51
|
Yang S, Zhao S, Ye Y, Jia L, Lou Y. Global research trends on the links between gut microbiota and cancer immunotherapy: A bibliometric analysis (2012-2021). Front Immunol 2022; 13:952546. [PMID: 36090978 PMCID: PMC9449151 DOI: 10.3389/fimmu.2022.952546] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
Background There is a crosstalk between gut microbiota (GM) and cancer immunotherapy (CI). The purpose of this study is to use bibliometric analysis to identify the highly cited papers relating to GM/CI and explore the research status and development trends of the GM/CI research. Methods A literature search regarding GM/CI publications from 2012 to 2021 was undertaken on July 4, 2022. The article titles, journals, authors, institutions, countries, total citations, keywords, and other information were extracted from the Science Citation Index Expanded (SCIE) of Web of Science Core Collection (WoSCC). The Bibliometrix of R package and VOSviewer were used for bibliometric analysis. Results A total of 665 papers were extracted. The number of papers has increased rapidly over the past decade, especially after 2018. The United States and China had the most publications and made great contributions to this field. Th5e Univ Texas MD Anderson Canc Ctr and Univ Paris Saclay were absolutely in the leading position in GM/CI. The most influential authors were Zitvogel L and Routy B. Frontiers in Immunology had the most publications and Science had the most total citations. Historical direct citation analysis explained the historical evolution in GM/CI. Highly cited papers and high-frequency keywords illustrated the current status and trends of GM/CI. Four clusters were identified and the important topics included the role of GM and antibiotics in CI, the methods of targeting GM to improve CI outcomes, the mechanism by which GM affects CI and the application of ICIs in melanoma. “Tumor microbiome”, “proton pump inhibitors” and “prognosis” may be the new focus of attention in the next few years. Conclusion This study filtered global publications on GM/CI correlation and analyzed their bibliometric characteristics, identified the most cited papers in GM/CI, and gained insight into the status, hotspots and trends of global GM/CI research, which may inform researchers and practitioners of future directions.
Collapse
Affiliation(s)
- Shanshan Yang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Oncology Department of Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Suya Zhao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yixiang Ye
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Liqun Jia
- Oncology Department of Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Liqun Jia, ; Yanni Lou,
| | - Yanni Lou
- Oncology Department of Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Liqun Jia, ; Yanni Lou,
| |
Collapse
|
52
|
Zhou J, Huang G, Wong WC, Hu DH, Zhu JW, Li R, Zhou H. The impact of antibiotic use on clinical features and survival outcomes of cancer patients treated with immune checkpoint inhibitors. Front Immunol 2022; 13:968729. [PMID: 35967438 PMCID: PMC9367677 DOI: 10.3389/fimmu.2022.968729] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background Nowadays, immune checkpoint inhibitors (ICIs) have become one of the essential immunotherapies for cancer patients. However, the impact of antibiotic (ATB) use on cancer patients treated with ICIs remains controversial. Methods Our research included retrospective studies and a randomized clinical trial (RCT) with cancer patients treated with ICIs and ATB, from the public database of PubMed, Web of Science, Embase, Cochrane, clinical trials, and JAMA. The survival outcomes included progression-free survival (PFS) and overall survival (OS). Meanwhile, hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated, and subgroup analyses were performed to determine the concrete association between ATB use and the prognosis of cancer patients treated in ICIs. Results Our results revealed that ATB use was associated with poor survival outcomes, including OS (HR: 1.94, 95% CI: 1.68–2.25, p <0.001) and PFS (HR: 1.83, 95% CI: 1.53–2.19, p <0.001). The subgroup analysis learned about the association between ATB use and the prognosis of cancer patients with ICI treatment, including 5 cancer types, 3 kinds of ICI, 5 different ATP windows, broad-spectrum ATB class, and ECOG score. ATB treatment was associated with poor OS of non-small-cell lung cancer (NSCLC), renal cell carcinoma (RCC), esophageal cancer (EC), and melanoma (MEL) in patients treated in ICIs, while non-small-cell lung cancer (NSCLC) and renal cell carcinoma (RCC) were associated with poor PFS. Meanwhile, it was strongly related to the ICI type and ATB window. Furthermore, it is firstly mentioned that the use of broad-spectrum ATB class was strongly associated with poor PFS. Conclusion In conclusion, our meta-analysis indicated that ATB use was significantly associated with poor OS and PFS of cancer patients treated with ICI immunotherapy, especially for patients with ATB use in the period of (−60 days; +30 days) near the initiation of ICI treatment. Also, different cancer types and the ICI type can also impact the survival outcome. This first reveals the strong relationship between the broad-spectrum ATB class and poor PFS. Still, more studies are needed for further study.
Collapse
Affiliation(s)
- Jiaxin Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- International School, Jinan University, Guangzhou, China
| | - Guowei Huang
- Shunde Hospital Affiliated to Jinan University, Guangzhou, China
| | - Wan-Ching Wong
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Da-hai Hu
- International School, Jinan University, Guangzhou, China
| | - Jie-wen Zhu
- College of Science and Engineering, Jinan University, Guangzhou, China
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Ruiman Li, ; Hong Zhou,
| | - Hong Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Ruiman Li, ; Hong Zhou,
| |
Collapse
|
53
|
Daley D. The Role of the Microbiome in Pancreatic Oncogenesis. Int Immunol 2022; 34:447-454. [PMID: 35863313 DOI: 10.1093/intimm/dxac036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/20/2022] [Indexed: 11/12/2022] Open
Abstract
Bacterial dysbiosis is evolving as an advocate for carcinogenesis and has been associated with pancreatic cancer progression and survival outcomes. The gut and pancreas of cancer patients harbor a unique microbiome that differs significantly from that of healthy individuals. We believe that the pancreatic cancer microbiome regulates tumorigenesis by altering host cell function and modulating immune cells, skewing them towards an immunosuppressive phenotype. Moreover, altering this pathogenic microbiome may enhance the efficacy of current therapies in pancreatic cancer and improve survival outcomes. This review highlights the findings on microbial modulation across various pre-clinical and clinical studies and provides insight into the potential of targeting the microbiome for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Donnele Daley
- Department of Surgery, University of Michigan, 1500 E Medical Center Drive, Ann Arbor, MI, USA
| |
Collapse
|
54
|
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; 15:47. [PMID: 35488243 PMCID: PMC9052532 DOI: 10.1186/s13045-022-01273-9] [Citation(s) in RCA: 225] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota have long been recognized to play a key role in human health and disease. Currently, several lines of evidence from preclinical to clinical research have gradually established that the gut microbiota can modulate antitumor immunity and affect the efficacy of cancer immunotherapies, especially immune checkpoint inhibitors (ICIs). Deciphering the underlying mechanisms reveals that the gut microbiota reprogram the immunity of the tumor microenvironment (TME) by engaging innate and/or adaptive immune cells. Notably, one of the primary modes by which the gut microbiota modulate antitumor immunity is by means of metabolites, which are small molecules that could spread from their initial location of the gut and impact local and systemic antitumor immune response to promote ICI efficiency. Mechanistic exploration provides novel insights for developing rational microbiota-based therapeutic strategies by manipulating gut microbiota, such as fecal microbiota transplantation (FMT), probiotics, engineered microbiomes, and specific microbial metabolites, to augment the efficacy of ICI and advance the age utilization of microbiota precision medicine.
Collapse
Affiliation(s)
- Yuting Lu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Xiangliang Yuan
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Miao Wang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Zhihao He
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ji Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
55
|
Messaoudene M, Pidgeon R, Richard C, Ponce M, Diop K, Benlaifaoui M, Nolin-Lapalme A, Cauchois F, Malo J, Belkaid W, Isnard S, Fradet Y, Dridi L, Velin D, Oster P, Raoult D, Ghiringhelli F, Boidot R, Chevrier S, Kysela DT, Brun YV, Falcone EL, Pilon G, Oñate FP, Gitton-Quent O, Le Chatelier E, Durand S, Kroemer G, Elkrief A, Marette A, Castagner B, Routy B. A Natural Polyphenol Exerts Antitumor Activity and Circumvents Anti-PD-1 Resistance through Effects on the Gut Microbiota. Cancer Discov 2022; 12:1070-1087. [PMID: 35031549 PMCID: PMC9394387 DOI: 10.1158/2159-8290.cd-21-0808] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/26/2021] [Accepted: 01/11/2022] [Indexed: 01/07/2023]
Abstract
Several approaches to manipulate the gut microbiome for improving the activity of cancer immune-checkpoint inhibitors (ICI) are currently under evaluation. Here, we show that oral supplementation with the polyphenol-rich berry camu-camu (CC; Myrciaria dubia) in mice shifted gut microbial composition, which translated into antitumor activity and a stronger anti-PD-1 response. We identified castalagin, an ellagitannin, as the active compound in CC. Oral administration of castalagin enriched for bacteria associated with efficient immunotherapeutic responses (Ruminococcaceae and Alistipes) and improved the CD8+/FOXP3+CD4+ ratio within the tumor microenvironment. Moreover, castalagin induced metabolic changes, resulting in an increase in taurine-conjugated bile acids. Oral supplementation of castalagin following fecal microbiota transplantation from ICI-refractory patients into mice supported anti-PD-1 activity. Finally, we found that castalagin binds to Ruminococcus bromii and promoted an anticancer response. Altogether, our results identify castalagin as a polyphenol that acts as a prebiotic to circumvent anti-PD-1 resistance. SIGNIFICANCE The polyphenol castalagin isolated from a berry has an antitumor effect through direct interactions with commensal bacteria, thus reprogramming the tumor microenvironment. In addition, in preclinical ICI-resistant models, castalagin reestablishes the efficacy of anti-PD-1. Together, these results provide a strong biological rationale to test castalagin as part of a clinical trial. This article is highlighted in the In This Issue feature, p. 873.
Collapse
Affiliation(s)
- Meriem Messaoudene
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Reilly Pidgeon
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Corentin Richard
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Mayra Ponce
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Khoudia Diop
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Myriam Benlaifaoui
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Alexis Nolin-Lapalme
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Florent Cauchois
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Julie Malo
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Wiam Belkaid
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Stephane Isnard
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | - Yves Fradet
- Centre de recherche du CHU de Québec, Oncology Division, CHU de Québec, Université Laval, Québec City, Quebec, Canada
| | - Lharbi Dridi
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Dominique Velin
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Paul Oster
- Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Didier Raoult
- Aix Marseille Université, IRD, AP-HM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | | | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-François Leclerc Cancer Center, UNICANCER, Dijon, France
- UMR CNRS 6302, Dijon, France
| | - Sandy Chevrier
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-François Leclerc Cancer Center, UNICANCER, Dijon, France
| | - David T. Kysela
- Faculté de Médecine, Département de Microbiologie, Infectiologie et Immunologie, University of Montreal, Montreal, Quebec, Canada
| | - Yves V. Brun
- Faculté de Médecine, Département de Microbiologie, Infectiologie et Immunologie, University of Montreal, Montreal, Quebec, Canada
| | - Emilia Liana Falcone
- Department of Immunity and Viral Infections, Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Geneviève Pilon
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute and Institute of Nutrition and Functional Foods, Laval University, Québec City, Quebec, Canada
| | | | | | | | - Sylvere Durand
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Centre de Recherche des Cordeliers, Équipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Centre de Recherche des Cordeliers, Équipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Arielle Elkrief
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute and Institute of Nutrition and Functional Foods, Laval University, Québec City, Quebec, Canada
| | - Bastien Castagner
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Bertrand Routy
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
- Hematology-Oncology Division, Department of Medicine, University of Montreal Healthcare Centre (CHUM), Montreal, Quebec, Canada
- Corresponding Author: Bertrand Routy, Hemato-Oncology, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec H2X 3H8, Canada. Phone: 514-890-8000; E-mail:
| |
Collapse
|
56
|
von Itzstein MS, Gonugunta AS, Sheffield T, Homsi J, Dowell JE, Koh AY, Raj P, Fattah F, Wang Y, Basava VS, Khan S, Park JY, Popat V, Saltarski JM, Gloria-McCutchen Y, Hsiehchen D, Ostmeyer J, Xie Y, Li QZ, Wakeland EK, Gerber DE. Association between Antibiotic Exposure and Systemic Immune Parameters in Cancer Patients Receiving Checkpoint Inhibitor Therapy. Cancers (Basel) 2022; 14:1327. [PMID: 35267634 PMCID: PMC8909108 DOI: 10.3390/cancers14051327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 01/19/2023] Open
Abstract
Antibiotic administration is associated with worse clinical outcomes and changes to the gut microbiome in cancer patients receiving immune checkpoint inhibitors (ICI). However, the effects of antibiotics on systemic immune function are unknown. We, therefore, evaluated antibiotic exposure, therapeutic responses, and multiplex panels of 40 serum cytokines and 124 antibodies at baseline and six weeks after ICI initiation, with p < 0.05 and false discovery rate (FDR) < 0.2 considered significant. A total of 251 patients were included, of whom the 135 (54%) who received antibiotics had lower response rates and shorter survival. Patients who received antibiotics prior to ICI initiation had modestly but significantly lower baseline levels of nucleolin, MDA5, c-reactive protein, and liver cytosol antigen type 1 (LC1) antibodies, as well as higher levels of heparin sulfate and Matrigel antibodies. After ICI initiation, antibiotic-treated patients had significantly lower levels of MDA5, CENP.B, and nucleolin antibodies. Although there were no clear differences in cytokines in the overall cohort, in the lung cancer subset (53% of the study population), we observed differences in IFN-γ, IL-8, and macrophage inflammatory proteins. In ICI-treated patients, antibiotic exposure is associated with changes in certain antibodies and cytokines. Understanding the relationship between these factors may improve the clinical management of patients receiving ICI.
Collapse
Affiliation(s)
- Mitchell S. von Itzstein
- Department of Internal Medicine, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.S.v.I.); (J.H.); (J.E.D.); (D.H.)
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
| | - Amrit S. Gonugunta
- School of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.S.G.); (V.P.)
| | - Thomas Sheffield
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (T.S.); (Y.W.); (J.O.)
| | - Jade Homsi
- Department of Internal Medicine, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.S.v.I.); (J.H.); (J.E.D.); (D.H.)
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
| | - Jonathan E. Dowell
- Department of Internal Medicine, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.S.v.I.); (J.H.); (J.E.D.); (D.H.)
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
| | - Andrew Y. Koh
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.R.); (S.K.); (Q.-Z.L.); (E.K.W.)
| | - Farjana Fattah
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
| | - Yiqing Wang
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (T.S.); (Y.W.); (J.O.)
| | - Vijay S. Basava
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
| | - Shaheen Khan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.R.); (S.K.); (Q.-Z.L.); (E.K.W.)
| | - Jason Y. Park
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Vinita Popat
- School of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.S.G.); (V.P.)
| | - Jessica M. Saltarski
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
| | - Yvonne Gloria-McCutchen
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
| | - David Hsiehchen
- Department of Internal Medicine, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.S.v.I.); (J.H.); (J.E.D.); (D.H.)
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
| | - Jared Ostmeyer
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (T.S.); (Y.W.); (J.O.)
| | - Yang Xie
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (T.S.); (Y.W.); (J.O.)
| | - Quan-Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.R.); (S.K.); (Q.-Z.L.); (E.K.W.)
| | - Edward K. Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (P.R.); (S.K.); (Q.-Z.L.); (E.K.W.)
| | - David E. Gerber
- Department of Internal Medicine, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.S.v.I.); (J.H.); (J.E.D.); (D.H.)
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (F.F.); (V.S.B.); (J.M.S.); (Y.G.-M.); (Y.X.)
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (T.S.); (Y.W.); (J.O.)
| |
Collapse
|
57
|
Jiang S, Geng S, Chen Q, Zhang C, Cheng M, Yu Y, Zhang S, Shi N, Dong M. Effects of Concomitant Antibiotics Use on Immune Checkpoint Inhibitor Efficacy in Cancer Patients. Front Oncol 2022; 12:823705. [PMID: 35223505 PMCID: PMC8864310 DOI: 10.3389/fonc.2022.823705] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/14/2022] [Indexed: 12/30/2022] Open
Abstract
Objective Immune checkpoint inhibitors (ICIs) have changed the outcomes of a variety of cancers in an unprecedented manner. Gut microbiome plays a crucial regulatory role in the antineoplastic therapy of ICIs, which can be influenced by antibiotic (ABX) administration. In this efficacy evaluation, we aimed to clarify the correlations of ABX administration with the survival of cancer patients receiving ICIs treatment. Method The eligible literatures were searched using PubMed, Cochrane Library, Web of Science, and Clinical trials.gov databases before Nov 2021. The correlations of ABX administration with progression-free survival (PFS) and overall survival (OS) were determined using Hazard ratios (HRs) coupled with 95% confidence intervals (CIs). Results A total of 12 studies enrolling 6010 cancer patients receiving ICIs treatment were included in this efficacy evaluation. ABX administration was significantly correlated worse PFS (HR=1.60, 95%CI=1.33-1.92, P<0.00001) and OS (HR=1.46, 95%CI=1.32-1.61, P<0.00001). Similar results were found in the subgroup analysis of non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC) and melanoma. Conclusions ABX use during ICIs treatment of cancer may significantly shorten PFS and OS. ABX should be used cautiously in cancer patients receiving ICIs. However, further validations are still essential due to existing publication bias.
Collapse
Affiliation(s)
- Shuai Jiang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuai Geng
- Department of Pharmacy, Strategic Support Force Medical Center, Beijing, China
| | - Qian Chen
- Department of Pharmacy, Beijing Boren Hospital, Beijing, China
| | - Chen Zhang
- Department of Medical Imaging, Strategic Support Force Medical Center, Beijing, China
| | - Mengfei Cheng
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Yu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuo Zhang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ning Shi
- Department of Pharmacy, Strategic Support Force Medical Center, Beijing, China
| | - Mei Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
58
|
Characterization of Alistipes montrealensis sp. nov., Isolated from Human Feces of a Patient with Metastatic Melanoma Treated with Immune Checkpoint Inhibitors. MICROBIOLOGY RESEARCH 2022. [DOI: 10.3390/microbiolres13010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Fecal microbiome culturomics of a cancer patient treated with immune checkpoint inhibitors led to the identification of a Gram-negative, rod-shaped, obligate anaerobic, non-motile, non-spore-forming bacterium, designated strain kh20T, which was phylogenetically assigned to the genus Alistipes. Strain kh20T demonstrated a 98.61% 16S rRNA sequence similarity with A.shahii WAL 8301T. The bacteria cells generated catalase but no oxidase. Iso-C15:0 (26.6%), anteiso-C15:0 (19.9%), and iso-C17:0 (17.2%) were the major cellular fatty acids identified in its composition. The G+C content of its genome was 57.2%. Strain kh20T showed significantly low values for DNA-DNA Hybridation (DDH ≤ 33.70%) and Average Nucleotide Identity (ANI ≤ 86.35%) compared with other Alistipes species. Based on these findings, we concluded that strain kh20T represented a novel bacterium, and we proposed the name Alistipes montrealensis sp. nov. (CECT 30384 and CSUR Q6005).
Collapse
|
59
|
Araji G, Maamari J, Ahmad FA, Zareef R, Chaftari P, Yeung SCJ. The Emerging Role of the Gut Microbiome in the Cancer Response to Immune Checkpoint Inhibitors: A Narrative Review. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2022; 5:13-25. [PMID: 35663831 PMCID: PMC9138420 DOI: 10.36401/jipo-21-10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/10/2021] [Accepted: 09/16/2021] [Indexed: 12/02/2022]
Abstract
The discovery of immune checkpoint inhibitors (ICIs) has revolutionized the care of cancer patients. However, the response to ICI therapy exhibits substantial interindividual variability. Efforts have been directed to identify biomarkers that predict the clinical response to ICIs. In recent years, the gut microbiome has emerged as a critical player that influences the efficacy of immunotherapy. An increasing number of studies have suggested that the baseline composition of a patient's gut microbiota and its dysbiosis are correlated with the outcome of cancer immunotherapy. This review tackles the rapidly growing body of evidence evaluating the relationship between the gut microbiome and the response to ICI therapy. Additionally, this review highlights the impact of antibiotic-induced dysbiosis on ICI efficacy and discusses the possible therapeutic interventions to optimize the gut microbiota composition to augment immunotherapy efficacy.
Collapse
Affiliation(s)
- Ghada Araji
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Julian Maamari
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Fatima Ali Ahmad
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Rana Zareef
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Patrick Chaftari
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
60
|
Bourgin M, Kepp O, Kroemer G. Immunostimulatory effects of vitamin B5 improve anticancer immunotherapy. Oncoimmunology 2022; 11:2031500. [PMID: 35096488 PMCID: PMC8794238 DOI: 10.1080/2162402x.2022.2031500] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Vitamin B5 (panthotenic acid), the precursor of coenzyme A (CoA), is contained in most food items and is produced by the intestinal microbiota. A recent study published in Cell Metabolism reports that vitamin B5 and CoA favor the differentiation of CD8+ cytotoxic T cells into interleukin-22 (IL-22)-producing Tc22 cells, likely through fueling mitochondrial metabolism. Importantly, in a small cohort of melanoma patients, the plasma levels of vitamin B5 positively correlate with responses to PD-1-targeted immunotherapy. Moreover, in mice, supplementation with vitamin B5 increases the efficacy of PD-L1-targeted cancer immunotherapy, and in vitro culture of T cells with CoA enhances their antitumor activity upon adoptive transfer into mice. These finding suggest that vitamin B5 is yet another B vitamin that stimulates anti-cancer immunosurveillance.
Collapse
Affiliation(s)
- Melanie Bourgin
- Centre de Recherche Des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche Des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Guido Kroemer
- Centre de Recherche Des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP,Paris, France
| |
Collapse
|
61
|
Abstract
Colorectal cancer (CRC) is still one of the most common types of cancer in the world, and the gut microbiome plays an important role in its development. The microbiome is involved in the carcinogenesis, formation and progression of CRC as well as its response to different systemic therapies. The composition of bacterial strains and the influence of geography, race, sex, and diet on the composition of the microbiome serve as important information for screening, early detection and prediction of the treatment outcome of CRC. Microbiome modulation is one of the most prospective new strategies in medicine to improve the health of individuals. Therefore, future research and clinical trials on the gut microbiome in oncology as well as in the treatment of CRC patients are warranted to determine the efficacy of systemic treatments for CRC, minimize adverse effects and increase survival rates.
Collapse
Affiliation(s)
- Martina Rebersek
- Department of Medical Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000, Ljubljana, Slovenia. .,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
62
|
Zhang J, Dai Z, Yan C, Zhang W, Wang D, Tang D. A new biological triangle in cancer: intestinal microbiota, immune checkpoint inhibitors and antibiotics. Clin Transl Oncol 2021; 23:2415-2430. [PMID: 34125407 PMCID: PMC8557192 DOI: 10.1007/s12094-021-02659-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy has revolutionized the treatment of many malignant tumors. Although immune checkpoint inhibitors (ICIs) can reactivate the anti-tumor activity of immune cells, sensitivity to immune checkpoint inhibitor therapy depends on the complex tumor immune processes. In recent years, numerous researches have demonstrated the role of intestinal microbiota in immunity and metabolism of the tumor microenvironment, as well as the efficacy of immunotherapy. Epidemiological studies have further demonstrated the efficacy of antibiotic therapy on the probability of patients' response to ICIs and predictability of the short-term survival of cancer patients. Disturbance to the intestinal microbiota significantly affects ICIs-mediated immune reconstitution and is considered a possible mechanism underlying the development of adverse effects during antibiotic-based ICIs treatment. Intestinal microbiota, antibiotics, and ICIs have gradually become important considerations for the titer of immunotherapy. In the case of immunotherapy, the rational use of antibiotics and intestinal microbiota is expected to yield a better prognosis for patients with malignant tumors.
Collapse
Affiliation(s)
- Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Zhujiang Dai
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Cheng Yan
- Dalian Medical University, Dalian, China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu Province Hospital, Yangzhou University, Yangzhou, 225001, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu Province Hospital, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
63
|
Varrone F, Mandrich L, Caputo E. Melanoma Immunotherapy and Precision Medicine in the Era of Tumor Micro-Tissue Engineering: Where Are We Now and Where Are We Going? Cancers (Basel) 2021; 13:5788. [PMID: 34830940 PMCID: PMC8616100 DOI: 10.3390/cancers13225788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/16/2022] Open
Abstract
Malignant melanoma still remains a cancer with very poor survival rates, although it is at the forefront of personalized medicine. Most patients show partial responses and disease progressed due to adaptative resistance mechanisms, preventing long-lasting clinical benefits to the current treatments. The response to therapies can be shaped by not only taking into account cancer cell heterogeneity and plasticity, but also by its structural context as well as the cellular component of the tumor microenvironment (TME). Here, we review the recent development in the field of immunotherapy and target-based therapy and how, in the era of tumor micro-tissue engineering, ex-vivo assays could help to enhance our melanoma biology knowledge in its complexity, translating it in the development of successful therapeutic strategies, as well as in the prediction of therapeutic benefits.
Collapse
Affiliation(s)
| | - Luigi Mandrich
- Research Institute on Terrestrial Ecosystem—IRET-CNR Via Pietro Castellino 111, I-80131 Naples, Italy;
| | - Emilia Caputo
- Institute of Genetics and Biophysics—IGB-CNR, “A. Buzzati-Traverso”, Via Pietro Castellino 111, I-80131 Naples, Italy
| |
Collapse
|
64
|
Yang W, Lei C, Song S, Jing W, Jin C, Gong S, Tian H, Guo T. Immune checkpoint blockade in the treatment of malignant tumor: current statue and future strategies. Cancer Cell Int 2021; 21:589. [PMID: 34727927 PMCID: PMC8565029 DOI: 10.1186/s12935-021-02299-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/24/2021] [Indexed: 01/08/2023] Open
Abstract
After being stagnant for decades, there has finally been a paradigm shift in the treatment of cancer with the emergence and application of immune checkpoint inhibitors (ICIs). The most extensively utilized ICIs are targeting the pathways involving programmed death-1 (PD-1) and cytotoxic T-lymphocyte associated protein 4 (CTLA-4). PD-1, as an crucial immune inhibitory molecule, by and large reasons the immune checkpoint response of T cells, making tumor cells get away from immune surveillance. Programmed cell death ligand-1 (PD-L1) is exceptionally expressed in most cancers cells and approves non-stop activation of the PD-1 pathway in the tumor microenvironment. PD-1/PD-L1 inhibitors can block the combination of PD-1 and PD-L1, inhibit hostile to regulatory signals, and restore the activity of T cells, thereby bettering immune response. The current researchers assume that the efficacy of these drugs is related to PD-L1 expression in tumor tissue, tumor mutation burden (TMB), and other emerging biomarkers. Although malignant tumors can benefit from the immunotherapy of PD-1/PD-L1 inhibitors, formulating a customized medication model and discovering biomarkers that can predict efficacy are the new trend in the new era of malignant tumor immunotherapy. This review summarizes the mechanism of action of PD-1/PD-L1 inhibitors, their clinical outcomes on various malignant tumors, their efficacy biomarkers, as well as predictive markers of irAEs.
Collapse
Affiliation(s)
- Wenwen Yang
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - Caining Lei
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
- The First Clinical Medicine College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, People's Republic of China
| | - Shaoming Song
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - Wutang Jing
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - Chuanwei Jin
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
- The First Clinical Medicine College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, People's Republic of China
| | - Shiyi Gong
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - Hongwei Tian
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China.
| | - Tiankang Guo
- Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
65
|
Jin X, Wu S, Bai Y. Risk factors and characteristics of bacterial infection during first-line chemotherapy for metastatic gastric or gastroesophageal junction adenocarcinoma. Support Care Cancer 2021; 30:2121-2129. [PMID: 34677650 DOI: 10.1007/s00520-021-06557-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/08/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE Chemotherapy-related bacterial infection is a common side effect in patients receiving chemotherapy. The purpose of this study was to determine the risk factors and characteristics of bacterial infection in metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma patients treated with combined chemotherapy. METHODS Patients with metastatic gastric or GEJ adenocarcinoma were followed up from 2013 to 2016 at Peking University First Hospital in China. Patients were treated with multiple cycles of combined chemotherapy. The incidence rate of bacterial infection and patients' clinical data were manually reviewed. RESULTS A total of 154 patients were eligible and were enrolled in this study. A median of 6 chemotherapy cycles were administered (range, 1-14 cycles). Chemotherapy-related bacterial infections were observed in 36 of 154 patients (23.4%). Pulmonary is the most common site of infections. Ninety-four percent of patients with bacterial infection during chemotherapy received broad-spectrum antibiotics. The independent risk factors for chemotherapy-related bacterial infection identified by multivariable analysis were Nutritional Risk Screening 2002 (NRS2002) ≥ 3 (P = 0.008), ≥ grade 3 neutropenia (P = 0.028), and Eastern Cooperative Oncology Group Performance Status (ECOG PS) ≥ 2 (P = 0.042). CONCLUSIONS Nearly a quarter of patients with metastatic gastric or GEJ adenocarcinoma who received combined chemotherapy had bacterial infection in this study. The proportion of broad-spectrum antibiotics used in patients with infection is very high. Improving nutritional status may help reduce the incidence of bacterial infection.
Collapse
Affiliation(s)
- Xuan Jin
- Department of Medical Oncology, Peking University First Hospital, Beijing, 100034, China
| | - Shikai Wu
- Department of Medical Oncology, Peking University First Hospital, Beijing, 100034, China.
| | - Yu Bai
- Department of Medical Oncology, Peking University First Hospital, Beijing, 100034, China
| |
Collapse
|
66
|
Kostine M, Mauric E, Tison A, Barnetche T, Barre A, Nikolski M, Rouxel L, Dutriaux C, Dousset L, Prey S, Beylot-Barry M, Seneschal J, Veillon R, Vergnenegre C, Daste A, Domblides C, Sionneau B, Gross-Goupil M, Ravaud A, Forcade E, Schaeverbeke T. Baseline co-medications may alter the anti-tumoural effect of checkpoint inhibitors as well as the risk of immune-related adverse events. Eur J Cancer 2021; 157:474-484. [PMID: 34649118 DOI: 10.1016/j.ejca.2021.08.036] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE As gut microbiota composition is an important determinant of response to immune checkpoint inhibitors (ICIs), we examined the effect of various co-medications known for their interaction with microbiota, when given at ICI initiation. PATIENTS AND METHODS We identified patients with advanced cancer treated with ICI between May 2015 and September 2017 in our institution. Co-medications given within 1 month before or 1 month after the first administration of ICI were reviewed from medical records. Survival data were analysed with univariable Cox regression, and the combined effect of multiple factors was assessed with factor analysis of mixed data (FAMD). The impact of co-medications on immune-related adverse events (irAEs) occurrence was also assessed. RESULTS A total of 635 patients were included. Psychotropic drugs (41%), proton pump inhibitors (PPIs; 38%), angiotensin-converting enzyme (ACE) inhibitors and/or angiotensin II receptor blockers (ARBs; 32%), glucocorticoids (26%), antibiotics (24%), statins (21%) and morphine (20%) were the most prescribed co-medications. Baseline use of antibiotics, glucocorticoids >10 mg/day, PPIs, psychotropic drugs, morphine and insulin was associated with significantly shortened overall survival and decreased tumour response, whereas coadministration of statins, ACEs and/or ARBs, non-steroidal anti-inflammatory drugs, aspirin and oral antidiabetic drugs did not impact patient outcomes. Treatments that altered the response to ICI were also associated with a decreased incidence of irAEs. FAMD revealed the respective weight of each factor or co-medication on the oncological outcomes. CONCLUSION Co-medications must be carefully assessed at the time of ICI initiation and clinicians aware of their possible deleterious effect, notably for PPIs, glucocorticoids, antibiotics and psychotropic drugs.
Collapse
Affiliation(s)
- Marie Kostine
- Department of Rheumatology, Bordeaux University Hospital, Bordeaux, France.
| | - Eleonora Mauric
- Department of Rheumatology, Bordeaux University Hospital, Bordeaux, France
| | - Alice Tison
- Department of Rheumatology, Bordeaux University Hospital, Bordeaux, France
| | - Thomas Barnetche
- Department of Rheumatology, Bordeaux University Hospital, Bordeaux, France
| | | | | | - Léa Rouxel
- Department of Rheumatology, Bordeaux University Hospital, Bordeaux, France
| | - Caroline Dutriaux
- Department of Dermatology, Bordeaux University Hospital, Bordeaux, France
| | - Léa Dousset
- Department of Dermatology, Bordeaux University Hospital, Bordeaux, France
| | - Sorilla Prey
- Department of Dermatology, Bordeaux University Hospital, Bordeaux, France
| | - Marie Beylot-Barry
- Department of Dermatology, Bordeaux University Hospital, Bordeaux, France
| | - Julien Seneschal
- Department of Dermatology, Bordeaux University Hospital, Bordeaux, France
| | - Rémi Veillon
- Department of Pulmonology, Bordeaux University Hospital, Bordeaux, France
| | | | - Amaury Daste
- Department of Oncology, Bordeaux University Hospital, Bordeaux, France
| | | | - Baptiste Sionneau
- Department of Oncology, Bordeaux University Hospital, Bordeaux, France
| | | | - Alain Ravaud
- Department of Oncology, Bordeaux University Hospital, Bordeaux, France
| | - Edouard Forcade
- Department of Hematology, Bordeaux University Hospital, Bordeaux, France
| | | | | |
Collapse
|
67
|
Li B, Gong T, Hao Y, Zhou X, Cheng L. Mining the Gut Microbiota for Microbial-Based Therapeutic Strategies in Cancer Immunotherapy. Front Oncol 2021; 11:721249. [PMID: 34589427 PMCID: PMC8473692 DOI: 10.3389/fonc.2021.721249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/23/2021] [Indexed: 02/05/2023] Open
Abstract
The past two decades witnessed a revolution in our understanding of host–microbiota interactions that led to the concept of the super-organism consisting of a eukaryotic part and a prokaryotic part. Owing to the critical role of gut microbiota in modulating the host immune system, it is not beyond all expectations that more and more evidence indicated that the shift of gut microbiota influenced responses to numerous forms of cancer immunotherapy. Therapy targeting gut microbiota is becoming a promising strategy to improve cancer immunotherapy. In this review, we discuss the role of the gut microbiota in response to cancer immunotherapy, the mechanisms that the gut microbiota influences cancer immunotherapy, and therapeutic strategies targeting gut microbiota to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Bolei Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Tao Gong
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Yu Hao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
68
|
Roberto M, Carconi C, Cerreti M, Schipilliti FM, Botticelli A, Mazzuca F, Marchetti P. The Challenge of ICIs Resistance in Solid Tumours: Could Microbiota and Its Diversity Be Our Secret Weapon? Front Immunol 2021; 12:704942. [PMID: 34489956 PMCID: PMC8417795 DOI: 10.3389/fimmu.2021.704942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
The human microbiota and its functional interaction with the human body were recently returned to the spotlight of the scientific community. In light of the extensive implementation of newer and increasingly precise genome sequencing technologies, bioinformatics, and culturomic, we now have an extraordinary ability to study the microorganisms that live within the human body. Most of the recent studies only focused on the interaction between the intestinal microbiota and one other factor. Considering the complexity of gut microbiota and its role in the pathogenesis of numerous cancers, our aim was to investigate how microbiota is affected by intestinal microenvironment and how microenvironment alterations may influence the response to immune checkpoint inhibitors (ICIs). In this context, we show how diet is emerging as a fundamental determinant of microbiota’s community structure and function. Particularly, we describe the role of certain dietary factors, as well as the use of probiotics, prebiotics, postbiotics, and antibiotics in modifying the human microbiota. The modulation of gut microbiota may be a secret weapon to potentiate the efficacy of immunotherapies. In addition, this review sheds new light on the possibility of administering fecal microbiota transplantation to modulate the gut microbiota in cancer treatment. These concepts and how these findings can be translated into the therapeutic response to cancer immunotherapies will be presented.
Collapse
Affiliation(s)
- Michela Roberto
- Department of Clinical and Molecular Medicine, Sant' Andrea University Hospital, Sapienza University of Rome, Rome, Italy.,Medical Oncology Unit, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Catia Carconi
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant' Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Micaela Cerreti
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant' Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Francesca Matilde Schipilliti
- Department of Clinical and Molecular Medicine, Sant' Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sant' Andrea University Hospital, Sapienza University of Rome, Rome, Italy.,Medical Oncology Unit, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, Sant' Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sant' Andrea University Hospital, Sapienza University of Rome, Rome, Italy.,Medical Oncology Unit, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
69
|
Acharya M, Kim T, Li C. Broad-Spectrum Antibiotic Use and Disease Progression in Early-Stage Melanoma Patients: A Retrospective Cohort Study. Cancers (Basel) 2021; 13:4367. [PMID: 34503177 PMCID: PMC8431240 DOI: 10.3390/cancers13174367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
Animal studies and a few clinical studies have reported mixed findings on the association between antibiotics and cancer incidence. Antibiotics may inhibit tumor cell growth, but could also alter the gut-microbiome-modulated immune system and increase the risk of cancer. Studies that assess how antibiotics affect the progression of cancer are limited. We evaluated the association between broad-spectrum antibiotic use and melanoma progression. We conducted a retrospective cohort study using IQVIA PharMetrics® Plus data (2008-2018). We identified patients with malignant melanoma who underwent wide local excision or Mohs micrographic surgery within 90 days of first diagnosis. Surgery date was the index date. Patients were excluded if they had any other cancer diagnosis or autoimmune disorders in 1 year before the index date ("baseline"). Exposure to broad-spectrum antibiotics was identified in three time windows using three cohorts: 3 months prior to the index date, 1 month after the index date, and 3 months after the index date. The covariates were patients' demographic and clinical characteristics identified in the 1-year baseline period. The patients were followed from the index date until cancer progression, loss of enrollment, or the end of 2 years after the index date. Progression was defined as: (i) any hospice care after surgery, (ii) a new round of treatment for melanoma (surgery, chemotherapy, immunotherapy, targeted therapy, or radiotherapy) 180 days after prior treatment, or (iii) a metastasis diagnosis or a diagnosis of a new nonmelanoma primary cancer at least 180 days after first melanoma diagnosis or prior treatment. A high-dimensional propensity score approach with inverse weighting was used to adjust for the patients' baseline differences. Cox proportional hazard regression was used for estimating the association. The final samples included 3930, 3831, and 3587 patients (mean age: 56 years). Exposure to antibiotics was 16% in the prior-3-months, 22% in the post-1-month, and 22% in the post-3-months. In the pre-3-months analysis, 9% of the exposed group and 9% of the unexposed group had progressed. Antibiotic use was not associated with melanoma progression (HR: 0.81; 95% CI: 0.57-1.14). However, antibiotic use in subsequent 1 month and subsequent 3 months was associated with 31% reduction (HR: 0.69; 95% CI: 0.51-0.92) and 32% reduction (HR: 0.68; 95% CI: 0.51-0.91) in progression, respectively. In this cohort of patients with likely early-stage melanoma cancer, antibiotic use in 1 month and 3 months after melanoma surgery was associated with a lower risk of melanoma progression. Future studies are warranted to validate the findings.
Collapse
Affiliation(s)
- Mahip Acharya
- Division of Pharmaceutical Evaluation and Policy, University of Arkansas for Medical Sciences College of Pharmacy, Little Rock, AR 72205, USA;
| | - Thomas Kim
- Department of Radiation Oncology, Rush University Medical College, Chicago, IL 60612, USA;
| | - Chenghui Li
- Division of Pharmaceutical Evaluation and Policy, University of Arkansas for Medical Sciences College of Pharmacy, Little Rock, AR 72205, USA;
| |
Collapse
|
70
|
Kulkarni AA, Ebadi M, Zhang S, Meybodi MA, Ali AM, DeFor T, Shanley R, Weisdorf D, Ryan C, Vasu S, Rashidi A, Patel MR. Comparative analysis of antibiotic exposure association with clinical outcomes of chemotherapy versus immunotherapy across three tumour types. ESMO Open 2021; 5:e000803. [PMID: 32900789 PMCID: PMC7477978 DOI: 10.1136/esmoopen-2020-000803] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/19/2020] [Accepted: 05/23/2020] [Indexed: 12/20/2022] Open
Abstract
Background In solid tumours, antibiotic use during immune checkpoint inhibitor (ICI) treatment is associated with shorter survival. Following allogeneic haematopoietic cell transplantation (allo-HCT), antibiotic-induced gut microbiome alterations are associated with risk of relapse and mortality. These findings suggest that the gut microbiota can modulate antitumour immune response across tumour types, though it is not clear if the impact on outcomes is specific to immune therapy. An important limitation of previous studies is that the analysis combined all antibiotic exposures irrespective of the antibiotic spectrum of activity. Whether antibiotic exposure during induction chemotherapy in acute myeloid leukaemia (AML) affects risk of relapse is also unknown. Patients and methods We performed a single-centred retrospective analysis of antibiotic exposures in metastatic/advanced non-small cell lung cancer (NSCLC) and renal cell cancer (RCC) receiving ICI and newly diagnosed AML patients receiving induction chemotherapy achieving a complete remission 1. Antibiotic use within 4 weeks before and 6 weeks after the ICI initiation were included. In AML patients, antibiotic exposures between days 1 and 28 of induction were collected. Antibiotics were a priori stratified based on spectrum of activity. Primary outcomes of interest were progression-free survival (PFS), overall survival (OS) in NSCLC and RCC and relapse-free survival (RFS) in AML. Results 140 patients with NSCLC, 55 with RCC and 143 with AML were included. In multivariable analysis, PFS and OS were shorter in NSCLC patients who received broad-spectrum anti-anaerobes (PFS, HR=3.2, 95% CI 1.6 to 6.2, p<0.01; OS, HR=1.7, 95% CI 0.8 to 3.6, p=0.19) or ‘other’ antibiotics (vancomycin-predominant) (PFS, HR=2.4, 95% CI 1.3 to 4.6, p<0.01; OS, HR=2.4, 95% CI 1.2 to 4.7, p=0.01). In RCC, patients who received penicillins/penicillin-class/early-generation cephalosporins had shorter PFS (HR=3.6, 95% CI 1.7 to 7.6, p<0.01) but similar OS (p=0.37). In the AML cohort, none of the exposures were associated with RFS. Conclusion In contrast to AML, antibiotic exposures in solid tumours affected clinical outcomes. The presence of an allogeneic effect (allo-HCT) or an augmented immune system (checkpoint blockade) may be necessary for microbiota mediation of relapse risk.
Collapse
Affiliation(s)
- Amit A Kulkarni
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maryam Ebadi
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shijia Zhang
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mohamad A Meybodi
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alaa M Ali
- Hematology, Oncology and Transplantation, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Todd DeFor
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ryan Shanley
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel Weisdorf
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Charles Ryan
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sumithira Vasu
- Hematology, Oncology and Transplantation, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Armin Rashidi
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Manish Ramesh Patel
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
71
|
Cruellas M, Yubero A, Zapata M, Galvez EM, Gascón M, Isla D, Lastra R, Martínez-Lostao L, Ocariz M, Pardo J, Ramírez A, Sesma A, Torres-Ramón I, Paño JR. How Could Antibiotics, Probiotics, and Corticoids Modify Microbiota and Its Influence in Cancer Immune Checkpoint Inhibitors: A Review. Infect Immun 2021; 89:e0066520. [PMID: 33526567 PMCID: PMC8370676 DOI: 10.1128/iai.00665-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has become a new paradigm in oncology, improving outcomes for several types of cancer. However, there are some aspects about its management that remain uncertain. One of the key points that needs better understanding is the interaction between immunotherapy and gut microbiome and how modulation of the microbiome might modify the efficacy of immunotherapy. Consequently, the negative impact of systemic antibiotics and corticosteroids on the efficacy of immunotherapy needs to be clarified.
Collapse
Affiliation(s)
- Mara Cruellas
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Alfonso Yubero
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - María Zapata
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | | | - Marta Gascón
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Dolores Isla
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Rodrigo Lastra
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Luis Martínez-Lostao
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
- Immunology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Department of Microbiology, Pediatrics, Radiology and Public Health, University of Zaragoza, Zaragoza, Spain
- Aragon Nanoscience institute, Zaragoza, Spain
- Aragon Materials Science Institute, Zaragoza, Spain
| | - Maitane Ocariz
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Julián Pardo
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
- ARAID Foundation (IIS Aragón), Zaragoza, Spain
- Microbiology, Preventive Medicine and Public Health Department, Medicine, University of Zaragoza, Zaragoza, Spain
- Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine Network (CIBER-BBN), Madrid, Spain
| | - Ariel Ramírez
- Nanotoxicology and Immunotoxicology Unit (IIS Aragón), Zaragoza, Spain
| | - Andrea Sesma
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Irene Torres-Ramón
- Medical Oncology Department, University Hospital Lozano Blesa, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - José Ramón Paño
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
- Infectious Disease Department, University Hospital Lozano Blesa, Zaragoza, Spain
| |
Collapse
|
72
|
Chau J, Zhang J. Tying Small Changes to Large Outcomes: The Cautious Promise in Incorporating the Microbiome into Immunotherapy. Int J Mol Sci 2021; 22:ijms22157900. [PMID: 34360663 PMCID: PMC8347117 DOI: 10.3390/ijms22157900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/16/2022] Open
Abstract
The role of the microbiome in immunology is a rapidly burgeoning topic of study. Given the increasing use of immune checkpoint inhibitor (ICI) therapy in cancers, along with the recognition that carcinogenesis has been linked to dysregulations of the immune system, much attention is now directed at potentiation of ICI efficacy, as well as minimizing the incidence of treatment-associated immune-related adverse events (irAEs). We provide an overview of the major research establishing links between the microbiome to tumorigenesis, chemotherapy and radiation potentiation, and ICI efficacy and irAE development.
Collapse
Affiliation(s)
- Justin Chau
- Division of Hematology, Oncology and Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA 52246, USA;
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Cancer Biology, University of Kansas Cancer Center, Kansas City, KS 66160, USA
- Correspondence: ; Tel.: +1-(913)-588-8150; Fax: +1-(913)-588-4085
| |
Collapse
|
73
|
Liu X, Chen Y, Zhang S, Dong L. Gut microbiota-mediated immunomodulation in tumor. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:221. [PMID: 34217349 PMCID: PMC8254267 DOI: 10.1186/s13046-021-01983-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/16/2021] [Indexed: 12/15/2022]
Abstract
Tumor immunity consists of various types of cells, which serve an important role in antitumor therapy. The gastrointestinal tract is colonized by trillions of microorganisms, which form the gut microbiota. In addition to pathogen defense and maintaining the intestinal ecosystem, gut microbiota also plays a pivotal role in various physiological processes. Recently, the association between these symbionts and cancer, ranging from oncogenesis and cancer progression to resistance or sensitivity to antitumor therapies, has attracted much attention. Metagenome analysis revealed a significant difference between the gut microbial composition of cancer patients and healthy individuals. Moreover, modulation of microbiome could improve therapeutic response to immune checkpoint inhibitors (ICIs). These findings suggest that microbiome is involved in cancer pathogenesis and progression through regulation of tumor immunosurveillance, although the exact mechanisms remain largely unknown. This review focuses on the interaction between the microbiome and tumor immunity, with in-depth discussion regarding the therapeutic potential of modulating gut microbiota in ICIs. Further investigations are warranted before gut microbiota can be introduced into clinical practice.
Collapse
Affiliation(s)
- Xinyi Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200030, People's Republic of China.,Shanghai Medical College of Fudan University, 130 Dongan Road, Shanghai, 200030, People's Republic of China
| | - Yanjie Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200030, People's Republic of China
| | - Si Zhang
- Shanghai Medical College of Fudan University, 130 Dongan Road, Shanghai, 200030, People's Republic of China.
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200030, People's Republic of China. .,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
74
|
Hayase E, Jenq RR. Role of the intestinal microbiome and microbial-derived metabolites in immune checkpoint blockade immunotherapy of cancer. Genome Med 2021; 13:107. [PMID: 34162429 PMCID: PMC8220726 DOI: 10.1186/s13073-021-00923-w] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that block immune inhibitory pathways. Administration of ICIs augments T cell-mediated immune responses against tumor, resulting in improved overall survival in cancer patients. It has emerged that the intestinal microbiome can modulate responses to ICIs via the host immune system and that the use of antibiotics can lead to reduced efficacy of ICIs. Recently, reports that fecal microbiota transplantation can lead to ICI therapy responses in patients previously refractory to therapy suggest that targeting the microbiome may be a viable strategy to reprogram the tumor microenvironment and augment ICI therapy. Intestinal microbial metabolites may also be linked to response rates to ICIs. In addition to response rates, certain toxicities that can arise during ICI therapy have also been found to be associated with the intestinal microbiome, including in particular colitis. A key mechanistic question is how certain microbes can enhance anti-tumor responses or, alternatively, predispose to ICI-associated colitis. Evidence has emerged that the intestinal microbiome can modulate outcomes to ICI therapies via two major mechanisms, including those that are antigen-specific and those that are antigen-independent. Antigen-specific mechanisms occur when epitopes are shared between microbial and tumor antigens that could enhance, or, alternatively, reduce anti-tumor immune responses via cross-reactive adaptive immune cells. Antigen-independent mechanisms include modulation of responses to ICIs by engaging innate and/or adaptive immune cells. To establish microbiome-based biomarkers of outcomes and specifically modulate the intestinal microbiome to enhance efficacy of ICIs in cancer immunotherapy, further prospective interventional studies will be required.
Collapse
Affiliation(s)
- Eiko Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert R Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- CPRIT Scholar in Cancer Research, Houston, TX, USA.
| |
Collapse
|
75
|
Mohiuddin JJ, Chu B, Facciabene A, Poirier K, Wang X, Doucette A, Zheng C, Xu W, Anstadt EJ, Amaravadi RK, Karakousis GC, Mitchell TC, Huang AC, Shabason JE, Lin A, Swisher-McClure S, Maity A, Schuchter LM, Lukens JN. Association of Antibiotic Exposure With Survival and Toxicity in Patients With Melanoma Receiving Immunotherapy. J Natl Cancer Inst 2021; 113:162-170. [PMID: 32294209 DOI: 10.1093/jnci/djaa057] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/20/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Gut microbial diversity is associated with improved response to immune checkpoint inhibitors (ICI). Based on the known detrimental impact that antibiotics have on microbiome diversity, we hypothesized that antibiotic receipt prior to ICI would be associated with decreased survival. METHODS Patients with stage III and IV melanoma treated with ICI between 2008 and 2019 were selected from an institutional database. A window of antibiotic receipt within 3 months prior to the first infusion of ICI was prespecified. The primary outcome was overall survival (OS), and secondary outcomes were melanoma-specific mortality and immune-mediated colitis requiring intravenous steroids. All statistical tests were two-sided. RESULTS There were 568 patients in our database of which 114 received antibiotics prior to ICI. Of the patients, 35.9% had stage III disease. On multivariable Cox proportional hazards analysis of patients with stage IV disease, the antibiotic-exposed group had statistically significantly worse OS (hazard ratio [HR] = 1.81, 95% confidence interval [CI] = 1.27 to 2.57; P <.001). The same effect was observed among antibiotic-exposed patients with stage III disease (HR = 2.78, 95% CI = 1.31 to 5.87; P =.007). When limited to only patients who received adjuvant ICI (n = 89), antibiotic-exposed patients also had statistically significantly worse OS (HR = 4.84, 95% CI = 1.09 to 21.50; P =.04). The antibiotic group had a greater incidence of colitis (HR = 2.14, 95% CI = 1.02 to 4.52; P =.046). CONCLUSION Patients with stage III and IV melanoma exposed to antibiotics prior to ICI had statistically significantly worse OS than unexposed patients. Antibiotic exposure was associated with greater incidence of moderate to severe immune-mediated colitis. Given the large number of antibiotics prescribed annually, physicians should be judicious with their use in cancer populations likely to receive ICI.
Collapse
Affiliation(s)
- Jahan J Mohiuddin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Chu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrea Facciabene
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Kendra Poirier
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Xingmei Wang
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Abigail Doucette
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Cathy Zheng
- Tara Miller Melanoma Center, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Xu
- Tara Miller Melanoma Center, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily J Anstadt
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi K Amaravadi
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Giorgos C Karakousis
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Tara C Mitchell
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander C Huang
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob E Shabason
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander Lin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Amit Maity
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lynn M Schuchter
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John N Lukens
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
76
|
Classen AY, Henze L, von Lilienfeld-Toal M, Maschmeyer G, Sandherr M, Graeff LD, Alakel N, Christopeit M, Krause SW, Mayer K, Neumann S, Cornely OA, Penack O, Weißinger F, Wolf HH, Vehreschild JJ. Primary prophylaxis of bacterial infections and Pneumocystis jirovecii pneumonia in patients with hematologic malignancies and solid tumors: 2020 updated guidelines of the Infectious Diseases Working Party of the German Society of Hematology and Medical Oncology (AGIHO/DGHO). Ann Hematol 2021; 100:1603-1620. [PMID: 33846857 PMCID: PMC8116237 DOI: 10.1007/s00277-021-04452-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/04/2021] [Indexed: 12/11/2022]
Abstract
Hematologic and oncologic patients with chemo- or immunotherapy-related immunosuppression are at substantial risk for bacterial infections and Pneumocystis jirovecii pneumonia (PcP). As bacterial resistances are increasing worldwide and new research reshapes our understanding of the interactions between the human host and bacterial commensals, administration of antibacterial prophylaxis has become a matter of discussion. This guideline constitutes an update of the 2013 published guideline of the Infectious Diseases Working Party (AGIHO) of the German Society for Hematology and Medical Oncology (DGHO). It gives an overview about current strategies for antibacterial prophylaxis in cancer patients while taking into account the impact of antibacterial prophylaxis on the human microbiome and resistance development. Current literature published from January 2012 to August 2020 was searched and evidence-based recommendations were developed by an expert panel. All recommendations were discussed and approved in a consensus conference of the AGIHO prior to publication. As a result, we present a comprehensive update and extension of our guideline for antibacterial and PcP prophylaxis in cancer patients.
Collapse
Affiliation(s)
- Annika Y Classen
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Herderstr. 52-54, 50931, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Larissa Henze
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Marie von Lilienfeld-Toal
- Department of Hematology and Oncology, Clinic for Internal Medicine II, University Hospital Jena, Jena, Germany
| | - Georg Maschmeyer
- Hematology, Oncology and Palliative Care, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Michael Sandherr
- Specialist Clinic for Haematology and Oncology, Medical Care Center Penzberg, Penzberg, Germany
| | - Luisa Durán Graeff
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Herderstr. 52-54, 50931, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Nael Alakel
- Department I of Internal Medicine, Hematology and Oncology, University Hospital Dresden, Dresden, Germany
| | - Maximilian Christopeit
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Stefan W Krause
- Department of Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Karin Mayer
- Medical Clinic III for Oncology, Hematology, Immunooncology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Silke Neumann
- Interdisciplinary Center for Oncology, Wolfsburg, Germany
| | - Oliver A Cornely
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Herderstr. 52-54, 50931, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Chair Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany
| | - Olaf Penack
- Medical Department for Hematology, Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Florian Weißinger
- Department for Internal Medicine, Hematology/Oncology, and Palliative Care, Evangelisches Klinikum Bethel v. Bodelschwinghsche Stiftungen Bethel, Bielefeld, Germany
| | - Hans-Heinrich Wolf
- Department IV of Internal Medicine, University Hospital Halle, Halle, Germany
| | - Jörg Janne Vehreschild
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Herderstr. 52-54, 50931, Cologne, Germany.
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany.
- Department of Internal Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
77
|
Naqash AR, Kihn-Alarcón AJ, Stavraka C, Kerrigan K, Maleki Vareki S, Pinato DJ, Puri S. The role of gut microbiome in modulating response to immune checkpoint inhibitor therapy in cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1034. [PMID: 34277834 PMCID: PMC8267312 DOI: 10.21037/atm-20-6427] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/12/2021] [Indexed: 12/16/2022]
Abstract
Immunotherapy has led to a paradigm shift in the treatment of several cancers. There have been significant efforts to identify biomarkers that can predict response and toxicities related to immune checkpoint inhibitor (ICPI) therapy. Despite these advances, it has been challenging to tease out why a subset of patients benefit more than others or why certain patients experience immune-related adverse events (irAEs). Although the immune-modulating properties of the human gut bacterial ecosystem are yet to be fully elucidated, there has been growing interest in evaluating the role of the gut microbiome in shaping the therapeutic response to cancer immunotherapy. Considerable research efforts are currently directed to utilizing metagenomic and metabolic profiling of stool microbiota in patients on ICPI-based therapies. Dysbiosis or loss of microbial diversity has been associated with a poor treatment response to ICPIs and worse survival outcomes in cancer patients. Emerging data have shown that certain bacterial strains, such as Faecalibacterium that confer sensitivity to ICPI, also have a higher propensity to increase the risk of irAEs. Additionally, the microbiome can modulate the local immune response at the intestinal interface and influence the trafficking of bacterial peptide primed T-cells distally, influencing the toxicity patterns to ICPI. Antibiotic or diet induced alterations in composition of the microbiome can also indirectly alter the production of certain bacterial metabolites such as deoxycholate and short chain fatty acids that can influence the anti-tumor tolerogenesis. Gaining sufficient understanding of the exact mechanisms underpinning the interplay between ICPI induced anti-tumor immunity and the immune modulatory role gut microbiome can be vital in identifying potential avenues of improving outcomes to cancer immunotherapy. In the current review, we have summarized and highlighted the key emerging data supporting the role of gut microbiome in regulating response to ICPIs in cancer.
Collapse
Affiliation(s)
- Abdul Rafeh Naqash
- Department of Investigational Cancer Therapeutics, National Cancer Institute, Bethesda, MD, USA
| | - Alba J Kihn-Alarcón
- Department of Research, Liga Nacional Contra el Cáncer & Instituto de Cancerología, Guatemala City, Guatemala
| | - Chara Stavraka
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, UK
| | - Kathleen Kerrigan
- Division of Medical Oncology Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| | - Saman Maleki Vareki
- Division of Experimental Oncology, Department of Oncology, University of Western Ontario, London, ON, Canada.,Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON, Canada.,Cancer Research Laboratory Program, Lawson Health Research Institute, London, ON, Canada
| | | | - Sonam Puri
- Division of Medical Oncology Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
78
|
Meriggi F, Zaniboni A. Antibiotics and steroids, the double enemies of anticancer immunotherapy: a review of the literature. Cancer Immunol Immunother 2021; 70:1511-1517. [PMID: 33165628 PMCID: PMC10991597 DOI: 10.1007/s00262-020-02786-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
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.
Collapse
Affiliation(s)
- Fausto Meriggi
- Oncology Department, Poliambulanza Foundation, Via Leonida Bissolati 57, 25124, Brescia, Italy.
| | - Alberto Zaniboni
- Oncology Department, Poliambulanza Foundation, Via Leonida Bissolati 57, 25124, Brescia, Italy
| |
Collapse
|
79
|
Schubert ML, Rohrbach R, Schmitt M, Stein-Thoeringer CK. The Potential Role of the Intestinal Micromilieu and Individual Microbes in the Immunobiology of Chimeric Antigen Receptor T-Cell Therapy. Front Immunol 2021; 12:670286. [PMID: 34135898 PMCID: PMC8200823 DOI: 10.3389/fimmu.2021.670286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Cellular immunotherapy with chimeric antigen receptor (CAR)-T cells (CARTs) represents a breakthrough in the treatment of hematologic malignancies. CARTs are genetically engineered hybrid receptors that combine antigen-specificity of monoclonal antibodies with T cell function to direct patient-derived T cells to kill malignant cells expressing the target (tumor) antigen. CARTs have been introduced into clinical medicine as CD19-targeted CARTs for refractory and relapsed B cell malignancies. Despite high initial response rates, current CART therapies are limited by a long-term loss of antitumor efficacy, the occurrence of toxicities, and the lack of biomarkers for predicting therapy and toxicity outcomes. In the past decade, the gut microbiome of mammals has been extensively studied and evidence is accumulating that human health, apart from our own genome, largely depends on microbes that are living in and on the human body. The microbiome encompasses more than 1000 bacterial species who collectively encode a metagenome that guides multifaceted, bidirectional host-microbiome interactions, primarily through the action of microbial metabolites. Increasing knowledge has been accumulated on the role of the gut microbiome in T cell-driven anticancer immunotherapy. It has been shown that antibiotics, dietary components and gut microbes reciprocally affect the efficacy and toxicity of allogeneic hematopoietic cell transplantation (allo HCT) as the prototype of T cell-based immunotherapy for hematologic malignancies, and that microbiome diversity metrics can predict clinical outcomes of allo HCTs. In this review, we will provide a comprehensive overview of the principles of CD19-CART immunotherapy and major aspects of the gut microbiome and its modulators that impact antitumor T cell transfer therapies. We will outline i) the extrinsic and intrinsic variables that can contribute to the complex interaction of the gut microbiome and host in CART immunotherapy, including ii) antibiotic administration affecting loss of colonization resistance, expansion of pathobionts and disturbed mucosal and immunological homeostasis, and ii) the role of specific gut commensals and their microbial virulence factors in host immunity and inflammation. Although the role of the gut microbiome in CART immunotherapy has only been marginally explored so far, this review may open a new chapter and views on putative connections and mechanisms.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Klinik fuer Haematologie, Onkologie und Rheumatologie, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Roman Rohrbach
- Research Division Microbiome and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Michael Schmitt
- Klinik fuer Haematologie, Onkologie und Rheumatologie, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Christoph K Stein-Thoeringer
- Research Division Microbiome and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany.,Klinik fuer Medizinische Onkologie, Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg, Germany
| |
Collapse
|
80
|
Bersanelli M, Giannarelli D, De Giorgi U, Pignata S, Di Maio M, Clemente A, Verzoni E, Giusti R, Di Napoli M, Aprile G, Ermacora P, Catino A, Scotti V, Mazzoni F, Guglielmini PF, Veccia A, Maruzzo M, Rossi E, Grossi F, Casadei C, Ficorella C, Montesarchio V, Verderame F, Rizzo M, Guaitoli G, Fratino L, Accettura C, Mencoboni M, Zustovich F, Baldessari C, Cinieri S, Camerini A, Laera L, Sorarù M, Zucali PA, Guadalupi V, Leonardi F, Tiseo M, Tognetto M, Di Costanzo F, Pinto C, Negrini G, Russo A, Migliorino MR, Filetti M, Buti S. INfluenza Vaccine Indication During therapy with Immune checkpoint inhibitors: a multicenter prospective observational study (INVIDIa-2). J Immunother Cancer 2021; 9:jitc-2021-002619. [PMID: 34016723 PMCID: PMC8141439 DOI: 10.1136/jitc-2021-002619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 01/03/2023] Open
Abstract
Background Until now, no robust data supported the efficacy, safety and recommendation for influenza vaccination in patients with cancer receiving immune checkpoint inhibitors (ICIs). Methods The prospective multicenter observational INfluenza Vaccine Indication During therapy with Immune checkpoint inhibitors (INVIDIa-2) study investigated the clinical effectiveness of influenza vaccination in patients with advanced cancer receiving ICIs, enrolled in 82 Italian centers from October 2019 to January 2020. The primary endpoint was the time-adjusted incidence of influenza-like illness (ILI) until April 30, 2020. Secondary endpoints regarded ILI severity and vaccine safety. Results The study enrolled 1279 patients; 1188 patients were evaluable for the primary endpoint analysis. Of them, 48.9% (581) received influenza vaccination. The overall ILI incidence was 8.2% (98 patients). Vaccinated patients were significantly more frequently elderly (p<0.0001), males (p=0.004), with poor European Cooperative Oncology Group performance status (p=0.009), affected by lung cancer (p=0.01), and by other non-cancer comorbidities (p<0.0001) when compared with unvaccinated. ILI incidence was not different basing on influenza vaccination: the time-to-ILI was similar in vaccinated and unvaccinated patients (p=0.62). ILI complications were significantly less frequent for patients receiving the vaccination (11.8% vs 38.3% in unvaccinated, p=0.002). ILI-related intravenous therapies were significantly less frequent in vaccinated patients than in unvaccinated (11.8% vs 29.8%, p=0.027). ILI lethality was, respectively, 0% in vaccinated and 4.3% in unvaccinated patients. Vaccine-related adverse events were rare and mild (1.5%, grades 1–2). Conclusion The INVIDIa-2 study results support a positive recommendation for influenza vaccination in patients with advanced cancer receiving immunotherapy.
Collapse
Affiliation(s)
- Melissa Bersanelli
- Medicine and Surgery Department, University of Parma, Parma, Italy .,Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Diana Giannarelli
- Biostatistical Unit, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sandro Pignata
- UC Oncologia Medica Uro-Ginecologica, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Napoli, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Torino, Italy.,Medical Oncology, Azienda Ospedaliera Ordine Mauriziano di Torino, Torino, Italy
| | - Alberto Clemente
- Biostatistics and Clinical Research Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Elena Verzoni
- SS.Oncologia Genitourinaria, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Marilena Di Napoli
- UC Oncologia Medica Uro-Ginecologica, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Napoli, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| | - Paola Ermacora
- Dipartimento di Oncologia, Presidio Ospedaliero Universitario Santa Maria della Misericordia, Azienda sanitaria universitaria integrata Friuli Centrale, Udine, Italy
| | - Annamaria Catino
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Vieri Scotti
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Firenze, Italy
| | | | | | | | - Marco Maruzzo
- Oncologia Medica 1, Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| | - Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Grossi
- Medical Oncology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Corrado Ficorella
- Department of Biotechnological and Applied Clinical Sciences, St Salvatore Hospital, University of L'Aquila, L'Aquila, Italy
| | - Vincenzo Montesarchio
- UOC Oncologia, Azienda Ospedaliera Specialistica dei Colli, Ospedale Monaldi, Napoli, Italy
| | | | - Mimma Rizzo
- Oncologia Traslazionale, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Giorgia Guaitoli
- Medical Oncology Unit, Universita degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Lucia Fratino
- Medical Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | | | | | - Fable Zustovich
- UOC Oncologia di Belluno, Dipartimento di Oncologia Clinica, AULSS 1 Dolomiti, Ospedale S.Martino, Belluno, Italy
| | - Cinzia Baldessari
- Medical Oncology Unit, Universita degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, Brindisi, Italy
| | - Andrea Camerini
- Oncologia Medica, Ospedale della Versilia, Lido di Camaiore, Italy
| | - Letizia Laera
- Medical Oncology, Ospedale Generale Regionale F Miulli, Acquaviva delle Fonti, Puglia, Italy
| | - Mariella Sorarù
- Medical Oncology, Camposampiero Hospital, ULSS 6 Euganea, Padova, Italy
| | - Paolo Andrea Zucali
- Department of Oncology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Valentina Guadalupi
- UC Oncologia Medica Uro-Ginecologica, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Marcello Tiseo
- Medicine and Surgery Department, University of Parma, Parma, Italy.,Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Michele Tognetto
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | - Carmine Pinto
- Medical Oncology Unit, AUSL-IRCCS of Reggio Emilia, Reggio Emilia, Italy.,Presidency of the Federation of Italian Cooperative Oncology Groups (FICOG), Milan, Italy
| | | | - Antonio Russo
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, Palermo, Italy
| | | | - Marco Filetti
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, Sapienza University of Rome, Roma, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | |
Collapse
|
81
|
Deng Y, Liu SY, Chua SL, Khoo BL. The effects of biofilms on tumor progression in a 3D cancer-biofilm microfluidic model. Biosens Bioelectron 2021; 180:113113. [DOI: 10.1016/j.bios.2021.113113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 12/13/2022]
|
82
|
Cvetkovic L, Régis C, Richard C, Derosa L, Leblond A, Malo J, Messaoudene M, Desilets A, Belkaid W, Elkrief A, Routy B, Juneau D. Physiologic colonic uptake of 18F-FDG on PET/CT is associated with clinical response and gut microbiome composition in patients with advanced non-small cell lung cancer treated with immune checkpoint inhibitors. Eur J Nucl Med Mol Imaging 2021; 48:1550-1559. [PMID: 33128571 DOI: 10.1007/s00259-020-05081-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) represent the backbone treatment for advanced non-small cell lung cancer (NSCLC). Emerging data suggest that increased gut microbiome diversity is associated with favorable response to ICI and that antibiotic-induced dysbiosis is associated with deleterious outcomes. 18F-FDG physiologic colonic uptake on PET/CT increases following treatment with antibiotics (ATB) and could act as a surrogate marker for microbiome composition and predict prognosis. The aim of this study was to determine if 18F-FDG physiologic colonic uptake prior to ICI initiation correlates with gut microbiome profiling and clinical outcomes in patients with advanced NSCLC. METHODS Seventy-one patients with advanced NSCLC who underwent a PET/CT prior to ICI were identified. Blinded colonic contouring was performed for each colon segment and patients were stratified according to the median of the average colon SUVmax as well as for each segment in low vs. high SUVmax groups. Response rate, progression-free survival (PFS), and overall survival (OS) were compared in the low vs. high SUVmax groups. Gut microbiome composition was analyzed for 23 patients using metagenomics sequencing. RESULTS The high colon SUVmax group had a higher proportion of non-responders (p = 0.033) and significantly shorter PFS (4.1 vs. 11.3 months, HR 1.94, 95% CI 1.11-3.41, p = 0.005). High caecum SUVmax correlated with numerically shorter OS (10.8 vs. 27.6 months, HR 1.85, 95% CI 0.97-3.53, p = 0.058). Metagenomics sequencing revealed distinctive microbiome populations in each group. Patients with low caecum SUVmax had higher microbiome diversity (p = 0.046) and were enriched with Bifidobacteriaceae, Lachnospiraceae, and Bacteroidaceae. CONCLUSIONS Lower colon physiologic 18F-FDG uptake on PET/CT prior to ICI initiation was associated with better clinical outcomes and higher gut microbiome diversity in patients with advanced NSCLC. Here, we propose that 18F-FDG physiologic colonic uptake on PET/CT could serve as a potential novel marker of gut microbiome composition and may predict clinical outcomes in this population.
Collapse
Affiliation(s)
- Lena Cvetkovic
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Claudine Régis
- Department of Radiology and Nuclear Medicine, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Radiology and Nuclear Medicine, Institut de Cardiologie de Montréal, Montréal, Québec, Canada
| | - Corentin Richard
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Lisa Derosa
- Department of Tumor Immunology and Immunotherapy INSERM U1015, Gustave Roussy Cancer Center, Villejuif, France
| | - Antoine Leblond
- Department of Radiology and Nuclear Medicine, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Radiology and Nuclear Medicine, Centre Hospitalier Affilié Universitaire Régional (CHAUR), Trois-Rivières, Québec, Canada
| | - Julie Malo
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Meriem Messaoudene
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Antoine Desilets
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Wiam Belkaid
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Arielle Elkrief
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada
| | - Bertrand Routy
- Department of Hematology and Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada.
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada.
| | - Daniel Juneau
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, Québec, Canada.
- Department of Radiology and Nuclear Medicine, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada.
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
83
|
Farmakiotis D. The Human Microbiome and Checkpoint Inhibition: Potential Benefits From Antibiotic Stewardship. Clin Infect Dis 2021; 71:701-702. [PMID: 31608406 DOI: 10.1093/cid/ciz1003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/07/2019] [Indexed: 12/19/2022] Open
|
84
|
Chandra V, McAllister F. Therapeutic potential of microbial modulation in pancreatic cancer. Gut 2021; 70:gutjnl-2019-319807. [PMID: 33906958 PMCID: PMC8292583 DOI: 10.1136/gutjnl-2019-319807] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 03/16/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Vidhi Chandra
- Department of Clinical Cancer Prevention, Houston, Texas, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, Houston, Texas, USA
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
85
|
Luo B, Zhang Y, Zhang C, Liu X, Shi C. Intestinal microbiota: A potential target for enhancing the antitumor efficacy and reducing the toxicity of immune checkpoint inhibitors. Cancer Lett 2021; 509:53-62. [PMID: 33845122 DOI: 10.1016/j.canlet.2021.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/06/2021] [Accepted: 04/05/2021] [Indexed: 12/14/2022]
Abstract
Accumulating evidence suggests that the intestinal microbiota is associated with the antitumor efficacy of immune checkpoint inhibitors (ICIs) and the occurrence of immune-related adverse events (irAEs) following ICI treatment. However, the mechanisms underlying these interactions remain unclear. Recent technological advances have allowed more extensive investigation into the interplay between the intestinal microbiota and the tumor immune microenvironment. Breakthroughs by two research groups revealed that Bifidobacterium enhanced the efficacy of ICIs via the stimulator of interferon genes (STING) and adenosine 2A receptor (A2AR) signaling pathways, highlighting the molecular mechanisms through which the intestinal microbiota modulates immunotherapy. In this review, we summarize recent findings related to the potential role and mechanisms of the gut microbiota in ICI therapy, available microbiota-targeting strategies, and ongoing clinical trials. Further we discuss the associated challenges that remain in this field of research. The current review aims to evaluate the potential of the intestinal microbiota in maximizing the antitumor efficacy of ICIs while minimizing their toxic effects and guiding the development of more specific treatment regimens.
Collapse
Affiliation(s)
- Baohua Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yongbin Zhang
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiaoqiu Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
86
|
Ochi N, Ichihara E, Takigawa N, Harada D, Inoue K, Shibayama T, Hosokawa S, Kishino D, Harita S, Oda N, Hara N, Hotta K, Maeda Y, Kiura K. The effects of antibiotics on the efficacy of immune checkpoint inhibitors in patients with non-small-cell lung cancer differ based on PD-L1 expression. Eur J Cancer 2021; 149:73-81. [PMID: 33838391 DOI: 10.1016/j.ejca.2021.02.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/22/2021] [Accepted: 02/24/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are essential for treatment of various malignancies, including non-small-cell lung cancer (NSCLC). Recently, several studies have shown that the gut microbiome plays an important role in ICI treatment of solid cancers, and antibiotic (ATB) use had a negative impact on the outcomes of ICI treatment via dysbiosis in the gut. However, whether this is applicable to NSCLC remains unclear. The impact of ATBs based on PD-L1 expression also remains unclear. METHODS We retrospectively reviewed the medical records of patients with NSCLC who received ICI monotherapy (anti-PD-1 or anti-PD-L1 antibody) at nine institutions from December 2015 to May 2018. Outcomes with use of ATBs during the 2 months before or a month after initiation of ICI treatment, including progression-free survival (PFS) and overall survival (OS), were investigated using the Kaplan-Meier method. Multivariate analysis was also conducted using a Cox proportional hazards model. RESULTS A total of 531 patients were included in this study, among whom 98 (18.5%) received ATBs before or after ICI treatment. ATB use was significantly associated with a shorter median OS (11.7 months in the ATB group vs. 16.1 months in the non-ATB group; p = 0.028), whereas the difference in PFS was not significant (3.5 months in both the groups; p = 0.287). We next investigated the association based on PD-L1 expression in the 265 patients for whom PD-L1 expression was determined. There was no significant difference in the median OS or PFS between patients with NSCLC and PD-L1 expression <50% receiving ATBs and those not receiving ATBs (PFS: 3.3 vs. 2.8 months, p = 0.88; OS: 9.5 vs. 17.1 months, p = 0.24). Conversely, patients with NSCLC and PD-L1 expression ≥50% receiving ATBs showed significantly shorter median PFS and OS (PFS: 4.2 vs. 9.4 months, p = 0.012; OS: 11.9 vs. 28.4 months, p = 0.011). The impact of ATBs in patients with NSCLC and PD-L1 expression ≥50% was more significant than that in the entire cohort. CONCLUSIONS Our results indicate that the impact of ATB use on the efficacy of ICIs differed based on PD-L1 expression in patients with advanced NSCLC. A negative impact of ATB use was found in patients with NSCLC and PD-L1 expression ≥50% but not in those with PD-L1 expression <50%.
Collapse
Affiliation(s)
- Nobuaki Ochi
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Eiki Ichihara
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Japan.
| | - Nagio Takigawa
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Daijiro Harada
- Department of Thoracic Oncology, National Hospital Organization Shikoku Cancer Center, Japan
| | - Koji Inoue
- Department of Respiratory Medicine, Ehime Prefectural Central Hospital, Japan
| | - Takuo Shibayama
- Department of Respiratory Medicine, National Hospital Organization Okayama Medical Center, Japan
| | - Shinobu Hosokawa
- Department of Respiratory Medicine, Japanese Red Cross Okayama Hospital, Japan
| | - Daizo Kishino
- Department of Respiratory Medicine, Himeji Red Cross Hospital, Japan
| | - Shingo Harita
- Department of Internal Medicine, Okayama Saiseikai General Hospital, Japan
| | - Naohiro Oda
- Department of Internal Medicine, Fukuyama City Hospital, Japan
| | - Naofumi Hara
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Japan
| | - Katsuyuki Hotta
- Center for Innovative Clinical Medicine, Okayama University Hospital, Japan
| | - Yoshinobu Maeda
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Japan
| | - Katsuyuki Kiura
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Japan
| |
Collapse
|
87
|
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; 106:142-154. [PMID: 33771672 DOI: 10.1016/j.ijid.2021.03.063] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Observational and experimental studies suggest that the use of antibiotics close to administration of immune checkpoint inhibitors (ICI) can have a negative effect on tumour response and patient survival, due to microbiome dysbiosis and the resultant suppression of host immune response against neoplastic cells. METHODS A systematic search of PUBMED and EMBASE was undertaken for studies published between 1 January 2017 and 1 June 2020, evaluating the association between the use of antibiotics and clinical outcomes in patients with cancer treated with ICIs. A meta-analysis of the association between the use of antibiotics and clinical outcomes was also performed. RESULTS Forty-eight studies met the inclusion criteria (12,794 patients). Use of antibiotics was associated with shorter overall survival [hazard ratio (HR) 1.88, 95% confidence interval (CI) 1.59-2.22; adjusted HR 1.87, 95% CI 1.55-2.25] and progression-free survival (HR 1.52, 95% CI 1.36-1.70; adjusted HR 1.93, 95% CI 1.59-2.36), decreased response rate [odds ratio (OR) 0.54, 95% CI 0.34-0.86] and more disease progression (OR 2.00, 95% CI 1.27-3.14). The negative association between the use of antibiotics and progression-free survival was stronger in patients with renal cell carcinoma or melanoma compared with lung cancer. Only antibiotic administration >1 month prior to ICI initiation was associated with increased disease progression. Heterogeneity was substantial for all outcomes. CONCLUSIONS Recent use of antibiotics in patients with cancer treated with ICIs was associated with worse clinical outcomes. Such patients may benefit from dedicated antimicrobial stewardship programmes.
Collapse
Affiliation(s)
- Maria Tsikala-Vafea
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Neel Belani
- Division of Hematology-Oncology, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Kendra Vieira
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Hina Khan
- Department of Internal Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Dimitrios Farmakiotis
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
88
|
Huang C, Li M, Liu B, Zhu H, Dai Q, Fan X, Mehta K, Huang C, Neupane P, Wang F, Sun W, Umar S, Zhong C, Zhang J. Relating Gut Microbiome and Its Modulating Factors to Immunotherapy in Solid Tumors: A Systematic Review. Front Oncol 2021; 11:642110. [PMID: 33816289 PMCID: PMC8012896 DOI: 10.3389/fonc.2021.642110] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Gut microbiome is proved to affect the activity of immunotherapy in certain tumors. However, little is known if there is universal impact on both the treatment response and adverse effects (AEs) of immune checkpoint inhibitors (ICIs) across multiple solid tumors, and whether such impact can be modulated by common gut microbiome modifiers, such as antibiotics and diet. Methods: A systematic search in PubMed followed by stringent manual review were performed to identify clinical cohort studies that evaluated the relevance of gut microbiome to ICIs (response and/or AEs, 12 studies), or association of antibiotics with ICIs (17 studies), or impact of diet on gut microbiome (16 studies). Only original studies published in English before April 1st, 2020 were used. Qualified studies identified in the reference were also included. Results: At the phylum level, patients who had enriched abundance in Firmicutes and Verrucomicrobia almost universally had better response from ICIs, whereas those who were enriched in Proteobacteria universally presented with unfavorable outcome. Mixed correlations were observed for Bacteroidetes in relating to treatment response. Regarding the AEs, Firmicutes correlated to higher incidence whereas Bacteroidetes were clearly associated with less occurrence. Interestingly, across various solid tumors, majority of the studies suggested a negative association of antibiotic use with clinical response from ICIs, especially within 1-2 month prior to the initiation of ICIs. Finally, we observed a significant correlation of plant-based diet in relating to the enrichment of “ICI-favoring” gut microbiome (P = 0.0476). Conclusions: Gut microbiome may serve as a novel modifiable biomarker for both the treatment response and AEs of ICIs across various solid tumors. Further study is needed to understand the underlying mechanism, minimize the negative impact of antibiotics on ICIs, and gain insight regarding the role of diet so that this important lifestyle factor can be harnessed to improve the therapeutic outcomes of cancer immunotherapy partly through its impact on gut microbiome.
Collapse
Affiliation(s)
- Chengliang Huang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Meizhang Li
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ben Liu
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, United States
| | - Huanbo Zhu
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States.,Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Xianming Fan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kathan Mehta
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Chao Huang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Prakash Neupane
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Fen Wang
- Department of Radiation Oncology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Weijing Sun
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Shahid Umar
- Department of Surgery, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States.,Department of Cancer Biology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, United States
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States.,Department of Cancer Biology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
89
|
Ma J, Zhu W, Liu B. Role of gut microbiome in the outcome of cancer immunotherapy. Int J Cancer 2021; 149:760-768. [PMID: 33600603 DOI: 10.1002/ijc.33524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/20/2020] [Accepted: 01/27/2021] [Indexed: 12/11/2022]
Abstract
Nearly 3 × 1013 types of bacteria colonize the human intestine. These colonized bacteria help in maintaining intestinal homeostasis by establishing a complex relationship with the intestinal epithelium and lymphoid tissue. Alteration in the composition of the intestinal microbiota is associated with susceptibility to various pathological conditions, such as autoimmune disorders, diabetes, inflammation and cancer. Of late, several researchers have focused on examining the effects of gut microbiota on the outcome of various cancer treatment protocols. Side effects and complications of traditional chemotherapy and allogeneic hematopoietic cell transplantation are associated with intestinal dysbiosis. Gut microbiota affects the efficacy of immune checkpoint inhibitor-based immunotherapy. The gut is inhabited by diverse resident bacteria, of which, few enhance, while others inhibit the host response to immunotherapy. This review focuses on the correlation between intestinal microbiota and the outcome of tumor immunotherapy. Additionally, the molecular mechanisms underlying the effects of gut microbiota on the efficacy of cancer immunotherapy have been reviewed. Further studies are needed for the identification of distinct gut microbiota and their efficacy in tumor immunotherapy as certain types of intestinal bacteria could function as novel adjuvant drugs to enhance the effectiveness of antitumor therapy in humans.
Collapse
Affiliation(s)
- Junting Ma
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, China
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Wenwen Zhu
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Beixing Liu
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
90
|
Oliva M, Mulet-Margalef N, Ochoa-De-Olza M, Napoli S, Mas J, Laquente B, Alemany L, Duell EJ, Nuciforo P, Moreno V. Tumor-Associated Microbiome: Where Do We Stand? Int J Mol Sci 2021; 22:1446. [PMID: 33535583 PMCID: PMC7867144 DOI: 10.3390/ijms22031446] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
The study of the human microbiome in oncology is a growing and rapidly evolving field. In the past few years, there has been an exponential increase in the number of studies investigating associations of microbiome and cancer, from oncogenesis and cancer progression to resistance or sensitivity to specific anticancer therapies. The gut microbiome is now known to play a significant role in antitumor immune responses and in predicting the efficacy of immune-checkpoint inhibitors in cancer patients. Beyond the gut, the tumor-associated microbiome-microbe communities located either in the tumor or within its body compartment-seems to interact with the local microenvironment and the tumor immune contexture, ultimately impacting cancer progression and treatment outcome. However, pre-clinical research focusing on causality and mechanistic pathways as well as proof-of-concept studies are still needed to fully understand the potential clinical utility of microbiome in cancer patients. Moreover, there is a need for the standardization of methodology and the implementation of quality control across microbiome studies to allow for a better interpretation and greater comparability of the results reported between them. This review summarizes the accumulating evidence in the field and discusses the current and upcoming challenges of microbiome studies.
Collapse
Affiliation(s)
- Marc Oliva
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Nuria Mulet-Margalef
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Maria Ochoa-De-Olza
- Service of Immuno-Oncology, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland;
- Ludwig Institute for Cancer Research, University of Lausanne, 1066 Lausanne, Switzerland
| | - Stefania Napoli
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (S.N.); (P.N.)
| | - Joan Mas
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| | - Berta Laquente
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Laia Alemany
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
- Cancer Epidemiology Research Program, Catalan Institute of Oncology, L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- EPIBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
| | - Eric J. Duell
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (S.N.); (P.N.)
| | - Victor Moreno
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| |
Collapse
|
91
|
Yu Y, Zheng P, Gao L, Li H, Tao P, Wang D, Ding F, Shi Q, Chen H. Effects of Antibiotic Use on Outcomes in Cancer Patients Treated Using Immune Checkpoint Inhibitors: A Systematic Review and Meta-Analysis. J Immunother 2021; 44:76-85. [PMID: 33208635 DOI: 10.1097/cji.0000000000000346] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
Abstract
Antibiotic (ATB) use seems to negatively affect the outcomes of immune checkpoint inhibitors (ICIs). The aim of this review is to clarify whether ATB use influences the efficacy of ICI treatment in cancer patients. Databases of MEDLINE, Embase, and Cochrane Library were searched for reports published in English between January 2007 and December 2019. We included studies that compared the outcomes of ATB use and no-ATB use in cancer patients using ICIs. Two reviewers independently selected eligible studies and extracted the data. Meta-analysis was performed with pooling of unadjusted hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS), and with pooling of odds ratios (ORs) for objective response rate (ORR). Thirty-eight studies involving 8409 patients were finally included for qualitative or quantitative analyses. Cancer types included renal cell carcinoma, non-small cell lung cancer, urothelial carcinoma, melanoma, gastrointestinal cancer, and others. Meta-analyses revealed that ATB use was associated with poor OS [HR: 1.80, 95% confidence interval (CI): 1.44-2.26, P<0.001], PFS (HR: 1.55, 95% CI: 1.26-1.91, P<0.001) and ORR (OR: 0.63, 95% CI: 0.42-0.95, P=0.03). Subgroup analysis found that these relationships were not influenced by cancer type or ICI regimens, but were dependent on the timing of ATB use. Narrative results of multivariable analyses further confirmed the negative effects of ATB use on OS and PFS. In cancer patients using ICIs, pre-ATB use close to the start of ICI treatment (within 60 d) was detrimental to outcomes in terms of OS, PFS, and ORR.
Collapse
Affiliation(s)
- Yang Yu
- Department of Tumor Surgery, Lanzhou University Second Hospital
- Second Clinical Medical College
| | - Peng Zheng
- Department of Tumor Surgery, Lanzhou University Second Hospital
- Second Clinical Medical College
| | - Lei Gao
- Department of Tumor Surgery, Lanzhou University Second Hospital
- Second Clinical Medical College
| | - Haiyuan Li
- Department of Tumor Surgery, Lanzhou University Second Hospital
- Second Clinical Medical College
| | - Pengxian Tao
- Department of Tumor Surgery, Lanzhou University Second Hospital
- Second Clinical Medical College
| | - Dengfeng Wang
- Department of Tumor Surgery, Lanzhou University Second Hospital
- Second Clinical Medical College
| | - Fanghui Ding
- First Clinical Medical College, Lanzhou University
- General Surgery Department Ward 5, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qianling Shi
- First Clinical Medical College, Lanzhou University
| | - Hao Chen
- Department of Tumor Surgery, Lanzhou University Second Hospital
| |
Collapse
|
92
|
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; 92:107303. [PMID: 33465728 DOI: 10.1016/j.intimp.2020.107303] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Pre-clinical and clinical data had revealed the gut microbiome plays a critical role in immune checkpoint inhibitors (ICIs) efficacy. This study was designed to investigate whether antibiotics (ATBs) affect the prognosis of malignancies treated with ICIs. METHODS Electronic databases were searched to identify relevant trials that evaluated the impact of ATBs on ICIs efficacy. The primary endpoints were overall survival (OS) and progression-free survival (PFS) measured by HRs with corresponding 95%CIs. Subgroup analyses were performed based on cancer type, study design, ICIs agent, and time of ATBs administration. RESULTS Totally, 12,492 individuals in the 44 cohorts were recruited. Pooled results showed that ATBs administration was significantly correlated with a worse objective remission rate (ORR) (OR = 0.61, 95%CI (0.42-0.90), P = 0.0128), PFS (HR = 1.18, 95%CI (1.11-1.25), P < 0.0001), and OS (HR = 1.20, 95%CI (1.15-1.25), P < 0.0001) in patients treated with ICIs. In subgroup analyses, patients treated with ICIs exposed to ATBs suffered an evidently worse ORR in arms of renal cell carcinoma (RCC) (OR = 0.30, 95%CI (0.14-0.67), P = 0.0034), multiple (OR = 0.44, 95%CI (0.27-0.73), P = 0.0016), and before ICIs initiation (OR = 0.47, 95%CI (0.32-0.71), P = 0.0003) without heterogeneity; experienced a worse PFS and OS in arms of non-small cell lung cancer, melanoma, RCC, urothelial carcinoma, multiple, prospective, retrospective, PD-(L)1 alone, PD-(L)1 plus CTLA-4, before ICIs initiation, before ICIs initiation and concurrent, and before or after ICIs within 1 month, while a better PFS and OS in concurrent with ICIs arm. CONCLUSIONS ATBs administration was negatively associated with ORR, PFS and OS in malignancies treated with ICIs, while the time of ATBs exposure might impact ICIs efficacy.
Collapse
Affiliation(s)
- Qing Wu
- Department of Oncology, Molecular Oncology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Junjin Liu
- Department of Infectious Diseases, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Sumei Wu
- Department of Oncology, Molecular Oncology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Xianhe Xie
- Department of Oncology, Molecular Oncology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China.
| |
Collapse
|
93
|
The biomarkers related to immune related adverse events caused by immune checkpoint inhibitors. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:284. [PMID: 33317597 PMCID: PMC7734811 DOI: 10.1186/s13046-020-01749-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023]
Abstract
The enthusiasm for immune checkpoint inhibitors (ICIs), an efficient tumor treatment model different from traditional treatment, is based on their unprecedented antitumor effect, but the occurrence of immune-related adverse events (irAEs) is an obstacle to the prospect of ICI treatment. IrAEs are a discrete toxicity caused by the nonspecific activation of the immune system and can affect almost all tissues and organs. Currently, research on biomarkers mainly focuses on the gastrointestinal tract, endocrine system, skin and lung. Several potential hypotheses concentrate on the overactivation of the immune system, excessive release of inflammatory cytokines, elevated levels of pre-existing autoantibodies, and presence of common antigens between tumors and normal tissues. This review lists the current biomarkers that might predict irAEs and their possible mechanisms for both nonspecific and organ-specific biomarkers. However, the prediction of irAEs remains a major clinical challenge to screen and identify patients who are susceptible to irAEs and likely to benefit from ICIs.
Collapse
|
94
|
Fidelle M, Yonekura S, Picard M, Cogdill A, Hollebecque A, Roberti MP, Zitvogel L. Resolving the Paradox of Colon Cancer Through the Integration of Genetics, Immunology, and the Microbiota. Front Immunol 2020; 11:600886. [PMID: 33381121 PMCID: PMC7768083 DOI: 10.3389/fimmu.2020.600886] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
While colorectal cancers (CRC) are paradigmatic tumors invaded by effector memory lymphocytes, the mechanisms accounting for the relative resistance of MSI negative CRC to immunogenic cell death mediated by oxaliplatin and immune checkpoint inhibitors has remained an open conundrum. Here, we propose the viewpoint where its microenvironmental contexture could be explained -at least in part- by macroenvironmental cues constituted by the complex interplay between the epithelial barrier, its microbial ecosystem, and the local immune system. Taken together this dynamic ménage-à-trois offers novel coordinated actors of the humoral and cellular immune responses actionable to restore sensitivity to immune checkpoint inhibition. Solving this paradox involves breaking tolerance to crypt stem cells by inducing the immunogenic apoptosis of ileal cells in the context of an ileal microbiome shifted towards immunogenic bacteria using cytotoxicants. This manoeuver results in the elicitation of a productive Tfh and B cell dialogue in mesenteric lymph nodes culminating in tumor-specific memory CD8+ T cell responses sparing the normal epithelium.
Collapse
Affiliation(s)
- Marine Fidelle
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Satoru Yonekura
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Marion Picard
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Unit Biology and Genetics of the Bacterial Cell Wall, Institut Pasteur, Paris, France
| | - Alexandria Cogdill
- Department of Immunology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
- Department of Genomic Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Antoine Hollebecque
- Gustave Roussy, Villejuif, France
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Maria Paula Roberti
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| |
Collapse
|
95
|
Karpinets TV, Solley TN, Mikkelson MD, Dorta-Estremera S, Nookala SS, Medrano AYD, Petrosino JF, Mezzari MP, Zhang J, Futreal PA, Sastry KJ, Colbert LE, Klopp A. Effect of Antibiotics on Gut and Vaginal Microbiomes Associated with Cervical Cancer Development in Mice. Cancer Prev Res (Phila) 2020; 13:997-1006. [PMID: 32917644 PMCID: PMC10583130 DOI: 10.1158/1940-6207.capr-20-0103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/06/2020] [Accepted: 08/25/2020] [Indexed: 11/16/2022]
Abstract
Antibiotics affect microbial diversity in the gut, leading to dysbiosis and impaired immunity. However, the impact of antibiotics on microbial communities at other sites, such as vagina is less understood. It is also not clear whether changes induced by antibiotics in both microbiomes affect the development of cervical cancer. In this study, we utilized the murine model to evaluate these questions. We show that oral application of broad-spectrum antibiotics in mice changed not only diversity, but composition and sharing of gut and vaginal microbiomes in mice and influenced cervical cancer development in an orthotopic tumor model. Antibiotics decreased richness and diversity indexes in the gut but increased them in the vagina. Some beneficial taxa, such as Bacteroides, Ruminococcaceae, and Lachnospiraceae increased their abundance in the vagina while other pathogenic species, such as Proteobacteria, were decreased. As a result of the changes, mice with greater richness and diversity of the vaginal microbiome after antibiotics exposure were less likely developed tumors. No association between richness and diversity of the gut microbiome and tumor development was identified.
Collapse
Affiliation(s)
| | - Travis N Solley
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Megan D Mikkelson
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | - Sita S Nookala
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas
| | - Melissa P Mezzari
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas
| | - Jinghua Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas
| | - P Andrew Futreal
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas
| | - K Jagannadha Sastry
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Lauren E Colbert
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas.
| | - Ann Klopp
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
96
|
Cren PY, Bertrand N, Le Deley MC, Génin M, Mortier L, Odou P, Penel N, Chazard E. Is the survival of patients treated with ipilimumab affected by antibiotics? An analysis of 1585 patients from the French National hospital discharge summary database (PMSI). Oncoimmunology 2020; 9:1846914. [PMID: 33299658 PMCID: PMC7714497 DOI: 10.1080/2162402x.2020.1846914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: The gut microbiota has a key role in the regulation of the immune system. Disruption of the gut microbiota’s composition by antibiotics might significantly affect the efficacy of immune checkpoint inhibitors. In a study of patients treated with ipilimumab, we sought to assess the relationship between overall survival and in-hospital antibiotic administration. Methods: Patients having been treated with ipilimumab between January 2012 and November 2014 were selected from the French National Hospital Discharge Summary Database. Exposure to antibiotics was defined as the presence of a hospital stay with a documented systemic bacterial infection in the 2 months before or the month after initiation of the patient’s first ever course of ipilimumab. The primary outcome was overall survival. Results: We studied 43,124 hospital stays involving 1585 patients from 97 centers. All patients had received ipilimumab monotherapy for advanced melanoma. Overall, 117 of the 1585 patients (7.4%) were documented as having received systemic antibiotic therapy in hospital during the defined exposure period. The median overall survival time was shorter in patients with infection (6.3 months, vs. 15.4 months in patients without an infection; hazard ratio (HR) = 1.88, 95% confidence interval [1.46; 2.43], p = 10−6). In a multivariate analysis adjusted for covariates, infection was still significantly associated with overall survival (HR = 1.68, [1.30; 2.18], p = 10−5). Conclusions: In patients treated with ipilimumab for advanced melanoma, infection, and antibiotic administration in hospital at around the time of the patient’s first ever course of ipilimumab appears to be associated with significantly lower clinical benefit.
Collapse
Affiliation(s)
- Pierre-Yves Cren
- CERIM, ULR 2694 METRICS, Univ. Lille, CHU Lille, Lille, France.,Methodology and Biostatistics Unit, Centre Oscar Lambret, Lille, France
| | | | - Marie-Cécile Le Deley
- Methodology and Biostatistics Unit, Centre Oscar Lambret, Lille, France.,CESP, INSERM, Paris-Saclay University, Paris-Sud University, UVSQ, Villejuif, France
| | - Michaël Génin
- CERIM, ULR 2694 METRICS, Univ. Lille, CHU Lille, Lille, France
| | - Laurent Mortier
- Clinique de Dermatologie, Unit#xE9; d#x27;Onco-Dermatologie, U1189, INSERM, Univ. Lille, CHU Lille, Lille, France
| | - Pascal Odou
- ULR 7365 GRITA, Univ. Lille, CHU Lille, Lille, France
| | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret and Univ. Lille, Lille, France
| | | |
Collapse
|
97
|
Atypical immunometabolism and metabolic reprogramming in liver cancer: Deciphering the role of gut microbiome. Adv Cancer Res 2020; 149:171-255. [PMID: 33579424 DOI: 10.1016/bs.acr.2020.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related mortality worldwide. Much recent research has delved into understanding the underlying molecular mechanisms of HCC pathogenesis, which has revealed to be heterogenous and complex. Two major hallmarks of HCC include: (i) a hijacked immunometabolism and (ii) a reprogramming in metabolic processes. We posit that the gut microbiota is a third component in an entanglement triangle contributing to HCC progression. Besides metagenomic studies highlighting the diagnostic potential in the gut microbiota profile, recent research is pinpointing the gut microbiota as an instigator, not just a mere bystander, in HCC. In this chapter, we discuss mechanistic insights on atypical immunometabolism and metabolic reprogramming in HCC, including the examination of tumor-associated macrophages and neutrophils, tumor-infiltrating lymphocytes (e.g., T-cell exhaustion, regulatory T-cells, natural killer T-cells), the Warburg effect, rewiring of the tricarboxylic acid cycle, and glutamine addiction. We further discuss the potential involvement of the gut microbiota in these characteristics of hepatocarcinogenesis. An immediate highlight is that microbiota metabolites (e.g., short chain fatty acids, secondary bile acids) can impair anti-tumor responses, which aggravates HCC. Lastly, we describe the rising 'new era' of immunotherapies (e.g., immune checkpoint inhibitors, adoptive T-cell transfer) and discuss for the potential incorporation of gut microbiota targeted therapeutics (e.g., probiotics, fecal microbiota transplantation) to alleviate HCC. Altogether, this chapter invigorates for continuous research to decipher the role of gut microbiome in HCC from its influence on immunometabolism and metabolic reprogramming.
Collapse
|
98
|
Multivariable Analysis of 169 Cases of Advanced Cutaneous Melanoma to Evaluate Antibiotic Exposure as Predictor of Survival to Anti-PD-1 Based Immunotherapies. Antibiotics (Basel) 2020; 9:antibiotics9110740. [PMID: 33120998 PMCID: PMC7692514 DOI: 10.3390/antibiotics9110740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 11/17/2022] Open
Abstract
Recently antibiotic exposure has been associated with worse outcomes in patients undergoing treatment with antibodies directed against programmed cell death protein-1 (PD-1). We reviewed data of 1264 patients enrolled at Melanoma Skin and Ocular Tissue Repositories at University of Iowa Hospitals and Clinic. Reviewed data included patient demographics, prior medical history, baseline hematologic and disease parameters and outcomes including progression-free survival (PFS) and overall survival (OS). Cox regression models were used to determine predictive markers. Overall, 169 patients with advanced cutaneous melanoma received anti-PD-1 based therapies. Median follow up was 18.46 (range 0.89 to 62.52) months. On multivariable analysis brain metastasis, higher absolute neutrophil count (ANC) and lower absolute lymphocyte count were associated with poorer PFS while brain and liver metastasis and lower albumin were associated with poorer OS. Prior antibiotics, radiation as well as age, gender, basal metabolic index (BMI), smoking status, BRAF mutation, line of therapy (first or latter), prior treatments (ipilimumab or BRAF inhibitors), hemoglobin, neutrophil-to-lymphocyte ratio, white blood cell, platelet and eosinophil counts were not associated with PFS or OS in multivariable analysis. Contrary to some prior studies BMI, radiation, and antibiotics were not associated with PFS or OS.
Collapse
|
99
|
Wang J, Yang HR, Wang DJ, Wang XX. Association between the gut microbiota and patient responses to cancer immune checkpoint inhibitors. Oncol Lett 2020; 20:342. [PMID: 33123253 PMCID: PMC7583737 DOI: 10.3892/ol.2020.12205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Studies are increasingly investigating the association between the gut microbiota and the outcomes of immunotherapy in patients with cancer. Notably, certain studies have demonstrated that the gut microbiota serves a key role in regulating a patient's response to immunotherapy. In the present review, the potential associations between the gut microbiota, and cancer, host immunity and cancer immunotherapy are reviewed. Furthermore, the effects of fecal microbiota transplantation, antibiotics, probiotics, prebiotics, synbiotics, components of traditional Chinese medicine and various lifestyle factors on the gut microbiota and cancer immunotherapy outcomes are discussed. Certain dominant bacterial groups in the context of cancer immunotherapy and certain effective methods for optimizing immunotherapy by regulating the gut microbiota have been identified. Further investigation may enable the rapid conversion of these discoveries into practical products and clinically applicable methods.
Collapse
Affiliation(s)
- Jian Wang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Hong-Ru Yang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Dai-Jie Wang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Xing-Xia Wang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
100
|
Aminzadeh-Gohari S, Weber DD, Catalano L, Feichtinger RG, Kofler B, Lang R. Targeting Mitochondria in Melanoma. Biomolecules 2020; 10:biom10101395. [PMID: 33007949 PMCID: PMC7599575 DOI: 10.3390/biom10101395] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Drastically elevated glycolytic activity is a prominent metabolic feature of cancer cells. Until recently it was thought that tumor cells shift their entire energy production from oxidative phosphorylation (OXPHOS) to glycolysis. However, new evidence indicates that many cancer cells still have functional OXPHOS, despite their increased reliance on glycolysis. Growing pre-clinical and clinical evidence suggests that targeting mitochondrial metabolism has anti-cancer effects. Here, we analyzed mitochondrial respiration and the amount and activity of OXPHOS complexes in four melanoma cell lines and normal human dermal fibroblasts (HDFs) by Seahorse real-time cell metabolic analysis, immunoblotting, and spectrophotometry. We also tested three clinically approved antibiotics, one anti-parasitic drug (pyrvinium pamoate), and a novel anti-cancer agent (ONC212) for effects on mitochondrial respiration and proliferation of melanoma cells and HDFs. We found that three of the four melanoma cell lines have elevated glycolysis as well as OXPHOS, but contain dysfunctional mitochondria. The antibiotics produced different effects on the melanoma cells and HDFs. The anti-parasitic drug strongly inhibited respiration and proliferation of both the melanoma cells and HDFs. ONC212 reduced respiration in melanoma cells and HDFs, and inhibited the proliferation of melanoma cells. Our findings highlight ONC212 as a promising drug for targeting mitochondrial respiration in cancer.
Collapse
Affiliation(s)
- Sepideh Aminzadeh-Gohari
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
| | - Daniela D. Weber
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
| | - Luca Catalano
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
| | - René G. Feichtinger
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
- Correspondence: (B.K.); (R.L.); Tel.: +43-57255-26274 (B.K.); +43-57255-58200 (R.L.)
| | - Roland Lang
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
- Correspondence: (B.K.); (R.L.); Tel.: +43-57255-26274 (B.K.); +43-57255-58200 (R.L.)
| |
Collapse
|