1,325
Views
0
CrossRef citations to date
0
Altmetric
Immunotherapy - Cancer

Gut microbiota and immunotherapy of renal cell carcinoma

ORCID Icon, , , , , , & show all
Article: 2268982 | Received 25 Jan 2023, Accepted 06 Oct 2023, Published online: 13 Nov 2023

ABSTRACT

The gut microbiome has recently been proposed as a key player in cancer development and progression. Several studies have reported that the composition of the gut microbiome plays a role in the response to immune checkpoint inhibitors (ICIs). The gut microbiome modulation has been investigated as a potential therapeutic strategy for cancer, mainly in patients undergoing therapy with ICIs. In particular, modulation through probiotics, FMT or other microbiome-related approaches have proven effective to improve the response to ICIs. In this review, we examine the role of the gut microbiome in enhancing clinical responses to ICIs in the treatment of renal cancer.

This article is part of the following collections:
Immunotherapy of Renal Cell Carcinoma

Introduction

The human gut microbiome is an ecological entity of about 100 trillion microorganisms that includes bacteria, yeast, viruses and parasites.Citation1–4 At birth, the human gut is poorly colonized by bacteria, but gets rapidly colonized by the microbiome of the mother and environment. The type of delivery, vaginal or cesarean, plays a major role in shaping the microbiome of the newborn.Citation4 The composition of the healthy gut microbiome is dominated about 90% from the phyla Firmicutes and Bacteroidetes.Citation5 The phylum of Firmicutes includes several genera involved in human health, the most common (up to 95%) are Lactobacillus, Bacillus, Enterococcus, Ruminococcus and Clostridium.Citation5 Moreover, it is important to report that the microbiome colonizes the whole gastrointestinal tract, and the composition is quantitatively and qualitatively different in various areas of itfor example, the gastric microbiome differs significantly from the intestinal microbiotaCitation6,Citation7

The gut microbiome plays key roles for human health, including a ‘shield effect’ from pathogenic microorganisms, maintaining the integrity of gut barrier, shaping and maturation of the immune system, regulation of human metabolism, nutrients and drug absorption.Citation8–14 The definition of the ‘healthy microbiome’ is one of the main challenges of researchers who deal with this topic. The alpha diversity of the gut microbiome, an ecological concept that refers to the number and distribution of different species, appears to be a reliable marker of health.Citation1 A progressive increase in diversity and stability of the gut microbiome can be clearly observed within the first years of life, reflecting the dynamics of the developing intestinal ecosystem in relation to several disturbances.Citation15 Moreover, early disruption of the gut microbiome has been associated with several chronic diseases in infants (such as allergy or asthma),Citation16 or even linked with several autoimmune and metabolic disorders that share a considerable alteration of the gut microbiome composition compared to matched control patientsCitation13 (as has been reported for celiac disease or diabetes).Citation17,Citation18

Furthermore, increasing evidence suggests that an imbalance of the gut microbiome homeostasis is closely related to the occurrence and development of cancer.Citation19 Interestingly, in recent years, new frontiers are emerging. The microbiome and – particular – the “polymorphic microbes” have been characterized as an emerging hallmark of cancer.Citation20,Citation21 More specifically, the gut microbiome composition may reflect direct carcinogenic activity, or the gut microbiome of patients may influence the host immune responses against cancer cells, mediate a protective effect for cancer development and progression, and – finally – modulate the response to anti-cancer therapies.Citation19 Additionally, the microbiome is involved in shaping the antitumor immune response.Citation20 Several studies have clarified the intrinsic relationship between microbiome and cancer,Citation22 and consequently the microbiome can be considered as a fundamental actor in cancer development and as modifier of therapeutic efficacy and safety.Citation23,Citation24

Due to recent research, the role of the gut microbiome in tumor immune regulation – and the interplay between microbiome and immune checkpoint proteins, such as programmed death 1 (PD-1) and its ligand 1 (PD-L1), and with cytotoxic T-lymphocyte antigen-4 (CTLA-4) – has been widely recognized (described in the next paragraph). This topic represents a new frontier, especially for those cancers for which immunotherapy is indicated as a first-line treatment, such as metastatic renal cell carcinoma (mRCC).Citation25 This review aims to describe the role of the microbiome in the development and progression of RCC and in modulating the efficacy of ICIs treatment.

Renal cell carcinoma: epidemiology, risk factors and treatment

Renal cell carcinoma (RCC) includes a heterogeneous group of cancers derived from renal tubular epithelial cells.Citation26 The major subtypes are papillary RCC, chromophobe RCC and clear cell RCC, which accounts for approximately 80% of all RCCs.Citation27–29 RCC is among the ten most common cancers worldwide, together with breast, pulmonary, colorectal cancer (CRC) and melanoma, with an estimated 79,000 new cases per year. Citation30–32 As reported by SEER database in the US, the median age of patients with RCC was 64 years with a near normal distribution,Citation30 it is more frequent in males (RCC accounts for 5% of all adult malignancies in male versus only 3% in female)Citation30,Citation31 The most common risk factors related to the development of RCC include excess body weight, cigarette smoking and hypertension.Citation32 RCC is also associated with other specific medical condition, such as chronic kidney disease, loss of glomerular filtration capacity requiring hemodialysis, kidney transplantation, acquired kidney cystic disease, previous RCC diagnosis and diabetes mellitus.Citation32

Approximately one-third of patients with RCC have metastatic disease at diagnosis, with a high mortality and an approximately 5-year relative survival rate of 14%.Citation30,Citation32–35,Citation36

Patients with localized RCC can be treated with surgical resection or loco-regional treatments (with or without adjuvant therapy (e.g. pembrolizumab)),Citation37 while patients with advanced stage or high risk of progression may benefit from first-line systemic therapy if surgery is not feasible, as reported by current guidelines,Citation27,Citation38 in particular, there was great hope in the improvement of the prognosis after the introduction of immunotherapy, which acts on precise molecular targets typical of this disease.

Indeed, clear cell RCC is also closely associated with a loss-of-function mutation in the von Hippel – Lindau (VHL) gene.Citation39 Loss of the VHL gene leads to the upregulation of hypoxia-inducible factor 2α (HIF-2α) and resultant overexpression of hypoxia-inducible genes, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor-β (PDGF-β) and transforming growth factor-α (TGF-α), which are involved in tumorigenesis and the progression of clear cell RCC.Citation40,Citation41

Therefore, targeting angiogenesis was hypothesized to be especially important in clear cell RCC treatment. Several clinical trials have shown that angiogenesis-targeted agents, such as sunitinib, pazopanib, bevacizumab, axitinib, cabozantinib, sorafenib and lenvatinib; furthermore, FDA recently approved Belzutifan for the treatment of VHL-linked RCC; in fact, clinical study has provided significant but not durable clinical benefits in clear cell RCC patients.Citation42–47

Moreover, RCC is influenced by immune modulation, as evidenced by its history of partial or complete response to high-dose interleukin-2.Citation48 For this reason, recently immune checkpoint inhibitors (ICIs) and combination strategies have changed the landscape of therapies for mRCC.Citation49–52 Immunotherapy targets are immune checkpoint proteins – such as PD-1, PD-L1 and CTLA-4 – that are often overexpressed by immune cells and/or tumor cells. The interaction between immune checkpoint proteins and these molecules leads to downregulation of antigen-specific T-cells mechanisms of programmed death (e.g. apoptosis), reduction in self-tolerance by suppression of T cell inflammatory activity and induced activation of T-cells that trigger an inflammatory response against cancer cells.Citation53

The relationship between gut microbiota and immunotherapy

The gut microbiome could be considered a ‘super organism,’ with a large amount of unique functions.Citation54 Among these, the gut microbiome plays a fundamental role in the development of the innate immune system.Citation55,Citation56

A considerable range of gut bacteria are involved in the gut immune response, especially Bacteroides fragilis, Clostridia, and Lactobacillus. Prevotella are able to induce Th17 cell-related cytokines, such as IL-6 and IL-23; consequently, the increase in Prevotella abundance is associated with the intensification of mucosal inflammation mediated by T helper type 17.Citation57 Lactobacillus and Bifidobacteria play a crucial role in shaping of proinflammatory Th17 lymphocytes, and they also induce tolerogenic dendritic cells and T regulatory lymphocytes (Tregs), involved in the regulation of immune response, whereas Clostridium cluster IV and XIVa, as well as Bacteroides, are involved in Th1 and Th17 immune responses.Citation58

Consequently, the microbiome can be considered as a regulator of the immune response. Therefore, in recent years, the scientific community has turned its attention toward the interaction between the microbiome and immunotherapy, with an interest in enhancing the clinical response to ICIs.Citation59 This close relationship was demonstrated first in animal models. In 2015, Vétizou et al.Citation60,Citation61 showed that antibiotic-treated or germ-free mice respond poorly to anti-CTLA4 therapy. They also demonstrated that the treatment of microbiota-depleted mice with either Bacteroides thetaiotaomicron or Bacteroides fragilis restores the anti-CTLA4 response by inducing dendritic cell maturation in tumors and a Th1 response.Citation60 Furthermore, a preclinical study reported that in mice with melanoma treated with anti-PD1 therapy, the concomitant administration of Bifidobacterium longum and Bifidobacterium breve resulted in increased response to therapy.Citation62

Furthermore, the association between the composition of the fecal microbiota and the response to anti-PD1 therapy is well established.Citation63 However, more limited data are available regarding the relationship between the microbiome and promoting anti-CTLA4 therapy clinical efficacy.Citation60

More specifically, a favorable outcome following anti PD 1 treatment was reported for patients with metastatic melanoma if their fecal microbiome demonstrated a higher abundance of key gut bacterial species belonging to Actinobacteria (Bifidobacteriaceae spp. and Coriobacteriaceae spp.) and Firmicutes (Ruminococcaceae spp. and Lachnospiraceae spp.).Citation64 In a recent prospective study of 69 patients, Derosaet al. showed that in mRCC patients treated with nivolumab, higher response rate was associated with an increase of Bacteroides salyersiae, Akkermansia muciniphila and Eubacterium siraeum, and a lower abundance of Clostridium clostridioforme and C. hathewayi in the fecal microbiota.Citation65

The role of Akkermansia muciniphila is controversial. Currently, available data suggest a possible bimodal effect of these bacterial species upon the anti-PD1 therapy response. First, certain studies reported that Akkermansia muciniphila may play an antitumor role in anti-PD1 therapy in patients with lung cancer, RCC or hepatocellular carcinoma.Citation63,Citation66,Citation67 Conversely, Derosa et al. found that higher fecal abundance of Akkermansia muciniphila induced by antibiotics was associated with resistance to anti-PD1 therapy in patients with advanced NSCLC.Citation68 Finally, in a cohort study of patients with advanced melanoma, a higher microbiota abundance of Faecalibacterium prausnitzii, Bacteroides thetaiotaomicron, Dorea formicogenerans and Holdemania filiformis were associated with better response to nivolumab or pembrolizumab.Citation69

Overall, clinical studies have reported gut microbial signatures that are not always concordant, which still makes it difficult to understand the pathophysiological mechanisms associated with these. For this reason, the increased knowledge from metabolomic and functional studies may allow to better understand the correlation between microbiome and response to ICIs, which to date is supported by pioneering clinical studies described below.

Improving immunotherapy efficacy by the modulation of gut microbiota

Immunotherapy has revolutionized cancer treatment, proving highly effective in epithelial cancers (e.g. melanoma). Unfortunately, however, despite an initial response, some patients develop resistance to anti-cancer treatment, and an ever-growing body of evidence suggests that the microbiota may play a role in this mechanism of resistance.Citation70

In consideration of this hypothesis, approaches based on the modulation of the microbiota have been tested with the aim of improving the efficacy of immunotherapy, especially through antibiotics, probiotics, diet or fecal microbiota transplantation.Citation71,Citation72

Antibiotics

In recent years, a growing body of data has been reported regarding the role of antibiotics in modulating the response to immunotherapy. Unfortunately, infections are a recurring problem in cancer patients, and therefore antibiotic therapy often overlaps with anti-cancer therapies, inducing a modulation on the efficacy of immunotherapy. In fact, a pioneering study suggests that in patients with solid tumors undergoing immunotherapy, antibiotic therapy is associated with worse response to treatment, especially if administered before immunotherapy.Citation61

To extend upon this observation, a recent study conducted on a cohort of mRCC and NSCLC patients reported that broad-spectrum β-lactam or quinolones were the most common antibiotics. Use of these antibiotics was associated with worsening of progression-free survival (HR 3.1, CI 95%, 1.4–6.9, P < .01) and the percentage of patients with primary progressive disease (75% versus 22%, P < .01).Citation73 Interestingly, another study reported that antibiotics had a greater effect in reducing progression-free survival in the RCC cohort (HR 2.13 95% CI 1.54–2.93), compared to both NSCLC (HR 1.64 95% CI 1.07–2.52) and melanoma (HR 1.54 95% CI 0.33–7.12).Citation74 This effect was statistically significant if antibiotic exposure occurred immediately before the initiation of immunotherapy (RCC HR 1.86 95% CI 1.16–2.98), suggesting a role during the induction. Other studies have shown how the use of antibiotics during therapy with ICIs (mainly in patients with RCC or melanoma) also reduces the overall survival (OS) (HR 1.88, 95% CI 1.59–2.22) and the response rate (OR 0.54, 95% CI 0.34–0.86).Citation75,Citation76 Moreover, antibiotics were statistically linked to a reduced progression-free survival (HR: 2.238, 95% CI: 1.284–3.900, P = .004) in a multicentre study of mRCC patients receiving nivolumab.Citation77 Similar results, with worst progression-free survival during antibiotic therapy (HR 1.96, 95% confidence interval [CI] 1.20–3.20, p = .007), were obtained in patients in therapy with mTOR inhibitors and VEGF target therapy during clinical trials.Citation78 Interestingly, in a recent study on patients with NSCLC, antibiotic therapy impacted the progression-free survival (HR 1.29, CI 95%, 1.04 to 1.59, p = .0192) and the overall survival (HR 1.42, CI 95%, 1.13 to 1.79, p = .0024) in patients treated with pembrolizumab, but not in patients treated with chemotherapy.Citation79 Furthermore, the same author demonstrated that the efficacy of first-line chemo-immunotherapy is not reduced by previous antibiotic exposure, in contrast with data reported for single ICIs.Citation80

Probiotics

Probiotics are defined as microorganisms that exert beneficial effect on host health.Citation63 This definition brings together a large number of products with characteristics that are not always well defined; therefore, the FDA has recently defined a new category of “live biotherapeutic products” (LBP), which have specific characteristics similar to other pharmacological products.Citation81 This regulation aims to promote specific indications for each probiotic strain as reported by clinical studies, with this approach necessary in the aim of moving toward personalized medicine. Indeed, considering the topic of this review, several studies have reported that probiotics have a potential role in enhancing clinical response to immunotherapy.Citation70 However, it is important to highlight that nonspecific probiotic supplementation is ineffective in improving immunotherapy outcomes. In fact, Spencer et al reported results from a series of melanoma patients undergoing ICIs who took commercial probiotics in clinical practice, and no significant difference was demonstrated in improvement of PFS or other outcomes in patients who took commercial probiotics or not.Citation82 These data confirm the importance of personalized medicine and the need to identify specific probiotic strains capable of modulating ICIs. For instance, a meta-analysis demonstrated superiority in terms of overall survival (HR = 0.50, 95% CI: 0.30–0.85, p = .01) and progression-free survival (HR = 0.51, 95% CI: 0.42–0.61, p < .01) in the groups of NSCLC patients who used probiotics containing Lactobacillus.Citation83 However, another study reported that probiotics reduced the progression of free survival (PFS) only in NSCLC (HR: 0.532, 95% CI: 0.354–0.798, p = .002), indeed it was not possible to demonstrate efficacy also in melanoma and mRCC due to the weakness of the available results.Citation84 Furthermore, Hahn AW et al, tried to verify the role of Bacteroides spp. in patients receiving first-line VEGF-TKIs in a retrospective cohort of 145 patients affected by mRCC.Citation85 finding that the administration of probiotics improved PFS in patients not receiving antibiotics (HR 0.92; 95% CI, 0.83–0.99; P = .04) as well as in patients receiving antibiotics (18 vs 8 months). Conversely, a subsequent randomized trial conducted on mRCC patients treated with VEGF-TKIs recorded no significant clinical benefit from eating Bifidobacterium-containing yogurt.Citation86 Recently, intriguing data have been published regarding the role of Clostridium butyricum CBM 588. In preclinical studies, CBM 588 is able to promote immune regulation and gut epithelial protection. Indeed, in a mice model, CBM 588 improved the intestinal barrier function and increased the abundance of Bifidobacterium, Lactobacillus and Lactococcus spp; moreover, this bacterium exerts an immunomodulatory role by favoring the expansion of IL-17A-producing γδT cells and IL-17A-producing CD4 cells in the colonic mucosa.Citation87 Therefore, considering pre-clinical results, this product has been studied in the modulation of the response to immunotherapy. A randomized prospective study was conducted on mRCC receiving immunotherapies with or without live Clostridium butyricum CBM588.Citation88 In particular, the administration of this bacterium was associated with a statistically significant increasing of PFS (12.7 months versus 2.5 months, HR 0.15, 95% confidence interval 0.05–0.47, P = .001) and improvement of response rate (58% versus 20%, P = 0.06); although, there were not reported significant differences in Bifidobacterium spp. or Shannon index related to the administration of this product. Further clinical studies are ongoing; in particular, interesting results are expected from a clinical study that will evaluate the efficacy of CBM588 in favoring the response to combined therapy approved by FDA with ICI and TKI (nivolumab and cabozantinib) in mRCC.Citation89 Finally, promising data concerning MRx0518 (that is, a gut microbiome-derived oral LBP consisting of a single strain of Enterococcus gallinarum), have recently been published; this bacterium may be able to modulate the immune response in treatment-naive cancer patients, translating to an improvement in the response to anti-cancer treatment.Citation90

Prebiotic

Prebiotics are defined as non-viable food components, mainly fibers or other non-digestible substances, that may stimulate the enrichment of particular gut microorganisms to prevent the colonization of pathogens and ensure effective immunological activity by modulating the release of inflammatory cytokines.Citation63 For instance, inulin was correlated with a higher presence of Bifidobacterium and Faecalibacterium that are able to enhance the activity of PD1/PD-L1 blockade therapy.Citation91 In in vitro models, oligosaccharides stimulate the production of arginine deiminase and L-asparaginase by Enterococcus durans, enzymes involved in cancer therapies.Citation92 This regulatory function might be used as an adjuvant strategy during immunotherapy, as suggested by an ongoing pilot study (NCT04552418).Citation93 A recent preclinical study demonstrated the impact of inulin gel in colorectal murine models (balb/c mice inoculated with colon carcinoma cells) treated with anti-PD1.Citation92 Inulin amplified the activity of immunotherapy in mice by remaining tumor-free, and it increased SCFA-producing bacteria that are involved in enhancing the efficacy of anti-PD1 therapy.Citation70,Citation92,Citation94

Dietary intervention and lifestyle

Diet and food ingestion are deeply linked to the gut microbiota.Citation94,Citation95 For example, high fat/low fiber diets are related to a low alpha diversity, reduction of Bifidobacterium spp., Bacteroides and F. prausnitzii and reduction of pro-inflammatory gut microbes.Citation96,Citation97 Inversely, high amounts of fiber are linked to an increased titer within the gut of Faecalibacterium prausnitzii, Akkermansia muciniphila and Bifidobacterium longum, which are involved in the metabolism of the intestinal epithelium and in the production or degradation of mucin, and are considered a key player in effectiveness of ICIs.Citation96 Many clinical trials (NCT03700437, NCT03950635 and NCT04909034) on dietary manipulation in patients with cancer who received immunotherapy are currently ongoing.Citation97–100 Published results by Spencer et al. showed that anti-PD-1 treatment for melanoma was five times more effective in patients with a higher fiber diet.Citation68,Citation101

Fecal microbiota transplantation

Fecal microbiota transplantation (FMT) consists in the transfer of feces from healthy screened donors into recipients with a disrupted microbiome, and it can be considered the most powerful among the therapeutic modulators of gut microbiome.

FMT is best established clinically as a therapy for recurrent Clostridioides difficile infection, where it achieves nearly 90% efficacy cure rates.Citation102,Citation103

More recently, in recognition of the fundamental role of gut microbiota in modulating the immune system, FMT has been proposed as a valid option to improve the clinical outcome of ICIs.Citation104 Clinical trials have proved that the microbiota could be modified with FMT to improve anti-PD1 therapy effectiveness in patients with melanoma. In the first one, ten patients with melanoma treated with anti-PD1 received repeated cycles of FMT from two different donors (the first by colonoscopy followed by capsule administration). Interestingly, one patient developed complete response, and two partial responses were also observed.Citation105 Moreover, responders to FMT also demonstrated an increase in inflammatory and antigen presentation signatures, associated with the shift toward an immune tumor rejection signature. In the second trial, 15 patients with melanoma with primary unresponsiveness to anti-PD1 therapy received a single FMT by colonoscopy. One patient had a complete response, two had a partial response, and three remained stable for more than 18 months. After FMT, the gut microbiota composition of patients with partial or complete clinical response appeared more similar than donor one; in these patients, there was an increased abundance of favorable taxa, such as Lachnospiraceae, Ruminococcaceae and Bifidobacteriaceae; in contrast, unfavorable species, such as those from the genus Bacteroides, were decreased.Citation106 In both trials, patients affected by melanoma and durable responders to anti-PD1 therapy have been used as donors. Indeed, these two pioneering clinical studies have confirmed that FMT is able to modulate the composition of the gut microbiome and the immune response, and in particular, they also underlie the importance of engraftment in favoring the enhancement of the response to ICIs; it is necessary to continue on this path by identifying pre-conditioning factors and the ideal donor to favor engraftment.Citation107 Finally, a clinical trial was recently published to evaluate the efficacy of FMT from healthy donors in enhancing clinical response to first-line regimen with ICIs in patients with advanced melanoma. The study enrolled 20 patients, with a partial response observed in 13/20 (65%) of the patients, with response of four of these 13 responders being complete. Longitudinal microbiome profiling revealed that microbiome similarity toward the pre-morbid composition was maintained over time only in responders.Citation108

To date, there are still no evident results regarding the efficacy of immunotherapy in mRCC; however, a pioneering study demonstrated the efficacy of FMT in reducing TKI-associated diarrhea through microbiome modulation and reduction of the mucosal inflammatory response.Citation109 Colitis associated with immunotherapy is a well-known complication, and as such therapeutic options to reduce the severity of symptoms can improve adherence to therapy and indirectly favor the clinical response.

Furthermore, FMT efficacy is also been explored in mRCC, CRC and in other solid cancers in several ongoing trials (NCT05273255, NCT05279677 and NCT04758507), but no results have been published yet.Citation110–112 Despite the reported promising results, more randomized clinical trials (RCTs) are advocated to establish the role of FMT in improving the efficacy of cancer immunotherapy, summarized in .

Table 1. Ongoing clinical trials investigating FMT and immunotherapy for renal cancer.

Final remarks and future perspectives

More than 100 drugs for the treatment of cancer have been approved by the FDA in the last two decades, yet the median survival has increased by a few months.Citation113 For this reason, the modulation of the microbiome associated with anti-cancer treatments could represent a pioneering strategy capable of improving the efficacy of these new drugs. Indeed, as previously described, much data have been published regarding the relationship between the microbiome and immune system. Similarly, immunotherapy is proving to be very effective, in particular, in solid tumors such as renal cancer. For this reason, the modulation of the microbiome and the development of immunotherapy protocols could represent a winning synergy. In particular, the demonstration that alteration of microbiome with antibiotics could play a role in reducing the efficacy of ICIs has encouraged the development of protocols based on microbiota modulation to improve the clinical response. In fact, several clinical trials have reported the efficacy of probiotics or prebiotics to improve the clinical outcome; in particular, considering probiotics, the results from studies on Clostridium butyricum CBM588 appear promising and worthy of further investigations. Nevertheless, the most promising results have emerged from preliminary studies on FMT, especially considering the deep impact upon gut microbiome modulation with this technique. Although the results are promisingto date, the evidence is still weak due to the limited number of patients enrolled in the studies and the heterogeneity of the protocols.

In conclusion, further preclinical and clinical studies to implement and standardize gut microbiome modulation in immunotherapy are needed. In particular, it will be fundamental to better understand the concept of engraftment and to learn how to predict the risk of complications or benefit from the administration of some consortia of bacteria compared to others in individuals undergoing immunotherapy. In particular, a greater knowledge of the relationship between microbiome and the immune system will favor the implementation of microbiome modulation in cancer treatment or – more specifically – in immunotherapy.

Abbreviations

CRC=

Colon Rectal Cancer

CTLA-4=

Cytotoxic T Lymphocyte Antigen-4

FMT=

Fecal Microbiota Transplantation

NSCLC=

Non-Small Cell Lung Cancer

ICI=

Immune Checkpoint Inhibitor

PD1=

Programmed Death 1

PD-L1=

Programmed Death – Ligand 1

PDGF-β=

PLatelet-Derived Growth Factor-β

PFS=

Progression Free Survival

RCC=

Renal Cell Carcinoma

mRCC=

metastaticRCC

SEER=

Surveillance, Epidemiology and End Results

TGF-α=

Transforming Growth Factor-α

Th1=

T Helper 1 Lymphocyte

Th17=

T Helper 17 Lymphocyte

Treg=

T Regulatory Lymphocyte

TKI=

Tyrosine Kinase Inhibitor

VEGF=

Vascular Endothelial Growth Factor

VHL=

Von Hippel-Lindau

Disclosure statement

Declaration of competing interest: B.H.M. reports consultancy fees from Finch Therapeutics Group, Ferring Pharmaceuticals and Summit Therapeutics, outside of the submitted work. G.I. has received personal fees for acting as speaker for Biocodex, Danone, Sofar, Malesci, Metagenics and Tillotts Pharma and for acting as consultant and/or advisor for Ferring Therapeutics, Giuliani, Malesci and Tillotts Pharma. A.G. reports personal fees for consultancy from Eisai Srl, 3PSolutions, Real Time Meeting, Fondazione Istituto Danone, Sinergie Srl, Board MRGE and Sanofi SpA personal fees for acting as a speaker for Takeda SpA, AbbVie and Sandoz SpA and personal fees for acting on advisory boards for VSL3 and Eisai. G.C. has received personal fees for acting as an advisor for Ferring Therapeutics.  All other authors have no conflicts of interest to disclose.

Additional information

Funding

S.B., S.P., L.E.D.V., A.S., G.I., A.G., G.C., acknowledged for the support Ministero della Salute italiano, Ricerca Corrente 2023 and Fondazione Roma. BHM is the rcipient of an NIHR Academic Clinical Lectureship (CL -2019-21-002). The division of digestive at imperial college London receives financial and infrastructure sopport from the NIHR Imperial Biomedical Research Centre (BRC) based at Imperial College Healthcare NHS Trust and Imperial College London.

References

  • Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489(7415):220. doi:10.1038/NATURE11550.
  • Ianiro G, Bruno G, Lopetuso L, Beghella F, Laterza L, D’Aversa F, Gigante G, Cammarota G, Gasbarrini A. Role of yeasts in healthy and impaired gut microbiota: the gut mycome. Curr Pharm Des. 2014;20(28):4565–10. doi:10.2174/13816128113196660723.
  • Ianiro G, Iorio A, Porcari S, Masucci L, Sanguinetti M, Perno CF, Gasbarrini A, Putignani L, Cammarota G. How the gut parasitome affects human health. Therap Adv Gastroenterol. 2022;15:175628482210915. doi:https://doi.org/10.1177/17562848221091524.
  • De Siena M, Laterza L, Matteo MV, Mignini I, Schepis T, Rizzatti G, Ianiro G, Rinninella E, Cintoni M, Gasbarrini A, et al. Gut and reproductive tract microbiota adaptation during pregnancy: new insights for pregnancy-related complications and therapy. Microorganisms. 2021;9(3):1–15. doi:10.3390/MICROORGANISMS9030473.
  • Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto J-M, et al. Enterotypes of the human gut microbiome. Nature. 2011;473(7346):174. doi:10.1038/NATURE09944.
  • Ianiro G, Molina-Infante J, Gasbarrini A. Gastric microbiota. Helicobacter. 2015;20(Suppl 1):68–71. doi:10.1111/HEL.12260.
  • Lopetuso LR, Severgnini M, Pecere S, Ponziani FR, Boskoski I, Larghi A, Quaranta G, Masucci L, Ianiro G, Camboni T, et al. Esophageal microbiome signature in patients with Barrett’s esophagus and esophageal adenocarcinoma. PloS One. 2020;15(5):e0231789. doi:10.1371/JOURNAL.PONE.0231789.
  • Viggiano D, Ianiro G, Vanella G, Bibbò S, Bruno G, Simeone G, Mele G. Gut barrier in health and disease: focus on childhood. Eur Rev Med Pharmacol Sci. 2015;19:1077–85.
  • Cardinale V, Capurso G, Ianiro G, Gasbarrini A, Arcidiacono PG, Alvaro D. Intestinal permeability changes with bacterial translocation as key events modulating systemic host immune response to SARS-CoV-2: a working hypothesis. Digestive Liver Dis. 2020;52(12):1383. doi:10.1016/J.DLD.2020.09.009.
  • Cianci R, Franza L, Schinzari G, Rossi E, Ianiro G, Tortora G, Gasbarrini A, Gambassi G, Cammarota G. The interplay between immunity and microbiota at intestinal immunological niche: the case of cancer. Int J Mol Sci. 2019;20(3):501. doi:https://doi.org/10.3390/IJMS20030501.
  • Bibbò S, Lopetuso LR, Ianiro G, Di Rienzo T, Gasbarrini A, Cammarota G. Role of microbiota and innate immunity in recurrent Clostridium difficile infection. J Immunol Res. 2014;2014:1–8. doi:https://doi.org/10.1155/2014/462740.
  • D’Aversa F, Tortora A, Ianiro G, Ponziani FR, Annicchiarico BE, Gasbarrini A. Gut microbiota and metabolic syndrome. Intern Emerg Med. 2013;8(Suppl 1):11–15. doi:10.1007/S11739-013-0916-Z.
  • Valdes AM, Walter J, Segal E, Spector TD. Role of the gut microbiota in nutrition and health. BMJ. 2018;361:36–44. doi:10.1136/BMJ.K2179.
  • Ianiro G, Mangiola F, Di Rienzo TA, Bibbò S, Franceschi F, Greco AV, Gasbarrini A. Levothyroxine absorption in health and disease, and new therapeutic perspectives. Eur Rev Med Pharmacol Sci. 2014;18:451–6.
  • Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, Angenent LT, Ley RE. Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4578–85. doi:https://doi.org/10.1073/PNAS.1000081107.
  • Jeong S. Factors influencing development of the infant microbiota: from prenatal period to early infancy. Clin Exp Pediatr. 2022;65(9):438. doi:10.3345/CEP.2021.00955.
  • Bibbò S, Abbondio M, Sau R, Tanca A, Pira G, Errigo A, Manetti R, Pes GM, Dore MP, Uzzau S, et al. Fecal microbiota signatures in celiac disease patients with poly-autoimmunity. Front Cell Infect Microbiol. 2020;10. doi:10.3389/FCIMB.2020.00349.
  • Bibbò S, Dore MP, Pes GM, Delitala G, Delitala AP. Is there a role for gut microbiota in type 1 diabetes pathogenesis? Ann Med. 2017;49(1):11–22. doi:10.1080/07853890.2016.1222449.
  • Christovich A, Luo XM. Gut microbiota, Leaky gut, and autoimmune diseases. Front Immunol. 2022;13. doi:10.3389/FIMMU.2022.946248.
  • Ali A, Ara A, Kashyap MK. Gut microbiota: role and association with tumorigenesis in different malignancies. Mol Biol Rep. 2022;49(8):8087–107. doi:10.1007/S11033-022-07357-6.
  • Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31–46. doi:10.1158/2159-8290.CD-21-1059.
  • Fulbright LE, Ellermann M, Arthur JC. The microbiome and the hallmarks of cancer. PLoS Pathog. 2017;13(9):e1006480. doi:https://doi.org/10.1371/JOURNAL.PPAT.1006480.
  • Lu Y, Yuan X, Wang M, He Z, Li H, Wang J, Li Q. Gut microbiota influence immunotherapy responses: mechanisms and therapeutic strategies. J Hematol Oncol. 2022;15(1). doi:10.1186/S13045-022-01273-9.
  • Eble JN, Sauter G, Epstein J, Sesterhenn I, editors. Pathology and genetics of tumours of the urinary system and male genital organs. Vol. 7. 3rd ed. WHO Classifications of Tumors IARC; 2004.
  • Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, Grünwald V, Gillessen S, Horwich A. Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2019;30(5):706–20. doi:10.1093/ANNONC/MDZ056.
  • Linehan WM, Spellman PT, Ricketts CJ, Creighton CJ, Fei SS, Davis C, Wheeler DA, Murray BA, Schmidt L, et al. Comprehensive molecular characterization of papillary renal-cell carcinoma. N Engl J Med. 2016;374(2):135–45. doi:10.1056/NEJMOA1505917.
  • Creighton CJ, Morgan M, Gunaratne PH, Wheeler DA, Gibbs RA, Robertson AG, Chu A, Beroukhim R., Cibulskis K., Signoretti S., et al. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499(7456):43–9. doi:10.1038/NATURE12222.
  • Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33. doi:10.3322/CAAC.21708.
  • Fay AP, McKay RR, Lin X, Simantov R, Choueiri TK. Impact of geographic regions on overall survival in patients with metastatic renal cell carcinoma: results from an International clinical trials database. J Glob Oncol. 2018;4(4):1–14. doi:10.1200/JGO.17.00119.
  • Key Statistics About Kidney Cancer. n.d. [accessed 2023 January 18]. https://www.cancer.org/cancer/kidney-cancer/about/key-statistics.html.
  • Zhuang TZ, Case K, Olsen TA, Brown JT, Carthon BC, Kucuk O, Goldman J, Harris W, Bilen MA, Nazha B, et al. Metastatic clear-cell renal cell carcinoma in the era of immune checkpoint inhibitors: therapies and ongoing trials. Cancers Basel. 2022;14(12):2867. doi:https://doi.org/10.3390/CANCERS14122867.
  • Padala SA, Barsouk A, Thandra KC, Saginala K, Mohammed A, Vakiti A, Rawla P, Barsouk A. Epidemiology of renal cell carcinoma. World J Oncol. 2020;11(3):79–87. doi:10.14740/WJON1279.
  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30. doi:10.3322/CAAC.21332.
  • Lipworth L, Tarone RE, Lund L, McLaughlin JK. Epidemiologic characteristics and risk factors for renal cell cancer. Clin Epidemiol. 2009;1:33. doi:10.2147/CLEP.S4759.
  • Choueiri TK, Tomczak P, Park SH, Venugopal B, Ferguson T, Chang Y-H, Hajek J, Symeonides SN, Lee JL, Sarwar N, et al. Adjuvant pembrolizumab after nephrectomy in renal-cell carcinoma. N Engl J Med. 2021;385(8):683–94. doi:https://doi.org/10.1056/NEJMoa2106391.
  • Powles T. Recent eUpdate to the ESMO clinical practice guidelines on renal cell carcinoma on cabozantinib and nivolumab for first-line clear cell renal cancer: renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021;32(3):422–3. doi:10.1016/J.ANNONC.2020.11.016.
  • Singer EA, Rumble RB, Van Veldhuizen PJ. Management of metastatic clear cell renal cell carcinoma: ASCO guideline Q&A. JCO Oncol Pract. 2023;19(3):127–31. doi:10.1200/OP.22.00660.
  • Choi WS, Boland J, Lin J. Hypoxia-inducible factor-2α as a novel target in renal cell carcinoma. J Kidney Cancer VHL. 2021;8(2):1–7. doi:10.15586/jkcvhl.v8i2.170.
  • Rini BI, Campbell SC, Escudier B. Renal cell carcinoma. Lancet. 2009;373(9669):1119–32. doi:10.1016/S0140-6736(09)60229-4.
  • Motzer RJ, Hutson TE, Glen H, Michaelson MD, Molina A, Eisen T, Jassem J, Zolnierek J, Maroto JP, Mellado B, et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial. Lancet Oncol. 2015;16(15):1473–82. doi:10.1016/S1470-2045(15)00290-9.
  • Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28(6):1061–8. doi:https://doi.org/10.1200/JCO.2009.23.9764.
  • Escudier B, Bellmunt J, Négrier S, Bajetta E, Melichar B, Bracarda S, Ravaud A, Golding S, Jethwa S, Sneller V, et al. Phase III trial of bevacizumab plus interferon alfa-2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. J Clin Oncol. 2010;28(13):2144–50. doi:10.1200/JCO.2009.26.7849.
  • Choueiri TK, Halabi S, Sanford BL, Hahn O, Michaelson MD, Walsh MK, Feldman DR, Olencki T, Picus J, Small EJ, et al. Cabozantinib versus sunitinib as initial targeted therapy for patients with metastatic renal cell carcinoma of poor or intermediate risk: the alliance A031203 CABOSUN trial. J Clin Oncol. 2017;35(6):591–7. doi:10.1200/JCO.2016.70.7398.
  • Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, Michaelson MD, Gorbunova VA, Gore ME, Rusakov IG, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet. 2011;378(9807):1931–9. doi:10.1016/S0140-6736(11)61613-9.
  • Burris HA. Overcoming acquired resistance to anticancer therapy: focus on the PI3K/AKT/mTOR pathway. Cancer Chemother Pharmacol. 2013;71(4):829–42. doi:10.1007/s00280-012-2043-3.
  • Rosenberg SA, Lotze MT, Muul LM, Chang AE, Avis FP, Leitman S, Linehan WM, Robertson CN, Lee RE, Rubin JT, et al. A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin-2 alone. N Engl J Med. 1987;316(15):889–97. doi:10.1056/NEJM198704093161501.
  • Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373(19):1803–13. doi:10.1056/NEJMOA1510665.
  • Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–90. doi:10.1056/NEJMOA1712126.
  • Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulières D, Melichar B, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1116–27. doi:10.1056/NEJMoa1816714.
  • Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, Kollmannsberger C, Negrier S, Uemura M, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1103–15. doi:10.1056/NEJMOA1816047.
  • Liu B, Zhang Y, Wang D, Hu X, Zhang Z. Single-cell meta-analyses reveal responses of tumor-reactive CXCL13+ T cells to immune-checkpoint blockade. Nat Cancer. 2022;3(9):1123–36. doi:10.1038/S43018-022-00433-7.
  • Kramer P, Bressan P. Humans as Superorganisms: how microbes, viruses, imprinted genes, and other selfish entities shape our behavior. Perspect Psychol Sci. 2015;10(4):464–81. doi:10.1177/1745691615583131.
  • Wu HJ, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes. 2012;3(1):4. doi:10.4161/GMIC.19320.
  • Riazi-Rad F, Behrouzi A, Mazaheri H, Katebi A, Ajdary S. Impact of gut microbiota on immune system. Acta Microbiol Immunol Hung. 2021;68(3):135–44. doi:10.1556/030.2021.01532.
  • Larsen JM. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology. 2017;151(4):363–74. doi:10.1111/IMM.12760.
  • Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–5. doi:10.1038/NATURE12726.
  • Shahbaz A, Mahmood T, Javed MU, Abbasi BH. Current advances in microbial-based cancer therapies. Med Oncol. 2023;40(7):207. doi:10.1007/S12032-023-02074-X.
  • Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CPM, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Sci. 2015;350(6264):1079–84. doi:10.1126/SCIENCE.AAD1329.
  • Pinato DJ, Howlett S, Ottaviani D, Urus H, Patel A, Mineo T, Brock C, Power D, Hatcher O, Falconer A, et al. Association of prior antibiotic treatment with survival and response to immune checkpoint Inhibitor therapy in patients with cancer. JAMA Oncol. 2019;5(12):1774–8. doi:10.1001/JAMAONCOL.2019.2785.
  • Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Man Lei Y, Jabri B, Alegre M-L, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science. 2015;350(6264):1084–9. doi:https://doi.org/10.1126/SCIENCE.AAC4255.
  • Zhou Y, Liu Z, Chen T. Gut microbiota: a promising milestone in enhancing the efficacy of PD1/PD-L1 blockade therapy. Front Oncol. 2022;12:476. doi:10.3389/fonc.2022.847350.
  • Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre M-L, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8. doi:10.1126/SCIENCE.AAO3290.
  • Derosa L, Routy B, Fidelle M, Iebba V, Alla L, Pasolli E, Segata N, Desnoyer A, Pietrantonio F, Ferrere G, et al. Gut bacteria composition drives primary resistance to cancer immunotherapy in renal cell carcinoma patients. Eur Urol. 2020;78(2):195–206. doi:https://doi.org/10.1016/J.EURURO.2020.04.044.
  • Zheng Y, Wang T, Tu X, Huang Y, Zhang H, Tan D, Jiang W, Cai S, Zhao P, Song R, et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J Immunother Cancer. 2019;7(1). doi:https://doi.org/10.1186/S40425-019-0650-9.
  • He D, Li X, An R, Wang L, Wang Y, Zheng S, Chen X, Wang X. Response to PD-1-Based immunotherapy for non-small cell lung cancer altered by gut microbiota. Oncol Ther. 2021;9(2):647–57. doi:10.1007/S40487-021-00171-3.
  • Derosa L, Routy B, Thomas AM, Iebba V, Zalcman G, Friard S, Mazieres J, Audigier-Valette C, Moro-Sibilot D, Goldwasser F, et al. Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat Med. 2022;28(2):315–24. doi:10.1038/S41591-021-01655-5.
  • Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, Koh AY. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia. 2017;19(10):848–55. doi:10.1016/J.NEO.2017.08.004.
  • Li X, Zhang S, Guo G, Han J, Yu J. Gut microbiome in modulating immune checkpoint inhibitors. EBioMedicine. 2022;82:104163. doi:https://doi.org/10.1016/J.EBIOM.2022.104163.
  • Cammarota G, Ianiro G, Bibbò S, Gasbarrini A. Gut microbiota modulation: probiotics, antibiotics or fecal microbiota transplantation? Intern Emerg Med. 2014;9(4):365–73. doi:10.1007/S11739-014-1069-4.
  • Bibbò S, Ianiro G, Giambò F, Settanni CR, Cammarota G, Gasbarrini A. Role of gut microbiome on immunotherapy efficacy in melanoma. Hum Vaccin Immunother. 2022;18(3). doi:10.1080/21645515.2021.1926759.
  • Derosa L, Hellmann MD, Spaziano M, Halpenny D, Fidelle M, Rizvi H, Long N, Plodkowski AJ, Arbour KC, Chaft JE, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann Oncol. 2018;29(6):1437–44. doi:10.1093/ANNONC/MDY103.
  • 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 Immunol Immunother. 2020;69(3):343–54. doi:10.1007/S00262-019-02453-2.
  • Tsikala-Vafea M, Belani N, Vieira K, Khan H, Farmakiotis D. Use of antibiotics is associated with worse clinical outcomes in patients with cancer treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Int J Infect Dis. 2021;106:142–54. doi:10.1016/J.IJID.2021.03.063.
  • Wu Q, Liu J, Wu S, Xie X. The impact of antibiotics on efficacy of immune checkpoint inhibitors in malignancies: a study based on 44 cohorts. Int Immunopharmacol. 2021;92. doi:10.1016/J.INTIMP.2020.107303.
  • Guven DC, Acar R, Yekeduz E, Bilgetekin I, Baytemur NK, Erol C, Ceylan F, Sendur MA, Demirci U, Urun Y, et al. The association between antibiotic use and survival in renal cell carcinoma patients treated with immunotherapy: a multi-center study. Curr Probl Cancer. 2021;45(6):100760. doi:https://doi.org/10.1016/J.CURRPROBLCANCER.2021.100760.
  • Lalani AKA, Xie W, Braun DA, Kaymakcalan M, Bossé D, Steinharter JA, Martini DJ, Simantov R, Lin X, Wei XX, et al. Effect of antibiotic use on outcomes with systemic therapies in metastatic renal cell carcinoma. Eur Urol Oncol. 2019;3(3):372–81. doi:https://doi.org/10.1016/J.EUO.2019.09.001.
  • Cortellini A, Di Maio M, Nigro O, Leonetti A, Cortinovis DL, Aerts JG, Guaitoli G, Barbieri F, Giusti R, Ferrara MG, et al. Differential influence of antibiotic therapy and other medications on oncological outcomes of patients with non-small cell lung cancer treated with first-line pembrolizumab versus cytotoxic chemotherapy. J Immunother Cancer. 2021;9(4):e002421. doi:https://doi.org/10.1136/JITC-2021-002421.
  • Cortellini A, Ricciuti B, Facchinetti F, Alessi JVM, Venkatraman D, Dall’Olio FG, Cravero P, Vaz VR, Ottaviani D, Majem M, et al. Antibiotic-exposed patients with non-small-cell lung cancer preserve efficacy outcomes following first-line chemo-immunotherapy. Ann Oncol. 2021;32(11):1391–9. doi:10.1016/J.ANNONC.2021.08.1744.
  • Cordaillat-Simmons M, Rouanet A, Pot B. Live biotherapeutic products: the importance of a defined regulatory framework. Exp Mol Med. 2020;52(9):1397–406. doi:10.1038/S12276-020-0437-6.
  • Spencer CN, McQuade JL, Gopalakrishnan V, McCulloch JA, Vetizou M, Cogdill AP, Khan MAW, Zhang X, White MG, Peterson CB, et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Sci. 2021;374(6575):1632–40. doi:10.1126/SCIENCE.AAZ7015.
  • Wan L, Wu C, Wu Q, Luo S, Liu J, Xie X. Impact of probiotics use on clinical outcomes of immune checkpoint inhibitors therapy in cancer patients. Cancer Med. 2022;12:1841–9. doi:https://doi.org/10.1002/CAM4.4994.
  • Zhang L, Jin Q, Chai D, Kuang T, Li C, Guan Y, Liu L, Wang W, Deng W. The correlation between probiotic use and outcomes of cancer patients treated with immune checkpoint inhibitors. Front Pharmacol. 2022;13. doi:10.3389/fphar.2022.937874.
  • Hahn AW, Froerer C, VanAlstine S, Rathi N, Bailey EB, Stenehjem DD, Agarwal N. Targeting Bacteroides in stool microbiome and response to treatment with first-line VEGF tyrosine kinase inhibitors in metastatic renal-cell carcinoma. Clin Genitourin Cancer. 2018;16(5):365–8. doi:10.1016/J.CLGC.2018.05.001.
  • Dizman N, Hsu JA, Bergerot PG, Gillece JD, Folkerts M, Reining L, Trent J, Highlander SK., Pal SK. Randomized trial assessing impact of probiotic supplementation on gut microbiome and clinical outcome from targeted therapy in metastatic renal cell carcinoma. Cancer Med. 2021;10(1):79–86. doi:10.1002/CAM4.3569.
  • Hagihara M, Kuroki Y, Ariyoshi T, Higashi S, Fukuda K, Yamashita R, Matsumoto A, Mori T, Mimura K, Yamaguchi N, et al. Clostridium butyricum modulates the microbiome to protect intestinal barrier function in mice with Antibiotic-Induced Dysbiosis. Science. 2020;23(1):100772. doi:https://doi.org/10.1016/J.ISCI.2019.100772.
  • Dizman N, Meza L, Bergerot P, Alcantara M, Dorff T, Lyou Y, Frankel P, Cui Y, Mira V, Llamas M, et al. Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial. Nat Med. 2022;28(4):704–12. doi:https://doi.org/10.1038/s41591-022-01694-6.
  • Meza LA, Malhotra J, Zengin ZB, Dizman N, Hsu J, Chawla NS, Chehrazi-Raffle A, Muddasani R, Govindarajan A, Castro DV, et al. A phase I trial to evaluate the biologic effect of CBM588 (Clostridium butyricum) in combination with cabozantinib plus nivolumab for patients with metastatic renal cell carcinoma (mRCC). 2022;40(16_suppl):TPS4606–TPS4606. doi:10.1200/JCO.2022.40.16_SUPPL.TPS4606.
  • Lythgoe M, Dama P, Frampton AE, Pickford E, Tookman L, Cunnea P, Fotopoulou C, Liu D, Clark J, Lozano-Kuehne J, et al. Immune modulation and the oral live biotherapeutic product, MRx0518, in treatment-naïve patients with cancer: updated safety data. J Clin Oncol. 2023;41(16_suppl):3145–3145. doi:https://doi.org/10.1200/JCO.2023.41.16_SUPPL.3145.
  • Saedisomeolia A, Wood LG, Garg ML, Gibson PG, Wark PAB. Anti-inflammatory effects of long-chain n -3 PUFA in rhinovirus-infected cultured airway epithelial cells. Br J Nutr. 2009;101(4):533–40. doi:10.1017/S0007114508025798.
  • Comerlato CB, Zhang X, Walker K, Brandelli A, Figeys D. Comparative proteomic analysis reveals metabolic variability of probiotic Enterococcus durans during aerobic and anaerobic cultivation. J Proteomics. 2020;220:103764. doi:https://doi.org/10.1016/J.JPROT.2020.103764.
  • Intestinal microbiome modification with resistant starch in patients treated with dual immune checkpoint inhibitors - full text view - ClinicalTrials.Gov. n.d [accessed 2022 December 11. https://clinicaltrials.gov/ct2/show/NCT04552418.
  • Han K, Nam J, Xu J, Sun X, Huang X, Animasahun O, Achreja A, Jeon JH, Pursley B, Kamada N, et al. Generation of systemic antitumour immunity via the in situ modulation of the gut microbiome by an orally administered inulin gel. Nat Biomed Eng. 2021;5(11):1377–88. doi:10.1038/S41551-021-00749-2.
  • Ley RE, Hamady M, Lozupone C, Turnbaugh PJ, Ramey RR, Bircher JS, Schlegel ML, Tucker TA, Schrenzel MD, Knight R, et al. Evolution of mammals and their gut microbes. Sci. 2008;320(5883):1647–51. doi:10.1126/SCIENCE.1155725.
  • Nava GM, Carbonero F, Ou J, Benefiel AC, O’Keefe SJ, Gaskins HR. Hydrogenotrophic microbiota distinguish native Africans from African and European Americans. Environ Microbiol Rep. 2012;4(3):307–15. doi:10.1111/J.1758-2229.2012.00334.X.
  • Devkota S, Wang Y, Musch MW, Leone V, Fehlner-Peach H, Nadimpalli A, Antonopoulos DA, Jabri B, Chang EB. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature. 2012;487(7405):104–8. doi:10.1038/NATURE11225.
  • Olivares-Hernández A, Figuero-Pérez L, González-Sarmiento R, León-Jiménez D, Cruz-Hernández JJ, Miramontes-González JP. The influence of diet and probiotics on the response of solid Tumours to immunotherapy: present and future perspectives. Appl Sci. 2021;11(18):8445. doi:10.3390/APP11188445.
  • Zhang X, Browman G, Siu W, Basen-Engquist KM, Hanash SM, Hoffman KL, Okhuysen PC, Scheet P, Petrosino JF, Kopetz S, et al. The BE GONE trial study protocol: a randomized crossover dietary intervention of dry beans targeting the gut microbiome of overweight and obese patients with a history of colorectal polyps or cancer. BMC Cancer. 2019;19(1):1–11. doi:https://doi.org/10.1186/s12885-019-6400-z.
  • Fasting-mimicking diet with chemo-immunotherapy in non-small cell lung cancer (NSCLC) - full text view - ClinicalTrials.Gov n.d. [accessed 2022 December 11]. https://clinicaltrials.gov/ct2/show/NCT03700437.
  • Safety and potential efficacy of MS-20 in combination with pembrolizumab for the treatment of NSCLC - Full text view - ClinicalTrials.Gov. n.d. [accessed 2022 December 11]. https://clinicaltrials.gov/ct2/show/NCT04909034.
  • Dietary intervention in patients with a history of melanoma - full text view - ClinicalTrials.Gov n.d. [accessed 2022 December 11]. https://clinicaltrials.gov/ct2/show/NCT03950635.
  • Spencer CN, Gopalakrishnan V, McQuade J, Andrews MC, Helmink B, Khan MAW, Sirmans E, Haydu L, Cogdill A, Burton E, et al. Abstract 2838: the gut microbiome (GM) and immunotherapy response are influenced by host lifestyle factors. Cancer Res. 2019;79(13_Supplement):2838–2838. doi:10.1158/1538-7445.AM2019-2838.
  • Ianiro G, Maida M, Burisch J, Simonelli C, Hold G, Ventimiglia M, Gasbarrini A, Cammarota G. Efficacy of different faecal microbiota transplantation protocols for Clostridium difficile infection: a systematic review and meta-analysis. United Eur Gastroent J. 2018;6(8):1232–44. doi:10.1177/2050640618780762.
  • Bibbò S, Settanni CR, Porcari S, Bocchino E, Ianiro G, Cammarota G, Gasbarrini A. Fecal microbiota transplantation: screening and selection to choose the optimal donor. J Clin Med. 2020;9(6):1–14. doi:10.3390/JCM9061757.
  • Lythgoe MP, Ghani R, Mullish BH, Marchesi JR, Krell J. The potential of fecal microbiota transplantation in oncology. Trends Microbiol. 2022;30(1):10–12. doi:10.1016/J.TIM.2021.10.003.
  • Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371(6529):602–9. doi:10.1126/SCIENCE.ABB5920.
  • Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin J-M, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, et al. Fecal microbiota transplant overcomes resistance to anti–PD-1 therapy in melanoma patients. Science. 2021;371(6529):595–602. doi:https://doi.org/10.1126/SCIENCE.ABF3363.
  • Elkrief A, Routy B. First clinical proof-of-concept that FMT can overcome resistance to ICIs. Nat Rev Clin Oncol. 2021;18(6):325–6. doi:https://doi.org/10.1038/s41571-021-00502-3.
  • Routy B, Lenehan JG, Miller WH, Jamal R, Messaoudene M, Daisley BA, Hes C, Al KF, Martinez-Gili L, Punčochář M, et al. Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: a phase I trial. Nat Med. 2023;29:2121–32. doi:https://doi.org/10.1038/s41591-023-02453-x.
  • Ianiro G, Rossi E, Thomas AM, Schinzari G, Masucci L, Quaranta G, Settanni CR, Lopetuso LR, Armanini F, Blanco-Miguez A, et al. Faecal microbiota transplantation for the treatment of diarrhoea induced by tyrosine-kinase inhibitors in patients with metastatic renal cell carcinoma. Nat Commun. 2020;11(1):1–6. doi:https://doi.org/10.1038/s41467-020-18127-y.
  • Porcari S, Ciccarese C, Pinto F, Quaranta G, De Giorgi S, Rondinella D, Settanni CR, Cortesi E, Roberto M, Primi F, et al. Fecal microbiota transplantation to improve efficacy of immune checkpoint inhibitors in renal cell carcinoma (TACITO trial). J Clin Oncol. 2022;40(6_suppl):TPS407–TPS407. doi:https://doi.org/10.1200/JCO.2022.40.6_SUPPL.TPS407.
  • Fecal microbiota transplantation in patients with malignancies not responding to immune checkpoint Inhibitor therapy - full text view - ClinicalTrials.Gov n.d. [accessed 2023 January 18]. https://clinicaltrials.gov/ct2/show/NCT05273255.
  • FMT combined with immune checkpoint Inhibitor and TKI in the treatment of CRC patients with advanced stage - full text view - ClinicalTrials.Gov n.d. [accessed 2023 January 18]. https://clinicaltrials.gov/ct2/show/NCT05279677.
  • Gloy V, Schmitt AM, Düblin P, Hirt J, Axfors C, Kuk H, Pereira TV, Locher C, Caquelin L, Walter‐Claudi M, et al. The evidence base of US food and Drug administration approvals of novel cancer therapies from 2000 to 2020. Int J Cancer. 2023;152(12):2474–84. doi:10.1002/IJC.34473.