Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Oct 6, 2024; 12(28): 6159-6164
Published online Oct 6, 2024. doi: 10.12998/wjcc.v12.i28.6159
Hidden army within: Harnessing the microbiome to improve cancer treatment outcomes
Ippokratis Messaritakis, Department of Microbiology, German Oncology Center & Yiannoukas Labs LTD, Bioiatriki Group, Limassol 4108, Cyprus
Georgios Vougiouklakis, Aris P Agouridis, Department of Internal Medicine, German Oncology Center, Limassol 4108, Cyprus
Asimina Koulouridi, Department of Oncology, German Oncology Center, Limassol 4108, Cyprus
Aris P Agouridis, School of Medicine, European University Cyprus, Nicosia 2404, Cyprus
Nikolaos Spernovasilis, Department of Infectious Diseases, German Oncology Center, Limassol 4108, Cyprus
ORCID number: Aris P Agouridis (0000-0002-9749-5075); Nikolaos Spernovasilis (0000-0002-6981-8535).
Author contributions: Messaritakis I, Vougiouklakis G, Koulouridi A, Agouridis AP, and Spernovasilis N contributed to this paper; Messaritakis I and Koulouridi A designed the overall concept and outline of the manuscript; Spernovasilis N contributed to the discussion and design of the manuscript; Messaritakis I and Vougiouklakis G contributed to the writing and review of literature; Koulouridi A, Agouridis AP, and Spernovasilis N contributed to editing the manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: Https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Nikolaos Spernovasilis, BSc, MD, MSc, PhD, Director, Department of Infectious Diseases, German Oncology Center, Nikis 1, Limassol 4108, Cyprus. nikspe@hotmail.com
Received: May 14, 2024
Revised: June 6, 2024
Accepted: June 24, 2024
Published online: October 6, 2024
Processing time: 90 Days and 10.6 Hours

Abstract

The gut microbiome has emerged as a critical player in cancer pathogenesis and treatment response. Dysbiosis, an imbalance in the gut microbial community, impacts tumor initiation, progression, and therapy outcomes. Specific bacterial species have been associated with either promoting or inhibiting tumor growth, offering potential targets for therapeutic intervention. The gut microbiome influences the efficacy and toxicity of conventional treatments and cutting-edge immunotherapies, highlighting its potential as a therapeutic target in cancer care. However, translating microbiome research into clinical practice requires addressing challenges such as standardizing methodologies, validating microbial biomarkers, and ensuring ethical considerations. Here, we provide a comprehensive overview of the gut microbiome's role in cancer highlighting the need for ongoing research, collaboration, and innovation to harness its full potential for improving patient outcomes in oncology. The current editorial aims to explore these insights and emphasizes the need for standardized methodologies, validation of microbial biomarkers, and interdisciplinary collaboration to translate microbiome research into clinical applications. Furthermore, it underscores ethical considerations and regulatory challenges surrounding the use of microbiome-based therapies. Together, this article advocates for ongoing research, collaboration, and innovation to realize the full potential of microbiome-guided oncology in improving patient care and outcomes.

Key Words: Gut microbiome, Cancer treatment, Immunotherapy, Microbiome-based interventions, Dysbiosis, Precision oncology

Core Tip: Understanding the intricate relationship between the gut microbiome and cancer treatment outcomes is pivotal in modern oncology. Dysbiosis, an imbalance in gut microbial communities, significantly influences tumor initiation, progression, and response to therapy. Harnessing the microbiome's potential offers novel avenues for personalized cancer care, including optimizing treatment strategies and enhancing immunotherapy efficacy. Multidisciplinary collaboration, standardized methodologies for microbiome profiling, and the development of microbial biomarkers are essential for translating microbiome research into clinical practice. Ethical considerations and regulatory oversight are paramount in ensuring equitable access to emerging microbiome-based interventions while promoting transparency and education among healthcare professionals and the public.



INTRODUCTION

Microbes appeared on Earth around 4 billion years ago, marking a critical point in the planet's history as it signifies the beginnings of life due to their role in oxygen production. It is well known that nowadays, microorganisms are ubiquitous, inhabiting diverse environments, including the human organism. In the continuing narrative of cancer treatment, a burgeoning field is emerging, shedding light on a potent ally inherent within our body: The gut microbiome. The article entitled "Bringing gut microbiota into the spotlight of clinical research and medical practice", authored by Davoutis et al[1] establishes a foundational comprehension of the crucial role the gut microbiota assumes in both clinical research and medical practice. The current editorial endeavors to further explore the insights provided by the article, elucidating how the gut microbiome possesses significant potential in reshaping cancer treatment outcomes and revolutionizing oncology practice.

GUT MICROBIOME: A HIDDEN PLAYER IN CANCER PATHOGENESIS

The symbiotic relationship between higher organisms and their commensal gut microbiota plays a crucial role in governing various aspects of human health, including but not limited to nutrition regulation, metabolic processes, homeostasis maintenance, developmental stages, and the functionality of both innate and adaptive immune responses[2-4]. Furthermore, this interaction exerts influence over inflammatory processes, albeit to a lesser degree, and contributes to the modulation of morphogenesis[4]. These multifaceted roles underscore the intricate interplay between the gut microbiome and human physiology.

Dysbiosis, an imbalance in the gut microbial community, has emerged as a hallmark of cancer pathogenesis, impacting tumor initiation, progression, and response to therapy[5]. Through intricate interactions with the host immune system and systemic physiology, the gut microbiome influences key hallmarks of cancer, including inflammation, immune evasion, and angiogenesis[6]. The article by Davoutis et al[1], underscores the significance of unraveling the complex interplay between the gut microbiome and cancer biology, shedding light on novel therapeutic avenues for combating malignancies.

Recent studies have highlighted the role of specific bacterial species in promoting or inhibiting tumor growth[7]. For instance, certain members of the Firmicutes phylum, such as Faecalibacterium and Clostridiales, have been associated with anti-tumorigenic effects, while others, like Fusobacterium spp. and Bacteroides fragilis, have been implicated in promoting tumor development[8,9].

MICROBIOME-MEDIATED EFFECTS ON CANCER THERAPY

One of the most compelling aspects highlighted in the article is the microbiome's influence on cancer therapy outcomes. By modulating the tumor microenvironment and systemic immune responses, the gut microbiome exerts profound effects on the efficacy and toxicity of conventional treatments such as chemotherapy and radiation therapy[7,10,11]. Moreover, emerging evidence suggests that the gut microbiome plays a pivotal role in shaping responses to cutting-edge immunotherapies, including immune checkpoint inhibitors and adoptive cell therapies[12]. Understanding the mechanisms underlying these microbiome-mediated effects is crucial for optimizing treatment strategies and personalizing cancer care.

Recent preclinical and clinical studies have provided compelling evidence supporting the role of the gut microbiome in modulating responses to immunotherapy. Routy et al[12], found that the gut microbiome composition predicts the response to PD-1 blockade in patients with advanced solid tumors, suggesting that microbiome-based biomarkers could guide patient selection and treatment strategies in the era of precision oncology. Similarly, Gopalakrishnan et al[13], demonstrated that the gut microbiome influences the efficacy of immune checkpoint inhibitors in melanoma patients, with specific bacterial taxa associated with enhanced anti-tumor immune responses and improved clinical outcomes.

It is noteworthy to mention that gut microbiome influences not only the efficacy of immunotherapy but also the efficacy and toxicity of other treatments, such as chemotherapy[14]. Recent research has elucidated how specific gut microbial communities can influence the effectiveness of chemotherapy drugs and mitigate their side effects[14,15]. Certain bacteria can metabolize chemotherapy agents, affecting their bioavailability and performance, while others can modulate inflammation and intestinal permeability, reducing the risk of gastrointestinal side effects[14]. More specifically, the elimination of intestinal bacteria through previous antibiotic therapy diminishes both myeloid and Th17 responses, subsequently lowering the effectiveness of oxaliplatin and cyclophosphamide[7,14]. Furthermore, the influence of microbiota on the efficacy of 5-fluorouracil (5-FU) may occur via the bacterial processing of vitamins and ribonucleotides, while the suppression of bacterial deoxyribonucleotide metabolism could potentially augment the effectiveness of 5-FU[16]. Additionally, a notable prevalence of Fusobacterium nucleatum (F. nucleatum) correlates with resistance to oxaliplatin and 5-FU. This phenomenon operates through a mechanistic pathway where F. nucleatum triggers the TLR4/MYD88-dependent pathway, thereby instigating innate immunity. This activation results in the depletion of microRNA segments, prompting a transition in cancer cell behavior from apoptosis to autophagy, consequently fostering the emergence of chemoresistance. Consequently, the targeting of F. nucleatum to diminish autophagy levels in colorectal cancer (CRC) cells holds promise for enhancing the chemosensitivity of CRC[11].

HARNESSING THE MICROBIOME TO ENHANCE IMMUNOTHERAPY

Immunotherapy has emerged as a game-changer in cancer treatment, offering durable responses and improved survival outcomes across a spectrum of malignancies. However, response rates to immunotherapy remain variable, underscoring the need to identify predictive biomarkers and strategies to enhance treatment efficacy[17]. Sivan et al[18] showed how the gut microbiome serves as a key determinant of immunotherapy response, with specific microbial signatures associated with favorable or adverse outcomes. The concept of modulating the gut microbiome as a therapeutic approach in cancer aligns with the increasing focus on microbiome-based interventions, including fecal microbiota transplantation, probiotics, selective antibiotics, or bacteriophages[19,20]. Furthermore, dietary habits, physical activity, and other lifestyle factors shape the composition and function of the gut microbiome, influencing an individual's susceptibility to cancer development and response to treatment[21,22]. These interventions aim to restore a healthy equilibrium in the gut microbiome. Leveraging microbiome-targeted interventions, such as fecal microbiota transplantation and microbial modulators, seems promising not only for enhancing the efficacy of immunotherapy but also on overcoming resistance mechanisms.

Recent preclinical studies have provided compelling evidence supporting the potential of microbiome-based interventions to enhance the efficacy of immunotherapy. For example, the administration of certain microbial metabolites, such as short chain fatty acids, has been shown to potentiate the anti-tumor effects of immune checkpoint blockade by modulating the activity of tumor-infiltrating immune cells and promoting the generation of memory T cells[18,23,24]. Moreover, researchers demonstrated that the efficacy of anti-PD-1 therapy in melanoma-bearing mice is dependent on the composition of the gut microbiome, with specific bacterial species, such as Bacteroidales and Burkholderiales, promoting anti-tumor immune responses and tumor regression[17].

TRANSLATING MICROBIOME RESEARCH INTO CLINICAL PRACTICE

Despite the growing body of evidence implicating the gut microbiome in cancer pathogenesis and treatment, several challenges hinder its translation into clinical practice. Standardization of methodologies for microbiome profiling, validation of microbial biomarkers, and integration of microbiome-targeted interventions into existing treatment paradigms represent critical areas of focus[25]. Moreover, the importance of multidisciplinary collaboration between oncologists, microbiologists, and computational biologists to harness the full potential of microbiome-based therapies and realize tangible benefits for cancer patients is also highlighted in the literature. Emerging research is focusing on the development of microbiome-based biomarkers for predicting cancer prognosis, treatment response, and therapeutic toxicity. Microbial signatures in blood, stool, or tumor tissue hold promise as non-invasive biomarkers for guiding treatment decisions and monitoring disease progression[12,26].

CONCLUSION

In navigating the intricate landscape of cancer treatment, the gut microbiome emerges as a beacon of hope, offering novel insights and therapeutic strategies to enhance patient outcomes. Through this path and by leveraging the microbiome's influence to tilt the scales in favor of the patient, we can unveil new avenues for personalized cancer therapy.

Ethical issues surrounding the use of microbiome-based interventions in cancer care, including patient consent, privacy concerns, and equitable access to emerging therapies, demand attention[27,28]. Regulatory hurdles in evaluating the safety and efficacy of microbiome-targeted interventions underscore the necessity of interdisciplinary collaboration and regulatory oversight to advance research. Expanding on these insights, it is imperative to stress the importance of further research to fully comprehend the complex interplay between the gut microbiome and cancer treatment outcomes. This entails investigating specific microbial signatures associated with treatment response and resistance, as well as elucidating the mechanisms through which the microbiome influences cancer progression and therapy efficacy. Additionally, efforts should be directed towards establishing standardized protocols for microbiome analysis and intervention implementation in clinical settings, ensuring reproducibility and consistency across studies. The article by Davoutis et al[1] serves as a catalyst for deeper exploration, and as the field of microbiome-guided oncology evolves, there is a pressing need for ongoing education and awareness among healthcare professionals and the public regarding the potential benefits and limitations of microbiome-based interventions in cancer care. This involves fostering dialogue around ethical considerations, promoting transparency in research practices, and dispelling misconceptions or unrealistic expectations surrounding microbiome therapies.

In summary, while the gut microbiome presents significant promise as a therapeutic target in cancer treatment, its integration into clinical practice demands careful consideration of ethical, regulatory, and scientific challenges. The article by Davoutis et al[1] lays the groundwork for further investigation in this domain, encouraging collaboration and innovation to unlock the full potential of microbiome-guided oncology in enhancing patient care and outcomes.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country of origin: Cyprus

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Chen K S-Editor: Liu H L-Editor: Filipodia P-Editor: Xu ZH

References
1.  Davoutis E, Gkiafi Z, Lykoudis PM. Bringing gut microbiota into the spotlight of clinical research and medical practice. World J Clin Cases. 2024;12:2293-2300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
2.  Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G, Quraishi MN, Kinross J, Smidt H, Tuohy KM, Thomas LV, Zoetendal EG, Hart A. The gut microbiota and host health: a new clinical frontier. Gut. 2016;65:330-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1341]  [Cited by in F6Publishing: 1401]  [Article Influence: 175.1]  [Reference Citation Analysis (0)]
3.  Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012;148:1258-1270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2236]  [Cited by in F6Publishing: 2368]  [Article Influence: 197.3]  [Reference Citation Analysis (0)]
4.  Belkaid Y, Harrison OJ. Homeostatic Immunity and the Microbiota. Immunity. 2017;46:562-576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 591]  [Cited by in F6Publishing: 701]  [Article Influence: 100.1]  [Reference Citation Analysis (0)]
5.  Arthur JC, Perez-Chanona E, Mühlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ, Abujamel T, Dogan B, Rogers AB, Rhodes JM, Stintzi A, Simpson KW, Hansen JJ, Keku TO, Fodor AA, Jobin C. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012;338:120-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1374]  [Cited by in F6Publishing: 1522]  [Article Influence: 126.8]  [Reference Citation Analysis (0)]
6.  Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell. 2018;33:570-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 677]  [Cited by in F6Publishing: 793]  [Article Influence: 132.2]  [Reference Citation Analysis (0)]
7.  Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, Dai RM, Kiu H, Cardone M, Naik S, Patri AK, Wang E, Marincola FM, Frank KM, Belkaid Y, Trinchieri G, Goldszmid RS. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342:967-970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1253]  [Cited by in F6Publishing: 1506]  [Article Influence: 136.9]  [Reference Citation Analysis (0)]
8.  Hibberd AA, Lyra A, Ouwehand AC, Rolny P, Lindegren H, Cedgård L, Wettergren Y. Intestinal microbiota is altered in patients with colon cancer and modified by probiotic intervention. BMJ Open Gastroenterol. 2017;4:e000145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 214]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
9.  Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Huang K, Guevara F, Nelson T, Chipashvili O, Hagan T, Walker M, Ramachandran A, Diosdado B, Serna G, Mulet N, Landolfi S, Ramon Y Cajal S, Fasani R, Aguirre AJ, Ng K, Élez E, Ogino S, Tabernero J, Fuchs CS, Hahn WC, Nuciforo P, Meyerson M. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. 2017;358:1443-1448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 649]  [Cited by in F6Publishing: 878]  [Article Influence: 125.4]  [Reference Citation Analysis (0)]
10.  Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, Vonderheide RH, Pittet MJ, Jain RK, Zou W, Howcroft TK, Woodhouse EC, Weinberg RA, Krummel MF. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24:541-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2019]  [Cited by in F6Publishing: 3119]  [Article Influence: 519.8]  [Reference Citation Analysis (0)]
11.  Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, Chen Y, Chen H, Hong J, Zou W, Fang JY. Fusobacterium nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy. Cell. 2017;170:548-563.e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 814]  [Cited by in F6Publishing: 1227]  [Article Influence: 175.3]  [Reference Citation Analysis (0)]
12.  Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, Fidelle M, Flament C, Poirier-Colame V, Opolon P, Klein C, Iribarren K, Mondragón L, Jacquelot N, Qu B, Ferrere G, Clémenson C, Mezquita L, Masip JR, Naltet C, Brosseau S, Kaderbhai C, Richard C, Rizvi H, Levenez F, Galleron N, Quinquis B, Pons N, Ryffel B, Minard-Colin V, Gonin P, Soria JC, Deutsch E, Loriot Y, Ghiringhelli F, Zalcman G, Goldwasser F, Escudier B, Hellmann MD, Eggermont A, Raoult D, Albiges L, Kroemer G, Zitvogel L. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2493]  [Cited by in F6Publishing: 3286]  [Article Influence: 469.4]  [Reference Citation Analysis (0)]
13.  Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, Cogdill AP, Zhao L, Hudgens CW, Hutchinson DS, Manzo T, Petaccia de Macedo M, Cotechini T, Kumar T, Chen WS, Reddy SM, Szczepaniak Sloane R, Galloway-Pena J, Jiang H, Chen PL, Shpall EJ, Rezvani K, Alousi AM, Chemaly RF, Shelburne S, Vence LM, Okhuysen PC, Jensen VB, Swennes AG, McAllister F, Marcelo Riquelme Sanchez E, Zhang Y, Le Chatelier E, Zitvogel L, Pons N, Austin-Breneman JL, Haydu LE, Burton EM, Gardner JM, Sirmans E, Hu J, Lazar AJ, Tsujikawa T, Diab A, Tawbi H, Glitza IC, Hwu WJ, Patel SP, Woodman SE, Amaria RN, Davies MA, Gershenwald JE, Hwu P, Lee JE, Zhang J, Coussens LM, Cooper ZA, Futreal PA, Daniel CR, Ajami NJ, Petrosino JF, Tetzlaff MT, Sharma P, Allison JP, Jenq RR, Wargo JA. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2999]  [Cited by in F6Publishing: 2823]  [Article Influence: 470.5]  [Reference Citation Analysis (0)]
14.  Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, Schlitzer A, Ginhoux F, Apetoh L, Chachaty E, Woerther PL, Eberl G, Bérard M, Ecobichon C, Clermont D, Bizet C, Gaboriau-Routhiau V, Cerf-Bensussan N, Opolon P, Yessaad N, Vivier E, Ryffel B, Elson CO, Doré J, Kroemer G, Lepage P, Boneca IG, Ghiringhelli F, Zitvogel L. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342:971-976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1147]  [Cited by in F6Publishing: 1391]  [Article Influence: 126.5]  [Reference Citation Analysis (0)]
15.  Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14:356-365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 451]  [Cited by in F6Publishing: 564]  [Article Influence: 80.6]  [Reference Citation Analysis (0)]
16.  Scott TA, Quintaneiro LM, Norvaisas P, Lui PP, Wilson MP, Leung KY, Herrera-Dominguez L, Sudiwala S, Pessia A, Clayton PT, Bryson K, Velagapudi V, Mills PB, Typas A, Greene NDE, Cabreiro F. Host-Microbe Co-metabolism Dictates Cancer Drug Efficacy in C. elegans. Cell. 2017;169:442-456.e18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 168]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
17.  Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, Poirier-Colame V, Roux A, Becharef S, Formenti S, Golden E, Cording S, Eberl G, Schlitzer A, Ginhoux F, Mani S, Yamazaki T, Jacquelot N, Enot DP, Bérard M, Nigou J, Opolon P, Eggermont A, Woerther PL, Chachaty E, Chaput N, Robert C, Mateus C, Kroemer G, Raoult D, Boneca IG, Carbonnel F, Chamaillard M, Zitvogel L. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1834]  [Cited by in F6Publishing: 2259]  [Article Influence: 251.0]  [Reference Citation Analysis (0)]
18.  Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre ML, Chang EB, Gajewski TF. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084-1089.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1979]  [Cited by in F6Publishing: 2501]  [Article Influence: 277.9]  [Reference Citation Analysis (0)]
19.  Dai Z, Coker OO, Nakatsu G, Wu WKK, Zhao L, Chen Z, Chan FKL, Kristiansen K, Sung JJY, Wong SH, Yu J. Multi-cohort analysis of colorectal cancer metagenome identified altered bacteria across populations and universal bacterial markers. Microbiome. 2018;6:70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 292]  [Article Influence: 48.7]  [Reference Citation Analysis (0)]
20.  Zhuang YP, Zhou HL, Chen HB, Zheng MY, Liang YW, Gu YT, Li WT, Qiu WL, Zhou HG. Gut microbiota interactions with antitumor immunity in colorectal cancer: From understanding to application. Biomed Pharmacother. 2023;165:115040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
21.  Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer. 2013;13:800-812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1054]  [Cited by in F6Publishing: 1127]  [Article Influence: 102.5]  [Reference Citation Analysis (1)]
22.  Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS, Sonnenburg JL. Diet-induced extinctions in the gut microbiota compound over generations. Nature. 2016;529:212-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 965]  [Cited by in F6Publishing: 1043]  [Article Influence: 130.4]  [Reference Citation Analysis (0)]
23.  Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1390]  [Cited by in F6Publishing: 1842]  [Article Influence: 307.0]  [Reference Citation Analysis (0)]
24.  Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, Boselli L, Routier E, Cassard L, Collins M, Vaysse T, Marthey L, Eggermont A, Asvatourian V, Lanoy E, Mateus C, Robert C, Carbonnel F. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28:1368-1379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 585]  [Cited by in F6Publishing: 816]  [Article Influence: 136.0]  [Reference Citation Analysis (0)]
25.  Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbé C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10799]  [Cited by in F6Publishing: 11263]  [Article Influence: 804.5]  [Reference Citation Analysis (0)]
26.  Jin Y, Dong H, Xia L, Yang Y, Zhu Y, Shen Y, Zheng H, Yao C, Wang Y, Lu S. The Diversity of Gut Microbiome is Associated With Favorable Responses to Anti-Programmed Death 1 Immunotherapy in Chinese Patients With NSCLC. J Thorac Oncol. 2019;14:1378-1389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 300]  [Article Influence: 60.0]  [Reference Citation Analysis (0)]
27.  Almeida A, Mitchell AL, Boland M, Forster SC, Gloor GB, Tarkowska A, Lawley TD, Finn RD. A new genomic blueprint of the human gut microbiota. Nature. 2019;568:499-504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 557]  [Cited by in F6Publishing: 717]  [Article Influence: 143.4]  [Reference Citation Analysis (0)]
28.  Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, Brochado AR, Fernandez KC, Dose H, Mori H, Patil KR, Bork P, Typas A. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555:623-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1160]  [Cited by in F6Publishing: 1121]  [Article Influence: 186.8]  [Reference Citation Analysis (0)]