Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2020 Nov 9;70(6):1511–1517. doi: 10.1007/s00262-020-02786-3

Antibiotics and steroids, the double enemies of anticancer immunotherapy: a review of the literature

Fausto Meriggi 1,, Alberto Zaniboni 1
PMCID: PMC10991597  PMID: 33165628

Abstract

The advent of immunotherapy in onco-haematology has represented a kind of revolution that has been able to modify the prognosis of numerous tumours that until recently would have been rapidly lethal. While much is known about the mechanism of action of these drugs, relatively little is known about the factors that represent potential predictors of response and toxicity. Among these factors, the simultaneous administration of antibiotics and/or steroids seems to have a negative impact. Furthermore, several retrospective studies have highlighted the strong link between cancer and gut microbiota, regardless of the tumour site, and how microbiota, playing a key role in the prevention of systemic inflammation at various levels and in the intestinal homeostasis, can be negatively influenced by the dysbiosis caused by antibiotic therapy administered during or in the weeks immediately preceding the start of immunotherapy. Moreover, we assume that the concurrent administration of steroids, which is often necessary in cancer patients, likely results in a deleterious effect on the therapeutic outcomes of immunotherapy. In this review, we will try to clarify the evidence on the possible detrimental effects of antibiotics and steroids, which are currently considered the double enemies of anticancer immunotherapy.

Keywords: Immunotherapy, Antibiotic therapy, Gut microbiota, Corticosteroids

Introduction

The advent and development of new cancer treatments such as immunotherapy (IO) have led to significant improvements, especially for some patients who have been able to benefit from or experience dramatic and lasting responses, even in advanced cancers that until a few years ago were considered to have a bad prognosis with extremely short survival times. The main mechanism of action of the immune checkpoint inhibitors (ICIs) consists of reversing the blockade of the inhibitory effect between the T lymphocyte receptor and its ligand located on neoplastic cells or myeloid cells, thus allowing the T lymphocyte-mediated response against tumour antigens. ICIs are monoclonal antibodies (mAbs) that target PD-1 (programmed cell death-1) and PD-L1 (programmed cell death ligand-1) or target cytotoxic T-lymphocyte antigen 4 (CTLA-4), which act to enable cytotoxic lymphocytes to regain their ability to attack tumour cells [1]. However, the magnitude of the responses and the spectrum of toxicities to IO are still extremely variable, and further research is needed to identify more reliable predictive factors, particularly those concerning the host. A refractoriness to ICIs is present, depending on study, in over 1/3 of treated patients, and often the main causes remain obscure [2]. Among the predictors of response to treatment with ICIs, the levels of PD-L1, tumour mutation burden (TMB), gene expression signatures of inflammation, neutrophil-to-lymphocyte ratio (NLR), and state of microsatellites at the baseline were recently identified [38]. However, in this context, the most promising would seem to be represented today by the modulation of gut microbiota by antibiotic therapy (ATB). Understanding the impact of the gut microbiome on immune function may be the key to developing consistently effective immunotherapy. It is now an accepted concept that, in the context of induced dysbiosis, the gut microbiota is able to alter the systemic response of the immune system, potentially contributing to the onset of chronic inflammatory disorders such as obesity, Crohn's disease and diabetes mellitus type 2 [9]. Therefore, gut microbiota is able to play a key role, even in the context of the responses to IO. Not surprisingly, this approach revealed that the gut microbiota from patients (pts) who failed to benefit from ICIs differed in its composition from those who responded [1020]. A similar potential detrimental effect is observed, but through a very different mechanism of action, as result of a concurrent treatment with steroids and IO. Steroids are often necessarily administered to cancer pts to relieve symptoms and have been able to reduce the response to IO, as demonstrated by overall response rate (ORR), progression free survival (PFS) and overall survival (OS). In this review, we try to clarify the current evidence on the possible detrimental effects of the associations between ATB and IO and between steroids and IO.

Antibiotics and immunotherapy

Putative mechanism of the detrimental effect

Tumours, especially at an advanced stage, are often accompanied by inflammation and infections that frequently require ATB, according to the guidelines [15]. It is known that the gut microbiota is implicated in numerous physiological and pathological processes in humans and is closely related to a good functioning of the immune system. Several preclinical studies have highlighted the role of the gut microbiota in regulating responses to IO [17, 18]. First, in mouse models, the lack of an intact gut microbiota (in germ-free or ATB-treated mice) dramatically decreased the antitumour activity of ICIs induced by CD4-positive T cells. As a result, the faecal microbiota transplantation in these germ-free mice from pts treated with immune checkpoint blockade restored the activity of ICIs [27]. Moreover, observational studies showed the relevance of geographical locations, differences in the composition of the gut microbiota and responses to the IO [19, 20, 27]. ATBs can induce intestinal dysbiosis through the diminution of the abundance of critical species or through the complete eradication of important commensals, therefore, altering normal gut homeostasis with effects on local and whole-body metabolism, as well as having major consequences for the immune system. Several retrospective studies have highlighted the strong link between cancer development and gut microbiota [1021, 23], regardless of the primary tumour site, and how microbiota, playing a key role in the prevention of systemic inflammation at various levels and in the intestinal homeostasis, can be negatively influenced by the dysbiosis caused by ATB administered during or in the weeks immediately preceding the start of IO [10, 2430, 30, 3335]. Recently, some studies showed the possible promoting effect of a previous ATB on the onset of cancer. This result could be explained merely by modulation of the intestinal microbiota. Bacteria induce carcinogenesis through two main mechanisms: the induction of chronic inflammatory processes leading to cancer in various human organs, or the production of carcinogenic metabolites. A recent large meta-analysis by Petrelli et al. has shown that the use of ATB represents an independent risk factor for the development of cancer (OR 1.18, 95% CI 1.12–1.24, p < 0.001), especially for lung, kidney and pancreatic cancer, as well as for some lymphomas and multiple myeloma [21]. A similar meta-analysis by Zhang et al. showed that the administration of oral broad-spectrum ATB may be associated with an increased risk of colon cancer, especially in the right colon, probably due to the differences in gut microbiota and carcinogenesis mechanisms [23].

Clinical evidence (Table 1)

Table 1.

Retrospective clinical trials addressing the impact of ATB on the activity of ICIs

Author
(ref)
Primary tumor site
(number of evaluable pts)
Antibiotic window Outcome (p-values)
Galli [29] NSCLC (157) 1 month PRE or 3 mos POST ↓ PFS (0.0001) and ↓ OS (0.0004)
Huemer [30] NSCLC (30) 1 month PRE or 1 month POST ↓ PFS (0.031) and ↓ OS (0.021)
Lurienne [34] Multiple Sites (2208 for PFS and 5560 for OS) 2 mos PRE or 2 mos POST ↓ PFS and ↓ OS
Thompson [61] NSCLC (74) 6 wks PRE ↓ PFS (0.02) and ↓ OS (0.004)
Rubio [62] NSCLC (168) 2 mos PRE or 1 month POST ↓ PFS (0.028) and ↓ OS (0.026)
Ouaknine [63] NSCLC (72) 2 mos PRE or 1 month POST ↓ OS (0.03)
Do [64] NSCLC (109) 1 month PRE or 1 month POST ↓ OS (0.0004)
Zhao [65] NSCLC (109) 1 month PRE or 1 month POST ↓ PFS (0.0001) and ↓ OS (0.0021)
Schett [66] NSCLC (218) 2 mos PRE ↓ PFS (0.001) and ↓ OS (0.001)
Lalani [67] RCC (146) 2 mos PRE or 1 month POST ↓ PFS (0.08) and ↓ ORR (0.026)
Mohiuddin [35] Melanoma (114) 45 days PRE or 45 days POST ↓ OS (0.001)
Elkrief [68] Melanoma (74) 30 days PRE ↓ PFS (0.01) and ↓ ORR (0.01)
Tinsley [24] NSCLC (58), RCC (46), Melanoma (201) 14 days PRE or 42 days POST ↓ PFS (0.049) and ↓ OS (0.001)
Routy [27] NSCLC (140), RCC (67), UC (32) 2 mos PRE or 1 month POST ↓ PFS (0.017) and ↓ OS (0.001)
Derosa [28] NSCLC (249), RCC (121) 1 month PRE ↓ PFS (0.001) and ↓ OS (0.001)
Pinato [30] NSCLC (119), Melanoma (38), Other (39) 30 days PRE or concurrently ↓ OS (0.001) and ↓ ORR (0.01) (PRE)
Masini [69] NSCLC (78), Melanoma (57), RCC (29), Other (5) Any time ↑ OS (0.028)
Kulkarni [70] NSCLC* (148), RCC** (55) 1 month PRE or any time POST

↑ PFS (0.01)*, ↑ OS (0.01)*, ↑ ORR (0,01)*

↓ PFS (0.04)**

Ref reference, pts patients, NSCLC non-small-cell lung cancer, RCC renal cell carcinoma, UC urothelial cancer, ATB antibiotic therapy, ICIs immune checkpoint inhibitors, PFS progression free survival, ORR overall response rate, OS overall survival, mos months

The main evidence derived from researches reported over the last 10 years confirms that ATB exposure tends to increase cancer risk and, unfortunately, that it reduces the efficacy of cancer therapies. Exposure to a broad-spectrum ATB has been shown to be able to negatively influence the results of IO with ICIs by the modulation of gut microbiota. Several studies have highlighted this close correlation between ATB and ICIs [2, 10, 2430, 30, 3335]. However, to date, little is known about the potentially detrimental timing of ATB administration and the start of IO. A recently published retrospective study by Tinsley et al. demonstrated that cumulative or prolonged use of ATB is an independent negative predictor of PFS and OS in pts with advanced cancer treated with ICIs [24]. In a prospective study, Pinato et al. evaluated the correlation of ATB with ORR and OS in approximately 200 cancer pts treated with ICIs. This study showed that there is a statistically negative correlation for all outcomes in the case of ATB administration before the beginning of treatment with ICIs but not in the case of the concurrent administration of ATB and IO [30]. Derosa et al. examined 121 pts with advanced renal cell carcinoma (RCC) and 239 with non-small cell lung cancer (NSCLC) treated with anti-PD-L1 mAbs. Those receiving ATB within 30 days of beginning ICIs were compared with those who did not. ATB administration was associated with an increased risk of primary progressive disease and shorter PFS and OS. The same trend was observed if ATB was administered within 60 days of starting ICI [2]. These findings suggest a time-dependent and partial repopulation of the gut microbiota after ATB discontinuation. Some studies have shown that it requires from 1 to 3 months after the suspension of the ATB for the microbiota to return to the initial situation, but it sometimes requires years to obtain a full recovery [31, 32]. Therefore, in the current state of knowledge, what has been highlighted with certainty is the existence of a correlation between ATB and a poor outcome in pts treated with ICIs. However, it is debated whether ATB use represents a true independent therapeutic biomarker or is a surrogate for pts with a worse overall prognosis (pts who required ATB use often had additional comorbidities). Wilson et al. recently published a systematic review and large meta-analyses of 766 observational studies on ATB and ICIs. Eighteen studies and 826 pts met the inclusion criteria for analyses. OS was 3.4 times longer in pts who did not receive any ATB in the 42 days prior to IO (HR 3.43, 95% CI 2.29–5.14, p < 0.0001), and PFS was also longer in pts who did not receive ATB (pooled HR 1.65, 95% CI 1.3–2.1, p < 0.0001). Conversely, exposure to ATB before 60 days starting or during IO seems not to influence the clinical outcomes of IO [33]. Moreover, two further reviews and meta-analyses of Lurienne et al. [34] and Mohiuddin et al. [35] explored and confirmed this detrimental link between the time of ATB exposure and the start of IO both in NSCLC and melanoma pts, respectively. Finally, it is known that there are still several obscure points, such as the time frame at which ATB administration is more deleterious for IO, the basal state of the gut microbiota and the possibility of intervening with treatments such as faecal transplantation [36, 37], diets, prebiotics and probiotics aimed at preventing and remediating ATB-induced dysbiosis. Therefore, manipulation of the microbiota for therapeutic benefits is a major goal. Further prospective studies are warranted to confirm these findings.

Steroids and immunotherapy

Mechanism of the detrimental effect

As a consequence of the well-known immunosuppressive effect of corticosteroids, today these drugs are considered able to reduce the efficacy of immunotherapy, and the guidelines report the use of simultaneous steroids as a relative contraindication. Giles et al. showed that the immunosuppressive effect of dexamethasone (DEX) consisted of upregulation of CTLA-4 mRNA and protein in CD4 and CD8 T cells and blocked CD28-mediated cell cycle entry and differentiation. Therefore, it seems that CTLA-4, but not PD-1 blockade, can partially prevent some of the negative effects of DEX on the immune response [38, 39]. Corticosteroids perform their immunosuppressive activity by inhibition of certain priority T-lymphocyte functions by suppressing IL-2-mediated activation of effector T cells [40] and the increase in regulatory T lymphocytes [41]. Furthermore, it seems that steroids are able to cause the alteration of the microbiome [42] and stimulate M2 polarization of macrophages [43]. However, their use and the clinical implications of an interaction between IO and steroids remain unclear. Pts with refractory anorexia, nausea, fatigue, pain, brain metastases, and dyspnoea require care and are often given corticosteroids as the main treatment for symptom palliation. Moreover, ICIs cause adverse events (irAEs) that can involve any organ, and steroid use represents the first antidote.

Clinical evidence

The relationship between steroids and IO remains very controversial. Because of their immunosuppressive properties, corticosteroids are both the principal treatment of irAEs due to ICIs [44] and an exclusion criterion for ICIs clinical trials considering a threshold of ≥ 10 mg of prednisone equivalent daily the usual cutoff [45, 46]. Usually, when irAEs are grade 2 or higher, steroid administration such as prednisone is used at a dosage of 0.5–2 mg/kg/day with a possible subsequent tapering of the induction dose if the symptoms resolve [47]. This management of irAEs does not seem to have consequences on the efficacy of IO [48] or the retreatment with ICIs after serious irAEs [4951] or significantly suppress the function of T cells [52, 53]. A large systematic review by Garant et al. showed that there was no objective data on the exact types of corticosteroids and the dose threshold above which an interaction could be measured clinically [54]. However, Arbor et al. demonstrated that pts on IO and who were taking 10 mg/day or more prednisone when they initiated ICIs for advanced NSCLC reported a significantly inferior objective RR, PFS and/or OS compared with those who used less than 10 mg/day of prednisone or the equivalent [55]. Doses > 10 mg of prednisone daily seem related to increasing infection rates in long-term steroids treated pts [56], and are considered immunosuppressive [57]. Another unknown point is the duration of simultaneous treatment with steroids. In a retrospective study recently published by Pan et al., it seems that prednisone > 10 mg/day for > 2 weeks may be associated with poorer survival outcomes [58]. De Giglio et al. found that early introduction of steroid therapy (≥ 10 mg of prednisone or equivalent) in 49 advanced NSCLC pts led to significantly poorer outcomes if steroids were administered for cancer-related symptoms but did not have a detrimental impact on prognosis if the steroids were started for other indications such as irAEs or COPD exacerbation [59]. In a large retrospective observational study of 2213 pts with NSCLC, advanced melanoma and advanced urothelial cancer, Drakaki et al. showed that pts taking baseline corticosteroids (19–30%) were more likely to have stage IV disease at diagnosis, brain and/or liver metastases and poorer ECOG PS scores at baseline. The administration of baseline corticosteroids was related to a 23–47% higher risk of death when compared with no use in the multivariable models (Table 2) [60].

Table 2.

Pts characteristics by baseline CS use, and OS HR by CS use (modified from Drakaki et al. [60])

NSCLC (n = 862) Melanoma (n = 742) UC (n = 609)
CS
(n = 258)
No CS
(n = 604)
CS
(n = 182)
No CS
(n = 560)
CS
(n = 116)
No CS
(n = 493)
Stage IV at diagnosis 74 61 34 29 42 35
ECOG PS ≥ 2 at ICI start 17 15 9 11 34 21
Site of metastases
 Brain 26 19 31 21 5 2
Multivariable OS, HR (95% CI), CS use vs. no CS use (reference)
 Model 1 1.35 (1.12, 1.62) 1.23 (0.97, 1.57) 1.47 (1.14, 1.90)
 Model 2 1.34 (1.12, 1.61) 1.24 (0.97, 1.57) 1.44 (1.12, 1.87)

Multivariable models adjusted for age at ICI start, stage at diagnosis, race/ethnicity, ECOG PS and Charlson Comorbidity Index at ICI start; treatment sequence, brain metastases at ICI start, smoking status (NSCLC, UC), histology (NSCLC), grade (UC) in model 1 and prior steroid use in model 2

Pts patients, CS corticosteroids, NSCLC non-small cell lung cancer, UC urothelial cancer, ICI immune checkpoint inhibitors, ECOG PS Eastern Cooperative Oncology Group Performance Status, HR hazard ratio

Discussion

IO has led to drastic positive changes in the prognosis of a broad range of advanced tumours such as melanoma, NSCLC, and renal cell carcinoma, which had a poor prognosis until only a few years ago. Today, due to IO, we can truly discuss the “chronicity” of advanced cancer. However, only a small proportion of pts benefit significantly from this treatment. For this reason, future research will focus on identifying new predictive response factors and on defining the roles of the old ones. Knowledge resulting mainly from retrospective studies has taught us that the two well-identified "enemies" of IO are represented by ATB, mainly due to the dysbiosis they cause at the intestinal level, and by steroids administered concurrently with ICIs. Many points still remain obscure and are worthy of further research. The real mechanisms that can determine these negative interferences are not yet completely known, such as the timing of administration of ATB (e.g., how long before the start of IO or whether simultaneous administration is acceptable). Furthermore, does this concept apply only to some broad-spectrum ATB or could it be extended to all ATB? What are the components of the gut microbiota that are most negatively affected? Are there effective remedies to overcome this problem, such as faecal transplantation, diets, prebiotics, or probiotics? These findings reaffirm the importance for physicians to use ATB judiciously, avoiding the use of ATB unless for life-threatening conditions, especially in responding or stable pts during IO.

On the other hand, with IO widely becoming the standard of care for pts with various advanced cancers, an important pragmatic concern regarding the concurrent use of ICIs and corticosteroids has emerged. It seems clear to avoid steroid use if possible, but there are some good practical points to consider. First, in the case of symptomatic brain metastases, we should try to use the lowest effective dose of prednisone or equivalent as soon as possible. Second, in the case of irAEs, we should provide a quick tapering of induction dose if clinically indicated. Finally, we should try to avoid use of steroids for relief of symptoms such as pain, fatigue, appetite loss or paraneoplastic fever. The type and the allowed dosage of administered corticosteroids with concurrent IO (e.g., prednisone < 10 mg/day) remain open questions, as well as the question of whether a simultaneous administration of chemotherapy plus IO may overcome these negative influences.

Providing answers to these questions remains one of the main exciting challenges in the field of onco-haematology awaiting us in the coming years.

Footnotes

Publisher's Note

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

References

  • 1.Pardoll D. Cancer and the immune system: basic concepts and targets for intervention. Semin Oncol. 2015;42:523–538. doi: 10.1053/j.seminoncol.2015.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Derosa L, 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;29:1437–1444. doi: 10.1093/annonc/mdy103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Motzer RJ, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373:1803–1813. doi: 10.1056/NEJMoa1510665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Escudier B, et al. CheckMate 025 randomized phase 3 study: outcomes by key baseline factors and prior therapy for nivolumab versus everolimus in advanced renal cell carcinoma. EurUrol. 2017 doi: 10.1016/j.eururo.2017.02.010. [DOI] [PubMed] [Google Scholar]
  • 5.Borghaei H, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373:1627–1639. doi: 10.1056/NEJMoa1507643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Reck M, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823–1833. doi: 10.1056/NEJMoa1606774. [DOI] [PubMed] [Google Scholar]
  • 7.Carbone DP, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376:2415–2426. doi: 10.1056/NEJMoa1613493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847–856. doi: 10.1158/1535-7163.MCT-14-0983. [DOI] [PubMed] [Google Scholar]
  • 9.Cho I, Blaser MJ. The human microbiome: at the interface of health and disease. Nat Rev Genet. 2012;13:260–270. doi: 10.1038/nrg3182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gori S, Inno A, Belluomini L, Bocus P, Bisoffi Z, Russo A, Arcaro G. Gut microbiota and cancer: how gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit Rev Oncol Hemat. 2019;143:139–147. doi: 10.1016/j.critrevonc.2019.09.003. [DOI] [PubMed] [Google Scholar]
  • 11.Picardo SL, Coburn B, Hansen AR. The microbiome and cancer for clinicians. Crit Rev Oncol Hemat. 2019;141:1–12. doi: 10.1016/j.critrevonc.2019.06.004. [DOI] [PubMed] [Google Scholar]
  • 12.Alexander JL, Kohoutova D, Powell N. Science in focus: the microbiome and cancer therapy. Clin Oncol. 2019;31:1–4. doi: 10.1016/j.clon.2018.09.004. [DOI] [PubMed] [Google Scholar]
  • 13.McQuade JL, Daniel CR, Helmink BA, Wargo JA. Modulating the microbiome to improve therapeutic response in cancer. Lancet Oncol. 2019;20:e77–91. doi: 10.1016/S1470-2045(18)30952-5. [DOI] [PubMed] [Google Scholar]
  • 14.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;15:382–396. doi: 10.1038/s41571-018-0006-2. [DOI] [PubMed] [Google Scholar]
  • 15.Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. 2017;15:465–478. doi: 10.1038/nrmicro.2017.44. [DOI] [PubMed] [Google Scholar]
  • 16.Wong SW, Kwong TNY, Wu C-Y, Yu J. Clinical applications of gut microbiota in cancer biology. Semin Cancer Biol. 2019;55:28–36. doi: 10.1016/j.semcancer.2018.05.003. [DOI] [PubMed] [Google Scholar]
  • 17.Vétizou M, Pitt JM, Daillère R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–1084. doi: 10.1126/science.aad1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–1089. doi: 10.1126/science.aac4255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97–103. doi: 10.1126/science.aan4236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104–108. doi: 10.1126/science.aao3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Petrelli F, Ghidini M, Ghidini A, Perego G, Cabiddu M, et al. Use of antibiotics and risk of cancer: a systematic review and meta-analysis of observational studies. Cancers. 2019;11:1174. doi: 10.3390/cancers11081174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Milano G. Efficacy of immunotherapy, gut microbiota and impact of antibiotic use: are the confounding factors? Letter to editor. Cancer Chemother Pharmacol. 2019;84:223–224. doi: 10.1007/s00280-019-03833-2. [DOI] [PubMed] [Google Scholar]
  • 23.Zhang J, Haines C, Watson AJM, Hart AR, Platt MJ, et al. Oral antibiotic use and risk of colorectal cancer in the United Kingdom, 1989–2012: a matched case-control study. Gut. 2019;68:1971–1978. doi: 10.1136/gutjnl-2019-318593. [DOI] [PubMed] [Google Scholar]
  • 24.Tinsley N, Zhou C, Tan G, Rack S, Lorigan P, et al. Cumulative antibiotic use significantly decreases efficacy of checkpoint inhibitors in patients with advanced cancer. Oncologist. 2020;25:55–63. doi: 10.1634/theoncologist.2019-0160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Elkrief A, Derosa L, Kroemer G, Zitvogel L, Routy B (2019) The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: a new independent prognostic factor? Review. Ann Oncol. Published online 3 July 2019 [DOI] [PubMed]
  • 26.Elkrief A, Derosa L, Zitvogel L, Kroemer G, Routy B. The intimate relationship between gut microbiota and cancer immunotherapy. Gut. 2019;10:424–428. doi: 10.1080/19490976.2018.1527167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–97. doi: 10.1126/science.aan3706. [DOI] [PubMed] [Google Scholar]
  • 28.Derosa L, Hellmann MD, Spaziano M, Halpenny D, Fidelle M, 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;29:1437–1444. doi: 10.1093/annonc/mdy103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Galli G, Triulzi T, Proto C, Signorelli D, Imbimbo M, et al. Association between antibiotic-immunotherapy exposure ratio and outcome in metastatic non-small cell lung cancer. Lung Cancer. 2019;132:72–78. doi: 10.1016/j.lungcan.2019.04.008. [DOI] [PubMed] [Google Scholar]
  • 30.Huemer F, Rinnerthaler G, Westphal T, et al. Impact of antibiotic treatment on immune checkpoint blockade efficacy in advanced non-squamous non-small cell lung cancer. Oncotarget. 2018;9:16512–16520. doi: 10.18632/oncotarget.24751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lange K, Buerger M, Stallmach A, Bruns T. Effects of antibiotics on gut microbiota. Dig Dis. 2016;34:260–268. doi: 10.1159/000443360. [DOI] [PubMed] [Google Scholar]
  • 32.Jakobsson HE, Jernberg C, Andersson AF, et al. Short-term antibiotic treatment has differing long-term impacts on the human throat and gut microbiome. PLoS ONE. 2010;5:e9836. doi: 10.1371/journal.pone.0009836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wilson BE, Routy B, Nagrial A, Chin VT. The effect of antibiotics on clinical outcomes in immune-checkpoint blockade: a systematic review and meta-analysis of observational studies. Cancer ImmunolImmunother. 2020;69:343–354. doi: 10.1007/s00262-019-02453-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lurienne L, Cervesi J, Duhalde L, de Gunzburg J, Andremont A, et al. NSCLC immunotherapy efficacy and antibiotic use: a systematic review and meta-analysis. J Thorac Oncol. 2020;S1556–0864(20):30194–30195. doi: 10.1016/j.jtho.2020.03.002. [DOI] [PubMed] [Google Scholar]
  • 35.Mohiuddin JJ, Chu B, Facciabene A, Poirier K, Wang X, et al. Association of antibiotic exposure with survival and toxicity in patients with melanoma receiving immunotherapy. J Nat Cancer Inst. 2020 doi: 10.1093/jnci/djaa057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Choi HH, Cho Y-S. Fecalmicrobiota transplantation: current applications, effectiveness, and future perspectives. ClinEndosc. 2016;49:257–265. doi: 10.5946/ce.2015.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Allegretti JR, Mullish BH, Kelly C, Fisher M. The evolution of the use of faecal microbiota transplantation and emerging therapeutics indications. Lancet. 2019;394:420–431. doi: 10.1016/S0140-6736(19)31266-8. [DOI] [PubMed] [Google Scholar]
  • 38.Libert C, Dejager L. How steroids steer T cells. Cell Rep. 2014;7:938–939. doi: 10.1016/j.celrep.2014.04.041. [DOI] [PubMed] [Google Scholar]
  • 39.Giles AJ, Hutchinson M-KND, Sonnermann HM, Jung J, Fecci PE, et al. Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer. 2018 doi: 10.1186/s40425-018-0371-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bianchi M, Meng C, Ivashkiv LB. Inhibition of IL-2-induced Jak-STAT signaling by glucocorticoids. Proc Natl Acad Sci USA. 2000 doi: 10.1073/pnas.160099797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chen X, Oppenheim JJ, Winkler-Pickett RT, Ortaldo JR, Howard OMZ. Glucocorticoid amplifies IL-2-dependent expansion of functional FoxP3 + CD4 + CD25 + T regulatory cells in vivo and enhances their capacity to suppress EAE. Eur J Immunol. 2006 doi: 10.1002/eji.200635873. [DOI] [PubMed] [Google Scholar]
  • 42.Tetel MJ, de Vries GJ, Melcangi RC, Panzica G, O’Mahony SM. Steroids, stress and the gut microbiome-brain axis. J Neuroendocr. 2018 doi: 10.1111/jne.12548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sica A, Mantovani A. Macrophage plasticity and polarization: In vivo veritas. J ClinInvestig. 2012 doi: 10.1172/JCI59643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Haanen JBAG, Carbonnel F, Robert C, Kerr KM, Peters S, Larkin J, Jordan K, ESMO Guidelines Committee Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017 doi: 10.1093/annonc/mdx225. [DOI] [PubMed] [Google Scholar]
  • 45.Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, Felip E, van den Heuvel MM, Ciuleanu TE, Badin F, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017 doi: 10.1056/NEJMoa1613493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823–1833. doi: 10.1056/NEJMoa1606774. [DOI] [PubMed] [Google Scholar]
  • 47.National Comprehensive Cancer Network (NCCN). Management of immunotherapy-related toxicities. (version 2.2019) [DOI] [PubMed]
  • 48.Grob J et al. Antitumor responses to ipililumab in advanced melanoma are not affected by systemic corticosteroids used to manage irAEs (abstract). In: EJ Cancer supplement 2009; abstr n.9312
  • 49.Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378:158–168. doi: 10.1056/NEJMra1703481. [DOI] [PubMed] [Google Scholar]
  • 50.Santini FC, Rizvi H, Wilkins O, et al. Safety of retreatment with immunotherapy after immune- related toxicity in patients with lung cancers treated with anti-PD(L)-1 therapy. J Clin Oncol. 2017;35(suppl):9012–9021. [Google Scholar]
  • 51.Horvat TZ, Adel NG, Dang T-O, et al. Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at Memorial Sloan Kettering Cancer Center. J Clin Oncol. 2015;33:3193–3198. doi: 10.1200/JCO.2015.60.8448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hinrichs CS, et al. Glucocorticoids do not inhibit anti-tumor activity of activated CD8 + T cells. J Immunother. 2005;28:517–524. doi: 10.1097/01.cji.0000177999.95831.7b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Franchimont D et al. (20022003) Glucorticoids and inflammation revisited: the state of the art. NIH clinical staff conference. Neuroimmunimodulation10:247–260 [DOI] [PubMed]
  • 54.Garant A, Guilbault C, Ekmekjian T, Greenwald Z, Murgoi P, et al. Concomitant use of corticosteroids and immune checkpoint inhibitors in patients with hematologic or solid neoplasms: a systematic review. Crit Rev Oncol Hematol. 2018;120:86–92. doi: 10.1016/j.critrevonc.2017.10.009. [DOI] [PubMed] [Google Scholar]
  • 55.Arbour KC, Mezquita L, Long N, Rizvi H, Auclin E, 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;28:2872–2878. doi: 10.1200/JCO.2018.79.0006. [DOI] [PubMed] [Google Scholar]
  • 56.Wolfe F, Caplan L, Michaud K. Treatment for rheumatoid arthritis and the risk of hospitalization for pneumonia: associations with prednisone, disease-modifying antirheumatic drugs, and anti-tumor necrosis factor therapy. Arthritis Rheum. 2006 doi: 10.1002/art.21568. [DOI] [PubMed] [Google Scholar]
  • 57.Petrelli F, Signorelli D, Ghidini M, Ghidini A, Pizzutilo EG, et al. Association of steroids use with survival in patients treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Cancers. 2020;12:546–558. doi: 10.3390/cancers12030546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Pan EY, Merl MY, Lin K (2019). The impact of corticosteroids use during anti-PD1 treatment. J Oncol Pharm Practice. (Epub ahead of print) [DOI] [PubMed]
  • 59.De Giglio A, Mezquita L, Auclin E, Blanc-Durand F, El-Amarti L, et al. Impact of early introduction of steroid on immune-checkpoint inhibitors (ICI) in patients with advanced non-small cell lung cancer treated. Ann Oncol. 2019 doi: 10.1093/annonc/mdz449. [DOI] [Google Scholar]
  • 60.Drakaki A, Luhn P, Wakelee H, Dhillon PK, Kent M, et al. 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. 2019 doi: 10.1093/annonc/mdz449.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Thompson J, Szabo A, Arce-Lara C, Menon S. Microbiome & immunotherapy: antibiotic use is associated with inferior survival for lung cancer patients receiving PD-1 inhibitors. J Thorac Oncol. 2017;12(suppl 2):S1998. [Google Scholar]
  • 62.Mielgo-Rubio X, Chara L, Sotelo-Lezama M, et al. 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(suppl):S389. [Google Scholar]
  • 63.Ouaknine J, De Tauriers PH, Dumenil C, et al. Plasmatic evaluation of the intestinal barrier and blood microbiota, and antibiotic use in non-small cell lung cancer patients treated with nivolumab. J Thorac Oncol. 2018;13(suppl):S389–S390. [Google Scholar]
  • 64.Do TP, Hegde AM, Cherry CR, et al. Antibiotic use and overall survival in lung cancer patients receiving nivolumab. J Clin Oncol. 2018;36:e15109. [Google Scholar]
  • 65.Zhao S, Gao G, Li W, 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. [DOI] [PubMed] [Google Scholar]
  • 66.Schett A, Rothschild SI, Mauti LA, et al. Prognostic impact of the use of antibiotics in patients with advanced non-small cell lung cancer receiving PD-(L)1 targeting monoclonal antibodies. ESMO Congress 2019. Abstract 157P. Presented September 29
  • 67.Lalani AKA, Xie W, Lin X, et al. Antibiotic use and outcomes with systemic therapy in metastatic renal cell carcinoma. J Clin Oncol. 2018;36:607. [Google Scholar]
  • 68.Elkrief A, El Raichani L, Richard C, et al. Antibiotics are associated with decreased progression free survival of advanced melanoma patients treated with immune checkpoint inhibitors. Oncoimmunology. 2019;8:e1568812. doi: 10.1080/2162402X.2019.1568812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Masini C, Berselli A, Romagnani A, et al. Results of an Italian CORE-IMMUNO study: safety and clinical-related biomarkers as predictors of immunotherapy benefit in real-world treatment of various advanced tumors. J Clin Oncol. 2019;37:e14156. [Google Scholar]
  • 70.Kulkarni A, Kumar M, Pease DF, et al. Impact of antibiotics and proton pump inhibitors on clinical outcomes of immune checkpoint blockers in advanced non-small cell lung cancers and metastatic renal cell cancer. J Clin Oncol. 2019;37:e20520. [Google Scholar]

Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

RESOURCES