patients), or urothelial cancer (42 patients) (14). They found that progression-free survival (PFS) (4.1 versus 3.5 months; P = 0.017) and OS (11.5 versus 20.8 months; P < 0.001) were significantly poorer in patients who had received antibiotics (14). The impact of antibiotic use and ICI efficacy was also explored in a retrospective cohort study including 360 patients with NSCLC and renal cancer from two U.S. centers (23). They reported a significant and independent PFS (1.9 versus 7.4 months; hazard ratio [HR], 3.1; 95% confidence interval [CI], 1.4 to 6.9; P < 0.01) and OS (median 17.3 versus 30.6 months; HR, 3.5; 95% CI, 1.1 to 10.8; P = 0.03) reduction in patients who had received systemic antibiotics. The analysis of antibiotic timing on ICI efficacy concluded that the reduction in ICI efficacy was most marked in those patients who had received antibiotics within 30 days before ICI compared with those receiving antibiotics within 60 days before ICI. The process of microbiome recovery in the latter could explain these differences.

In a recent retrospective observational study, which included 291 patients with advanced NSCLC, melanoma, or renal cancer, it was found that patients who received at least 1 course of systemic antibiotics 2 weeks before or during ICI therapy had lower PFS (3.1 versus 6.3 months; P = 0.003) and OS (10.4 versus 21.7 months; P = 0.002) (4). In the multivariate analysis including other prognostic factors, the authors concluded that the antibiotic received was an independent predictor of shorter PFS and OS. Interestingly, the authors categorized antibiotic use into two groups, a single antibiotic group (up to 7 days of a single antibiotic) or a cumulative antibiotic group (more than one antibiotic or a single antibiotic for more than 7 days), with poorest outcomes in the latter (4).

Recently, Hakozaki et al. observed shortened median PFS (1.2 versus 4.1 months) and OS (7.9 months versus not reached) among 13/90 patients with NSCLC treated with antibiotics prior to nivolumab but without statistically significant differences after multivariate analysis (7). In a retrospective single-center study by Galli et al., overall antibiotic use was not associated with PFS or OS. Nevertheless, when antibiotic exposure was considered by the means of the antibiotic-immunotherapy exposure ratio (AIER), defined as days of antibiotic/days of immunotherapy, high antibiotic exposure was independently associated with decreased PFS and OS (3). A pooled analysis, including 2,740 cancer patients from 19 eligible studies, found a significant decrease in PFS (HR, 1.84; 95% CI, 1.49 to 2.26; P < 0.001) and OS (HR, 2.37; 95% CI, 2.05 to 2.75; P < 0.001) (28). Recently, a pooled analysis including 1,512 patients from a phase II POPLAR and a phase III OAK group (both evaluated atezolizumab versus docetaxel in NSCLC) has been published (29). In the analysis, the effect of antibiotics taken between 30 days before and 30 days after the start of the study was evaluated. They observed unfavorable prognosis (shorter OS but unchanged PFS) in patients receiving antibiotics concomitantly with atezolizumab. Exploratory analysis suggests OS differences were detected across all classes of antibiotics used (29).

Very recently, a systematic review and meta-analysis conducted by Lurienne et al. was published (30). They concluded that the use of antibiotics before or during ICI administration reduces OS outcomes, especially their use shortly before or after starting ICI (30). Table 1 summarizes the key findings of studies assessing the impact of concomitant antibiotic therapy in cancer prognosis in patients treated with ICI.

TABLE 1.

Summary about studies assessing the impact of concomitant antibiotic therapy in cancer prognosis in patients treated with ICIsa

Reference No. of patients Type of cancer PFS (ATB vs no ATB) OS (ATB vs no ATB)
14 249 NSCLC, renal, and urothelial 4.1 vs 3.5 mo (P = 0.017) 20.8 vs 11.5 mo (P < 0.001)
24 360 NSCLC and renal 7.4 vs 1.9 mo (P < 0.01) 30.6 vs 17.3 mo (P  = 0.03)
4 291 NSCLC, melanoma, and renal 6.3 vs 3.1 mo (P  = 0.003) 21.7 vs 10.4 mo (P  = 0.002)
7 90 NSCLC 4.1 vs 1.2 mo (P = 0.049) NR vs 7.9 mo (P  = 0.037)
3 157 NSCLC P < 0.0001 if high AIER P = 0.0004 if high AIER
Pooled analysis
    28 2,740 Several P < 0.001 P < 0.001
    29 169 NSCLC No significant differences in PFS 14.1 vs 8.5 mo (P = 0.01)
a

NSCLC, non-small-cell lung cancer; PFS, progression-free survival; ATB, antibiotic; OS, overall survival; AIER, antibiotic-immunotherapy exposure ratio.

Corticosteriods.

Corticosteroids are used to minimize inflammation and decrease immune activity with effect on T-cell function (31). They also affect the microbiome (30). For example, in a paper published in 2015, the authors explained that mouse gut microbiota revealed phylogenetic shifts after dexamethasone treatment. They observed an increase in the Actinobacteria, Bifidobacterium, and Lactobacillus levels compared with controls (32). More research is needed to clearly establish the modification of the microbiota after corticosteroids.

Corticosteroids are used for mild to severe IrAE (19). Moreover, some patients could need corticosteroids during ICI treatment as part of best supportive care (2, 31, 33). However, there is some doubt whether corticosteroids could influence ICI response.

The baseline or early use of corticosteroids at the time of initiating ICI has an influence on ORR, PFS, and OS results (2, 3338). Curiously, in one study there were no significant differences in overall survival between patients who received steroids to treat IrAE compared with patients who did not develop IrAE (36). It has been postulated that baseline corticosteroids could blunt a proliferative burst of CD8-positive T cells that are otherwise needed for ICI effective response (33). Nevertheless, it should be noted that patients with early use of steroids tend to have worse ECOG, increased numbers of metastases, or metastases in the brain (2), all of which are independent predictors of unfavorable outcomes.

Recently, the concomitant use of ICI with chemotherapy in first-line NSCLC has been approved. It seems that the use of corticosteroids as premedication does not have a detrimental effect. This suggests that corticosteroid dose or duration thresholds are associated with decreased ICI response (31). Another explanation is that the benefit of combination has a synergic effect that offsets the detrimental corticosteroid action (33).

It has been speculated that glucocorticoid-induced tumor necrosis factor receptor-related-protein (GITR) could improve immune response, as it is an anti-glucocorticoid agent (39). In a phase 1/2a clinical trial using human GITR agonist IgG1 monoclonal antibody BMS-986156, which included 292 patients with advanced solid tumors, the researchers tried implementing this protein with dose-escalation in monotherapy and in combination with nivolumab. The study concluded that its efficacy with nivolumab is comparable to historical data and had a manageable safety profile (40).

Probiotics.

The gut microbiome can be altered by probiotics and is defined as a single or combination of bacterial species that, when administered in adequate amounts, confer a health benefit to the host (4, 41). Indeed, several clinical trials exploring the impact of probiotic therapy for cancer patients are currently ongoing (4). Probiotics could enhance the response to immunotherapy by modulating the individual species of the microbiome (18). Additionally, they can modulate production of anti-inflammatory cytokines, which are implicated in carcinogenesis and can activate phagocytes to destroy early-stage cancer cells (42).

Most probiotics used for therapy are lactic acid-producing bacteria, such as Lactobacillus and Bifidobacterium. Other possible probiotics are Streptococcus, Bacillus, Enterococcus, and Saccharomyces spp., with some safety limitations (41). The modulation capacity of Lactobacillus spp. and Bifidobacterium spp. in immunotherapy cancer responses has been observed in several studies (43). Sivan et al. assessed melanoma growth in mice that received a combination of Bifidobacterium with anti-PD-L1 therapy. They observed that this combination abolished tumor outgrowth (43). Lactobacillus spp. and Bifidobacterium spp. have been deeply studied in experimental models and appear to be safe (44, 45). These species produce lactic and acetic acids, decreasing luminal pH, which hampers the overgrowth of other undesired species (45). Furthermore, they increase the expression of tight junction proteins, improving barrier function in the gut (45).

CONCLUSIONS

It has been widely demonstrated that concomitant use of immunotherapy with antibiotics or corticosteroids implies changes in the expected response to ICI. Early use of antibiotics just prior to or immediately after the initiation of ICI has a negative impact on survival. A similar result occurs with the baseline or early use of corticosteroids at the beginning of ICI treatment. Therefore, it is important to make careful use of them and consider the influence that they may have on ICI response. Regarding probiotics, this is an area of potential development that can help modulate the effect of immunotherapy. Their relationship with the gut microbiome has been extensively demonstrated. This link is likely one of the modulating mechanisms of the response to immunotherapy.

ACKNOWLEDGMENTS

J.P. reports research funding from BMS and Gilead and speaker honoraria from Gilead and Pfizer. E.M.G. reports research funding from BMS and Gilead. J.P. and E.M.G. are funded by FEDER (Fondo Europeo de Desarrollo Regional), Gobierno de Aragón (Group B29_17R), Ministerio de Ciencia, Innovación e Universidades (MCNU), Agencia Estatal de Investigación (SAF2017‐83120‐C2‐1‐R), Fundación Inocente, ASPANOA, and Carrera de la Mujer de Monzón. J.P. is supported by ARAID Foundation. D.I. reports consultation honoraria from AbbVie, Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Eli Lilly Oncology, F. Hoffmann-La Roche, Merck, MSD, Novartis, Pierre Fabre, Pfizer, and Takeda. D.I. reports speaker honoraria from Amgen, AstraZeneca, BMS, Boehringer Ingelheim, Eli Lilly Oncology, F. Hoffmann-La Roche, MSD, Novartis, Pierre Fabre, and Pfizer. D.I. reports a research grant from AstraZeneca, BMS, F. Hoffmann-La Roche, MSD, and Pierre Fabre.

Contributor Information

Mara Cruellas, Email: mara1290@gmail.com.

Anthony R. Richardson, University of Pittsburgh

REFERENCES

  • 1.Johnson DB, Chandra S, Sosman JA. 2018. Immune checkpoint inhibitor toxicity in 2018. JAMA 320:1702–1703. doi: 10.1001/jama.2018.13995. [DOI] [PubMed] [Google Scholar]
  • 2.Fucà G, Galli G, Poggi M, Lo Russo G, Proto C, Imbimbo M, Ferrara R, Zilembo N, Ganzinelli M, Sica A, Torri V, Colombo MP, Vernieri C, Balsari A, de Braud F, Garassino MC, Signorelli D. 2019. 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 4:e000457–e000458. doi: 10.1136/esmoopen-2018-000457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Galli G, Triulzi T, Proto C, Signorelli D, Imbimbo M, Poggi M, Fucà G, Ganzinelli M, Vitali M, Palmieri D, Tessari A, de Braud F, Garassino MC, Colombo MP, Lo Russo G. 2019. Association between antibiotic-immunotherapy exposure ratio and outcome in metastatic non small cell lung cancer. Lung Cancer 132:72–78. doi: 10.1016/j.lungcan.2019.04.008. [DOI] [PubMed] [Google Scholar]
  • 4.Tinsley N, Zhou C, Tan G, Rack S, Lorigan P, Blackhall F, Krebs M, Carter L, Thistlethwaite F, Graham D, Cook N. 2020. Cumulative antibiotic use significantly decreases efficacy of checkpoint inhibitors in patients with advanced cancer. Oncologist 25:55–63. doi: 10.1634/theoncologist.2019-0160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, Felip E, van den Heuvel MM, Ciuleanu TE, Badin F, Ready N, Hiltermann TJN, Nair S, Juergens R, Peters S, Minenza E, Wrangle JM, Rodriguez-Abreu D, Borghaei H, BlumenscheinGR, Jr, Villaruz LC, Havel L, Krejci J, Corral Jaime J, Chang H, Geese WJ, Bhagavatheeswaran P, Chen AC, Socinski MA, CheckMate 026 Investigators. 2017. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med 376:2415–2426. doi: 10.1056/NEJMoa1613493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Schumacher TN, Schreiber RD. 2015. Neoantigens in cancer immunotherapy. Science 348:69–74. doi: 10.1126/science.aaa4971. [DOI] [PubMed] [Google Scholar]
  • 7.Hakozaki T, Okuma Y, Omori M, Hosomi Y. 2019. Impact of prior antibiotic use on the efficacy of nivolumab for non-small cell lung cancer. Oncol Lett 17:2946–2952. doi: 10.3892/ol.2019.9899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zitvogel L, Ma Y, Raoult D, Kroemer G, Gajewski TF. 2018. The microbiome in cancer immunotherapy: diagnostic tools and therapeutic strategies. Science 359:1366–1370. doi: 10.1126/science.aar6918. [DOI] [PubMed] [Google Scholar]
  • 9.Quigley EMM. 2016. Leaky gut-concept or clinical entity? Curr Opin Gastroenterol 32:74–79. doi: 10.1097/MOG.0000000000000243. [DOI] [PubMed] [Google Scholar]
  • 10.Geva-Zatorsky N, Sefik E, Kua L, Pasman L, Tan TG, Ortiz-Lopez A, Yanortsang TB, Yang L, Jupp R, Mathis D, Benoist C, Kasper DL. 2017. Mining the human gut microbiota for immunomodulatory organisms resource mining the human gut microbiota for immunomodulatory organisms. Cell 168:928–943. doi: 10.1016/j.cell.2017.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Gong J, Chehrazi-Raffle A, Placencio-Hickok V, Guan M, Hendifar A, Salgia R. 2019. The gut microbiome and response to immune checkpoint inhibitors: preclinical and clinical strategies. Clin Transl Med 8:9. doi: 10.1186/s40169-019-0225-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kuczma MP, Ding ZC, Li T, Habtetsion T, Chen T, Hao Z, Bryan L, Singh N, Kochenderfer JN, Zhou G. 2017. The impact of antibiotic usage on the efficacy of chemoimmunotherapy is contingent on the source of tumor-reactive T cells. Oncotarget 8:111931–111942. doi: 10.18632/oncotarget.22953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, Poirier-Colame V, Roux A, Becharef S, Formenti S, Golden E, Cording S, Eberl G, Schlitzer A, Ginhoux F, Mani S, Yamazaki T, Jacquelot N, Enot DP, Bérard M, Nigou J, Opolon P, Eggermont A, Woerther PL, Chachaty E, Chaput N, Robert C, Mateus C, Kroemer G, Raoult D, Boneca IG, Carbonnel F, Chamaillard M, Zitvogel L. 2015. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350:1079–1084. doi: 10.1126/science.aad1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, Fidelle M, Flament C, Poirier-Colame V, Opolon P, Klein C, Iribarren K, Mondragón L, Jacquelot N, Qu B, Ferrere G, Clémenson C, Mezquita L, Masip JR, Naltet C, Brosseau S, Kaderbhai C, Richard C, Rizvi H, Levenez F, Galleron N, Quinquis B, Pons N, Ryffel B, Minard-Colin V, Gonin P, Soria JC, Deutsch E, Loriot Y, Ghiringhelli F, Zalcman G, Goldwasser F, Escudier B, Hellmann MD, Eggermont A, Raoult D, Albiges L, Kroemer G, Zitvogel L. 2018. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359:91–97. doi: 10.1126/science.aan3706. [DOI] [PubMed] [Google Scholar]
  • 15.Goubet AG, Daillère R, Routy B, Derosa L, M Roberti P, Zitvogel L. 2018. The impact of the intestinal microbiota in therapeutic responses against cancer. C R Biol 341:284–289. doi: 10.1016/j.crvi.2018.03.004. [DOI] [PubMed] [Google Scholar]
  • 16.Villéger R, Lopès A, Carrier G, Veziant J, Billard E, Barnich N, Gagnière J, Vazeille E, Bonnet M. 2019. Intestinal microbiota: a novel target to improve anti-tumor treatment? Int J Mol Sci 20:4584. doi: 10.3390/ijms20184584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Vétizou M, Daillère R, Zitvogel L. 2016. Role du microbiote intestinal dans la réponse aux thérapies anti-tumorales. Med Sci 32:974–982. [DOI] [PubMed] [Google Scholar]
  • 18.Ma W, Mao Q, Xia W, Dong G, Yu C, Jiang F. 2019. Gut microbiota shapes the efficiency of cancer therapy. Front Microbiol 10:1050. doi: 10.3389/fmicb.2019.01050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, Chau I, Ernstoff MS, Gardner JM, Ginex P, Hallmeyer S, Holter Chakrabarty J, Leighl NB, Mammen JS, McDermott DF, Naing A, Nastoupil LJ, Phillips T, Porter LD, Puzanov I, Reichner CA, Santomasso BD, Seigel C, Spira A, Suarez-Almazor ME, Wang Y, Weber JS, Wolchok JD, Thompson JA, National Comprehensive Cancer Network. 2018. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol 36:1714–1768. doi: 10.1200/JCO.2017.77.6385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Drilon A, Wolchok JD, Carvajal RD, McHenry MB, Hosein F, Harbison CT, Grosso JF, Sznol M. 2019. Five-Year survival and correlates among patients with advanced melanoma, renal cell carcinoma, or non–small cell lung cancer treated with nivolumab. JAMA Oncol 5:1411–1420. doi: 10.1001/jamaoncol.2019.2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dubin K, Callahan MK, Ren B, Khanin R, Viale A, Ling L, No D, Gobourne A, Littmann E, Huttenhower C, Pamer EG, Wolchok JD. 2016. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun 7:10391–10398. doi: 10.1038/ncomms10391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, Boselli L, Routier E, Cassard L, Collins M, Vaysse T, Marthey L, Eggermont A, Asvatourian V, Lanoy E, Mateus C, Robert C, Carbonnel F. 2017. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol 28:1368–1379. doi: 10.1093/annonc/mdx108. [DOI] [PubMed] [Google Scholar]
  • 23.Ahmed J, Kumar A, Parikh K, Anwar A, Knoll BM, Puccio C, Chun H, Fanucchi M, Lim SH. 2018. Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors. Oncoimmunology 7:e1507670–e1507676. doi: 10.1080/2162402X.2018.1507670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Derosa L, Hellmann MD, Spaziano M, Halpenny D, Fidelle M, Rizvi H, Long N, Plodkowski AJ, Arbour KC, Chaft JE, Rouche JA, Zitvogel L, Zalcman G, Albiges L, Escudier B, Routy B. 2018. 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 29:1437–1444. doi: 10.1093/annonc/mdy103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Elkrief A, El Raichani L, Richard C, Messaoudene M, Belkaid W, Malo J, Belanger K, Miller W, Jamal R, Letarte N, Wong P, Routy B. 2019. Antibiotics are associated with decreased progression-free survival of advanced melanoma patients treated with immune checkpoint inhibitors. Oncoimmunology 8:e1568812–e1568816. doi: 10.1080/2162402X.2019.1568812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Farmakiotis D. 2020. The human microbiome and checkpoint inhibition: potential benefits from antibiotic stewardship. Clin Infect Dis 71:701–702. doi: 10.1093/cid/ciz1003. [DOI] [PubMed] [Google Scholar]
  • 27.Kaderbhai C, Richard C, Fumet JD, Aarnink A, Foucher P, Coudert B, Favier L, Lagrange A, Limagne E, Boidot R, Ghiringhelli F. 2017. Antibiotic use does not appear to influence response to nivolumab. Anticancer Res 3200:3195–3200. doi: 10.21873/anticanres.11680. [DOI] [PubMed] [Google Scholar]
  • 28.Huang XZ, Gao P, Song YX, Xu Y, Sun JX, Chen XW, Zhao JH, Wang ZN. 2019. Antibiotic use and the efficacy of immune checkpoint inhibitors in cancer patients: a pooled analysis of 2740 cancer patients. Oncoimmunology 8:e1665973. doi: 10.1080/2162402X.2019.1665973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chalabi M, Cardona A, Nagarkar DR, Dhawahir Scala A, Gandara DR, Rittmeyer A, Albert ML, Powles T, Kok M, Herrera FG, imCORE Working Group of Early Career Investigators. 2020. 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 31:525–531. doi: 10.1016/j.annonc.2020.01.006. [DOI] [PubMed] [Google Scholar]
  • 30.Lurienne L, Cervesi J, Duhalde L, de Gunzburg J, Andremont A, Zalcman G, Buffet R, Bandinelli PA. 2020. NSCLC immunotherapy efficacy and antibiotic use: a systematic review and meta-analysis. J Thorac Oncol 15:1147–1159. doi: 10.1016/j.jtho.2020.03.002. [DOI] [PubMed] [Google Scholar]
  • 31.Della Corte CM, Morgillo F. 2019. Early use of steroids affects immune cells and impairs immunotherapy efficacy. ESMO Open 4:e000477–e000479. doi: 10.1136/esmoopen-2018-000477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Huang EY, Inoue T, Leone VA, Dalal S, Touw K, Wang Y, Musch MW, Theriault B, Higuchi K, Donovan S, Gilbert J, Chang EB. 2015. Using corticosteroids to reshape the gut microbiome: implications for inflammatory bowel diseases. Inflamm Bowel Dis 21:963–972. doi: 10.1097/MIB.0000000000000332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Arbour KC, Mezquita L, Long N, Rizvi H, Auclin E, Ni A, Martínez-Bernal G, Ferrara R, Lai WV, Hendriks LEL, Sabari JK, Caramella C, Plodkowski AJ, Halpenny D, Chaft JE, Planchard D, Riely GJ, Besse B, Hellmann MD. 2018. 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 36:2872–2878. doi: 10.1200/JCO.2018.79.0006. [DOI] [PubMed] [Google Scholar]
  • 34.Martínez-Bernal G, Mezquita L, Auclin E, Ferrara R, Planchard D, Remon Masip J, Lahmar J, Boucher ME, Caramella C, Adam J, Gazzah A, Soria JC, Besse B. 2017. Baseline corticosteroids (CS) could be associated with absence of benefit to immune checkpoint inhibitors (ICI) in advanced non-small cell lung cancer (NSCLC) patients. Ann Oncol 28:1323P. doi: 10.1093/annonc/mdx380.025. [DOI] [Google Scholar]
  • 35.Dumenil C, Massiani MA, Dumoulin J, Giraud V, Labrune S, Chinet T, Giroux Leprieur E. 2018. Clinical factors associated with early progression and grade 3–4 toxicity in patients with advanced non-small-cell lung cancers treated with nivolumab. PLoS One 13:e0195945-15. doi: 10.1371/journal.pone.0195945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Scott SC, Pennell NA. 2018. Early use of systemic corticosteroids in patients with advanced NSCLC treated with nivolumab. J Thorac Oncol 13:1771–1775. doi: 10.1016/j.jtho.2018.06.004. [DOI] [PubMed] [Google Scholar]
  • 37.Drakaki A, Luhn P, Wakelee H, Dhillon PK, Kent M, Shim J, Degaonkar V, Hoang T, McNally V, Chui SY, Gutzmer R. 2019. Association of systemic corticosteroids with overall survival in patients receiving cancer immunotherapy for advanced melanoma, non-small cell lung cancer or urothelial cancer in routine clinical practice. Ann Oncol 30:xi16–xi17. doi: 10.1093/annonc/mdz449.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.De Giglio A, Mezquita L, Auclin E, Blanc-Durand F, El-Amarti L, Caramella C, Bernal GM, Hendriks L, Ferrara R, Naltet C, Lavaud P, Gazzah A, Adam J, Planchard D, Chaput N, Besse B. 2019. Impact of early introduction of steroids on immune checkpoint inhibitors (ICI) in patients with advanced non-small-cell lung cancer. Ann Oncol 30:xi32. doi: 10.1093/annonc/mdz449. [DOI] [Google Scholar]
  • 39.Indini A, Rijavec E, Grossi F. 2020. Immune related adverse events and response to immunotherapy: focus on corticosteroids. Lung Cancer 145:225. doi: 10.1016/j.lungcan.2020.02.009. [DOI] [PubMed] [Google Scholar]
  • 40.Heinhuis KM, Carlino M, Joerger M, Di Nicola M, Meniawy T, Rottey S, Moreno V, Gazzah A, Delord J-P, Paz-Ares L, Britschgi C, Schilder RJ, O’Byrne K, Curigliano G, Romano E, Patah P, Wang R, Liu Y, Bajaj G, Siu LL. 2020. Safety, tolerability, and potential clinical activity of a glucocorticoid-induced TNF receptor-related protein agonist alone or in combination with nivolumab for patients with advanced solid tumors: a phase 1/2a dose-escalation and cohort-expansion clinic. JAMA Oncol 6:100–107. doi: 10.1001/jamaoncol.2019.3848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Panebianco C, Andriulli A, Pazienza V. 2018. Pharmacomicrobiomics: exploting the drug-microbiota interactions in anticancer therapies. Microbiome 6:92. doi: 10.1186/s40168-018-0483-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Górska A, Przystupski D, Niemczura MJ, Kulbacka J. 2019. Probiotic bacteria: a promising tool in cancer prevention and therapy. Curr Microbiol 76:939–949. doi: 10.1007/s00284-019-01679-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre ML, Chang EB, Gajewski TF. 2015. Commensal Bifidobacterium promotes antitumor immunity and facilitates andi-PD-L1 efficacy. Science 350:1084–1089. doi: 10.1126/science.aac4255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Abdelhamid AG, El-Masry SS, El-Dougdoug NK. 2019. Probiotic Lactobacillus and Bifidobacterium strains possess safety characteristics, antiviral activities and host adherence factors revealed by genome mining. EPMA J 10:327–350. doi: 10.1007/s13167-019-00184-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. 2019. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol 16:605–616. doi: 10.1038/s41575-019-0173-3. [DOI] [PubMed] [Google Scholar]

Articles from Infection and Immunity are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES