1
|
Yan J, Yang L, Ren Q, Zhu C, Du H, Wang Z, Qi Y, Xian X, Chen D. Gut microbiota as a biomarker and modulator of anti-tumor immunotherapy outcomes. Front Immunol 2024; 15:1471273. [PMID: 39669573 PMCID: PMC11634861 DOI: 10.3389/fimmu.2024.1471273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/25/2024] [Indexed: 12/14/2024] Open
Abstract
Although immune-checkpoint inhibitors (ICIs) have significantly improved cancer treatment, their effectiveness is limited by primary or acquired resistance in many patients. The gut microbiota, through its production of metabolites and regulation of immune cell functions, plays a vital role in maintaining immune balance and influencing the response to cancer immunotherapies. This review highlights evidence linking specific gut microbial characteristics to increased therapeutic efficacy in a variety of cancers, such as gastrointestinal cancers, melanoma, lung cancer, urinary system cancers, and reproductive system cancers, suggesting the gut microbiota's potential as a predictive biomarker for ICI responsiveness. It also explores the possibility of enhancing ICI effectiveness through fecal microbiota transplantation, probiotics, prebiotics, synbiotics, postbiotics, and dietary modifications. Moreover, the review underscores the need for extensive randomized controlled trials to confirm the gut microbiota's predictive value and to establish guidelines for microbiota-targeted interventions in immunotherapy. In summary, the article suggests that a balanced gut microbiota is key to maximizing immunotherapy benefits and calls for further research to optimize microbiota modulation strategies for cancer treatment. It advocates for a deeper comprehension of the complex interactions between gut microbiota, host immunity, and cancer therapy, aiming for more personalized and effective treatment options.
Collapse
Affiliation(s)
- Jiexi Yan
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lu Yang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chan Zhu
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Haiyun Du
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhouyu Wang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Yaya Qi
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaohong Xian
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Dongsheng Chen
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Du C, Zhao Y, Shen F, Qian H. Effect of Brassica rapa L. Polysaccharide on Lewis Lung Cancer Mice by Inflammatory Regulation and Gut Microbiota Modulation. Foods 2024; 13:3704. [PMID: 39594117 PMCID: PMC11593872 DOI: 10.3390/foods13223704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related fatalities globally, related to inflammatory and gut microbiota imbalance. Brassica rapa L. polysaccharide (BP) is a functional compound, which is utilized by the gut microbiota to regulate immunity and metabolism. However, the effect of BP on lung cancer and whether it affects the "gut-lung" axis remains unclear. This study explored the intervention of BP in Lewis lung cancer (LLC) mice and its effect on the gut microbiota. The results revealed that BP reduced tumor weight and downregulated the expression of Ki67 protein. Additionally, BP reduced the content of inflammatory factors and growth factors, promoting tumor cell apoptosis and inhibiting the growth of LLC. The intervention of BP suppressed intestinal inflammation, preserved intestinal barrier integrity, and augmented the level of beneficial microbiota, such as Blautia and Bifidobacterium. Furthermore, BP significantly increased the production of short-chain fatty acids (SCFAs), particularly butyrate and propionate. A correlation analysis showed significant correlations among the gut microbiota, SCFAs, inflammatory factors, and tight junction proteins. A functional analysis indicated that BP promoted amino acid metabolism and fatty acid metabolism. These findings suggested that BP had the potential to act as prebiotics to prevent disease and improve lung cancer progression by regulating the gut microbiota.
Collapse
Affiliation(s)
- Changhui Du
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Yong Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China;
| | - Fanglin Shen
- School of Environmental Engineering, Wuxi University, Wuxi 214105, China;
| | - He Qian
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| |
Collapse
|
3
|
Bao P, Zhang XZ. Progress of tumor-resident intracellular bacteria for cancer therapy. Adv Drug Deliv Rev 2024; 214:115458. [PMID: 39383997 DOI: 10.1016/j.addr.2024.115458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/12/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Emerging studies have disclosed the pivotal role of cancer-associated microbiota in supporting cancer development, progression and dissemination, with the in-depth comprehending of tumor microenvironment. In particular, certain invasive bacteria that hide in various cells within the tumor tissues can render assistance to tumor growth and invasion through intricate mechanisms implicated in multiple branches of cancer biology. Thus, tumor-resident intracellular microbes are anticipated as next-generation targets for oncotherapy. This review is intended to delve into these internalized bacteria-driven cancer-promoting mechanisms and explore diversified antimicrobial therapeutic strategies to counteract the detrimental impact caused by these intruders, thereby improving therapeutic benefit of antineoplastic therapy.
Collapse
Affiliation(s)
- Peng Bao
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital, Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital, Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| |
Collapse
|
4
|
Iwan E, Grenda A, Bomba A, Bielińska K, Wasyl D, Kieszko R, Rolska-Kopińska A, Chmielewska I, Krawczyk P, Rybczyńska-Tkaczyk K, Olejnik M, Milanowski J. Gut resistome of NSCLC patients treated with immunotherapy. Front Genet 2024; 15:1378900. [PMID: 39170692 PMCID: PMC11335565 DOI: 10.3389/fgene.2024.1378900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
Background The newest method of treatment for patients with NSCLC (non-small cell lung cancer) is immunotherapy directed at the immune checkpoints PD-1 (Programmed Cell Death 1) and PD-L1 (Programmed Cell Death Ligand 1). PD-L1 is the only validated predictor factor for immunotherapy efficacy, but it is imperfect. Some patients do not benefit from immunotherapy and may develop primary or secondary resistance. This study aimed to assess the intestinal resistome composition of non-small cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors in the context of clinical features and potentially new prediction factors for assessing immunotherapy efficacy. Methods The study included 30 advanced NSCLC patients, 19 (57%) men and 11 (33%) women treated with first- or second-line immunotherapy (nivolumab, pembrolizumab or atezolizumab). We evaluated the patient's gut resistome composition using the high sensitivity of targeted metagenomics. Results Studies have shown that resistome richness is associated with clinical and demographic factors of NSCLC patients treated with immunotherapy. Smoking seems to be associated with an increased abundance of macrolides, lincosamides, streptogramins and vancomycin core resistome. The resistome of patients with progression disease appears to be more abundant and diverse, with significantly higher levels of genomic markers of resistance to lincosamides (lnuC). The resistance genes lnuC, msrD, ermG, aph(6), fosA were correlated with progression-free survival or/and overall survival, thus may be considered as factors potentially impacting the disease. Conclusion The results indicate that the intestinal resistome of NSCLC patients with immune checkpoint inhibitors treatment differs depending on the response to immunotherapy, with several distinguished markers. Since it might impact treatment efficacy, it must be examined more deeply.
Collapse
Affiliation(s)
- Ewelina Iwan
- Department of Omics Analyses, National Veterinary Research Institute, Pulawy, Poland
| | - Anna Grenda
- Department of Pneumology, Oncology and Allergology, Medical University in Lublin, Lublin, Poland
| | - Arkadiusz Bomba
- Department of Omics Analyses, National Veterinary Research Institute, Pulawy, Poland
| | - Katarzyna Bielińska
- Department of Omics Analyses, National Veterinary Research Institute, Pulawy, Poland
| | - Dariusz Wasyl
- Department of Omics Analyses, National Veterinary Research Institute, Pulawy, Poland
| | - Robert Kieszko
- Department of Pneumology, Oncology and Allergology, Medical University in Lublin, Lublin, Poland
| | - Anna Rolska-Kopińska
- Department of Pneumology, Oncology and Allergology, Medical University in Lublin, Lublin, Poland
| | - Izabela Chmielewska
- Department of Pneumology, Oncology and Allergology, Medical University in Lublin, Lublin, Poland
| | - Paweł Krawczyk
- Department of Pneumology, Oncology and Allergology, Medical University in Lublin, Lublin, Poland
| | | | - Małgorzata Olejnik
- Department of Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Janusz Milanowski
- Department of Pneumology, Oncology and Allergology, Medical University in Lublin, Lublin, Poland
| |
Collapse
|
5
|
Ji G, Zhao J, Si X, Song W. Targeting bacterial metabolites in tumor for cancer therapy: An alternative approach for targeting tumor-associated bacteria. Adv Drug Deliv Rev 2024; 211:115345. [PMID: 38834140 DOI: 10.1016/j.addr.2024.115345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/11/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Emerging evidence reveal that tumor-associated bacteria (TAB) can facilitate the initiation and progression of multiple types of cancer. Recent work has emphasized the significant role of intestinal microbiota, particularly bacteria, plays in affecting responses to chemo- and immuno-therapies. Hence, it seems feasible to improve cancer treatment outcomes by targeting intestinal bacteria. While considering variable richness of the intestinal microbiota and diverse components among individuals, direct manipulating the gut microbiota is complicated in clinic. Tumor initiation and progression requires the gut microbiota-derived metabolites to contact and reprogram neoplastic cells. Hence, directly targeting tumor-associated bacteria metabolites may have the potential to provide alternative and innovative strategies to bypass the gut microbiota for cancer therapy. As such, there are great opportunities to explore holistic approaches that incorporates TAB-derived metabolites and related metabolic signals modulation for cancer therapy. In this review, we will focus on key opportunistic areas by targeting TAB-derived metabolites and related metabolic signals, but not bacteria itself, for cancer treatment, and elucidate future challenges that need to be addressed in this emerging field.
Collapse
Affiliation(s)
- Guofeng Ji
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jingjing Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China.
| |
Collapse
|
6
|
Ullah H, Arbab S, Tian Y, Chen Y, Liu CQ, Li Q, Li K. Crosstalk between gut microbiota and host immune system and its response to traumatic injury. Front Immunol 2024; 15:1413485. [PMID: 39144142 PMCID: PMC11321976 DOI: 10.3389/fimmu.2024.1413485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/04/2024] [Indexed: 08/16/2024] Open
Abstract
Millions of microorganisms make up the complex microbial ecosystem found in the human gut. The immune system's interaction with the gut microbiota is essential for preventing inflammation and maintaining intestinal homeostasis. Numerous metabolic products that can cross-talk between immune cells and the gut epithelium are metabolized by the gut microbiota. Traumatic injury elicits a great and multifaceted immune response in the minutes after the initial offense, containing simultaneous pro- and anti-inflammatory responses. The development of innovative therapies that improve patient outcomes depends on the gut microbiota and immunological responses to trauma. The altered makeup of gut microbes, or gut dysbiosis, can also dysregulate immunological responses, resulting in inflammation. Major human diseases may become more common as a result of chronic dysbiosis and the translocation of bacteria and the products of their metabolism beyond the mucosal barrier. In this review, we briefly summarize the interactions between the gut microbiota and the immune system and human disease and their therapeutic probiotic formulations. We also discuss the immune response to traumatic injury.
Collapse
Affiliation(s)
- Hanif Ullah
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Safia Arbab
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yali Tian
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Yuwen Chen
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Chang-qing Liu
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Qijie Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Zhu Z, Huang J, Zhang Y, Hou W, Chen F, Mo YY, Zhang Z. Landscape of tumoral ecosystem for enhanced anti-PD-1 immunotherapy by gut Akkermansia muciniphila. Cell Rep 2024; 43:114306. [PMID: 38819989 DOI: 10.1016/j.celrep.2024.114306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/07/2024] [Accepted: 05/15/2024] [Indexed: 06/02/2024] Open
Abstract
Gut Akkermansia muciniphila (Akk) has been implicated in impacting immunotherapy or oncogenesis. This study aims to dissect the Akk-associated tumor immune ecosystem (TIME) by single-cell profiling coupled with T cell receptor (TCR) sequencing. We adopted mouse cancer models under anti-PD-1 immunotherapy, combined with oral administration of three forms of Akk, including live Akk, pasteurized Akk (Akk-past), or its membrane protein Amuc_1100 (Amuc). We show that live Akk is most effective in activation of CD8 T cells by rescuing the exhausted type into cytotoxic subpopulations. Remarkably, only live Akk activates MHC-II-pDC pathways, downregulates CXCL3 in Bgn(+)Dcn(+) cancer-associated fibroblasts (CAFs), blunts crosstalk between Bgn(+)Dcn(+) CAFs and PD-L1(+) neutrophils by a CXCL3-PD-L1 axis, and further suppresses the crosstalk between PD-L1(+) neutrophils and CD8 T cells, leading to the rescue of exhausted CD8 T cells. Together, this comprehensive picture of the tumor ecosystem provides deeper insights into immune mechanisms associated with gut Akk-dependent anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Zhuxian Zhu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jianguo Huang
- Earle A. Chiles Research Institute, a division of Providence Cancer Institute, Portland, OR 97213, USA
| | - Yanling Zhang
- Department of Emergency Medicine, Tongji University School of Medicine, Shanghai 200065, China
| | - Weiwei Hou
- Department of Clinical Laboratory, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Fei Chen
- Department of Emergency Medicine, Tongji University School of Medicine, Shanghai 200065, China
| | - Yin-Yuan Mo
- Institute of Clinical Medicine, Zhejiang Provincial People's Hospital of Hangzhou Medical College, Hangzhou 310014 , China.
| | - Ziqiang Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong Hospital of Fudan University, Shanghai 201399, China.
| |
Collapse
|
8
|
Derosa L, Iebba V, Silva CAC, Piccinno G, Wu G, Lordello L, Routy B, Zhao N, Thelemaque C, Birebent R, Marmorino F, Fidelle M, Messaoudene M, Thomas AM, Zalcman G, Friard S, Mazieres J, Audigier-Valette C, Sibilot DM, Goldwasser F, Scherpereel A, Pegliasco H, Ghiringhelli F, Bouchard N, Sow C, Darik I, Zoppi S, Ly P, Reni A, Daillère R, Deutsch E, Lee KA, Bolte LA, Björk JR, Weersma RK, Barlesi F, Padilha L, Finzel A, Isaksen ML, Escudier B, Albiges L, Planchard D, André F, Cremolini C, Martinez S, Besse B, Zhao L, Segata N, Wojcik J, Kroemer G, Zitvogel L. Custom scoring based on ecological topology of gut microbiota associated with cancer immunotherapy outcome. Cell 2024; 187:3373-3389.e16. [PMID: 38906102 DOI: 10.1016/j.cell.2024.05.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/16/2024] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
The gut microbiota influences the clinical responses of cancer patients to immunecheckpoint inhibitors (ICIs). However, there is no consensus definition of detrimental dysbiosis. Based on metagenomics (MG) sequencing of 245 non-small cell lung cancer (NSCLC) patient feces, we constructed species-level co-abundance networks that were clustered into species-interacting groups (SIGs) correlating with overall survival. Thirty-seven and forty-five MG species (MGSs) were associated with resistance (SIG1) and response (SIG2) to ICIs, respectively. When combined with the quantification of Akkermansia species, this procedure allowed a person-based calculation of a topological score (TOPOSCORE) that was validated in an additional 254 NSCLC patients and in 216 genitourinary cancer patients. Finally, this TOPOSCORE was translated into a 21-bacterial probe set-based qPCR scoring that was validated in a prospective cohort of NSCLC patients as well as in colorectal and melanoma patients. This approach could represent a dynamic diagnosis tool for intestinal dysbiosis to guide personalized microbiota-centered interventions.
Collapse
Affiliation(s)
- Lisa Derosa
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France.
| | - Valerio Iebba
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | | | - Guojun Wu
- Center for Nutrition, Microbiome and Health, New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA; Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA; Rutgers-Jiaotong Joint Laboratory for Microbiome and Human Health, New Brunswick, NJ, USA
| | - Leonardo Lordello
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Bertrand Routy
- Centre Hospitalier de l'Université de Montréal (CHUM), Hematology-Oncology Division, Department of Medicine, Montréal, QC, Canada; Centre de Recherche du CHUM (CRCHUM), Montréal, QC, Canada
| | - Naisi Zhao
- Department of Public Health and Community Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Cassandra Thelemaque
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Roxanne Birebent
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Federica Marmorino
- Unit of Medical Oncology 2, University Hospital of Pisa, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Marine Fidelle
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | | | | | - Gerard Zalcman
- Université Paris Cité, Thoracic Oncology Department-CIC1425/CLIP2 Paris-Nord, Bichat-Claude Bernard Hospital, AP-HP, Paris, France
| | - Sylvie Friard
- Pneumology Department, Foch Hospital, Suresnes, France
| | - Julien Mazieres
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | | | - Denis Moro- Sibilot
- Department of Thoracic Oncology, Centre Hospitalier Universitaire, Grenoble, France
| | - François Goldwasser
- INSERM U1016-CNRS UMR8104, Paris Cité University, Paris, France; Department of Medical Oncology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Paris, France
| | - Arnaud Scherpereel
- Department of Pulmonary and Thoracic Oncology, University of Lille, University Hospital (CHU), Lille, France
| | | | - François Ghiringhelli
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | | | - Cissé Sow
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Ines Darik
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Silvia Zoppi
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Pierre Ly
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Anna Reni
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | | | - Eric Deutsch
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Department of Radiation Oncology, Gustave Roussy, Villejuif, France; INSERM U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Villejuif, France
| | - Karla A Lee
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Laura A Bolte
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Johannes R Björk
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Fabrice Barlesi
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Lucas Padilha
- Bio-Me AS, Oslo Science Park, Gaustadalléen 21, Oslo, Norway
| | - Ana Finzel
- Bio-Me AS, Oslo Science Park, Gaustadalléen 21, Oslo, Norway
| | | | - Bernard Escudier
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Laurence Albiges
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - David Planchard
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Fabrice André
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Chiara Cremolini
- Unit of Medical Oncology 2, University Hospital of Pisa, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Stéphanie Martinez
- Service des Maladies Respiratoires, Centre Hospitalier d'Aix-en-Provence, Aix-en-Provence, France
| | - Benjamin Besse
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Liping Zhao
- Center for Nutrition, Microbiome and Health, New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA; Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA; Rutgers-Jiaotong Joint Laboratory for Microbiome and Human Health, New Brunswick, NJ, USA; State Key Laboratory of Microbial Metabolism, Ministry of Education Laboratory of Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy; IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Guido Kroemer
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée-Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Inserm U1138, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, ClinicObiome, Villejuif, France; Université Paris-Saclay, Ile-de-France, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France.
| |
Collapse
|
9
|
Blake SJ, Wolf Y, Boursi B, Lynn DJ. Role of the microbiota in response to and recovery from cancer therapy. Nat Rev Immunol 2024; 24:308-325. [PMID: 37932511 DOI: 10.1038/s41577-023-00951-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2023] [Indexed: 11/08/2023]
Abstract
Our understanding of how the microbiota affects the balance between response to and failure of cancer treatment by modulating the tumour microenvironment and systemic immune system has advanced rapidly in recent years. Microbiota-targeting interventions in patients with cancer are an area of intensive investigation. Promisingly, phase I-II clinical trials have shown that interventions such as faecal microbiota transplantation can overcome resistance to immune checkpoint blockade in patients with melanoma, improve therapeutic outcomes in treatment-naive patients and reduce therapy-induced immunotoxicities. Here, we synthesize the evidence showing that the microbiota is an important determinant of both cancer treatment efficacy and treatment-induced acute and long-term toxicity, and we discuss the complex and inter-related mechanisms involved. We also assess the potential of microbiota-targeting interventions, including bacterial engineering and phage therapy, to optimize the response to and recovery from cancer therapy.
Collapse
Affiliation(s)
- Stephen J Blake
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Yochai Wolf
- Ella Lemelbaum Institute for Immuno-oncology and Skin Cancer, Sheba Medical Center, Tel Hashomer, Israel
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ben Boursi
- School of Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Oncology, Sheba Medical Center, Tel Hashomer, Israel
- Center of Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Lynn
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
10
|
Yarahmadi A, Zare M, Aghayari M, Afkhami H, Jafari GA. Therapeutic bacteria and viruses to combat cancer: double-edged sword in cancer therapy: new insights for future. Cell Commun Signal 2024; 22:239. [PMID: 38654309 PMCID: PMC11040964 DOI: 10.1186/s12964-024-01622-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Cancer, ranked as the second leading cause of mortality worldwide, leads to the death of approximately seven million people annually, establishing itself as one of the most significant health challenges globally. The discovery and identification of new anti-cancer drugs that kill or inactivate cancer cells without harming normal and healthy cells and reduce adverse effects on the immune system is a potential challenge in medicine and a fundamental goal in Many studies. Therapeutic bacteria and viruses have become a dual-faceted instrument in cancer therapy. They provide a promising avenue for cancer treatment, but at the same time, they also create significant obstacles and complications that contribute to cancer growth and development. This review article explores the role of bacteria and viruses in cancer treatment, examining their potential benefits and drawbacks. By amalgamating established knowledge and perspectives, this review offers an in-depth examination of the present research landscape within this domain and identifies avenues for future investigation.
Collapse
Affiliation(s)
- Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Mitra Zare
- Department of Microbiology, Faculty of Sciences, Kerman Branch, Islamic Azad University, Kerman, Iran
| | - Masoomeh Aghayari
- Department of Microbiology, Faculty of Sciences, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | - Gholam Ali Jafari
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
11
|
Hurst R, Brewer DS, Gihawi A, Wain J, Cooper CS. Cancer invasion and anaerobic bacteria: new insights into mechanisms. J Med Microbiol 2024; 73:001817. [PMID: 38535967 PMCID: PMC10995961 DOI: 10.1099/jmm.0.001817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/27/2024] [Indexed: 04/07/2024] Open
Abstract
There is growing evidence that altered microbiota abundance of a range of specific anaerobic bacteria are associated with cancer, including Peptoniphilus spp., Porphyromonas spp., Fusobacterium spp., Fenollaria spp., Prevotella spp., Sneathia spp., Veillonella spp. and Anaerococcus spp. linked to multiple cancer types. In this review we explore these pathogenic associations. The mechanisms by which bacteria are known or predicted to interact with human cells are reviewed and we present an overview of the interlinked mechanisms and hypotheses of how multiple intracellular anaerobic bacterial pathogens may act together to cause host cell and tissue microenvironment changes associated with carcinogenesis and cancer cell invasion. These include combined effects on changes in cell signalling, DNA damage, cellular metabolism and immune evasion. Strategies for early detection and eradication of anaerobic cancer-associated bacterial pathogens that may prevent cancer progression are proposed.
Collapse
Affiliation(s)
- Rachel Hurst
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - Daniel S. Brewer
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
- Earlham Institute, Norwich Research Park Innovation Centre, Colney Lane, Norwich NR4 7UZ, UK
| | - Abraham Gihawi
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - John Wain
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
- Quadram Institute Biosciences, Colney Lane, Norwich, Norfolk, NR4 7UQ, UK
| | - Colin S. Cooper
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| |
Collapse
|
12
|
Mignini I, Piccirilli G, Galasso L, Termite F, Esposto G, Ainora ME, Gasbarrini A, Zocco MA. From the Colon to the Liver: How Gut Microbiota May Influence Colorectal Cancer Metastatic Potential. J Clin Med 2024; 13:420. [PMID: 38256554 PMCID: PMC10815973 DOI: 10.3390/jcm13020420] [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: 12/18/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
The gut microbiota's influence on human tumorigenesis is a burning topic in medical research. With the new ontological perspective, which considers the human body and its pathophysiological processes as the result of the interaction between its own eukaryotic cells and prokaryotic microorganisms living in different body niches, great interest has arisen in the role of the gut microbiota on carcinogenesis. Indeed, dysbiosis is currently recognized as a cancer-promoting condition, and multiple molecular mechanisms have been described by which the gut microbiota may drive tumor development, especially colorectal cancer (CRC). Metastatic power is undoubtedly one of the most fearsome features of neoplastic tissues. Therefore, understanding the underlying mechanisms is of utmost importance to improve patients' prognosis. The liver is the most frequent target of CRC metastasis, and new evidence reveals that the gut microbiota may yield an effect on CRC diffusion to the liver, thus defining an intriguing new facet of the so-called "gut-liver axis". In this review, we aim to summarize the most recent data about the microbiota's role in promoting or preventing hepatic metastasis from CRC, highlighting some potential future therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (G.P.); (L.G.); (F.T.); (G.E.); (M.E.A.); (A.G.)
| |
Collapse
|
13
|
Tsakmaklis A, Farowski F, Zenner R, Lesker TR, Strowig T, Schlößer H, Lehmann J, von Bergwelt-Baildon M, Mauch C, Schlaak M, Knuever J, Schweinsberg V, Heinzerling LM, Vehreschild MJGT. TIGIT + NK cells in combination with specific gut microbiota features predict response to checkpoint inhibitor therapy in melanoma patients. BMC Cancer 2023; 23:1160. [PMID: 38017389 PMCID: PMC10685659 DOI: 10.1186/s12885-023-11551-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/20/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Composition of the intestinal microbiota has been correlated to therapeutic efficacy of immune checkpoint inhibitors (ICI) in various cancer entities including melanoma. Prediction of the outcome of such therapy, however, is still unavailable. This prospective, non-interventional study was conducted in order to achieve an integrated assessment of the connection between a specific intestinal microbiota profile and antitumor immune response to immune checkpoint inhibitor therapy (anti-PD-1 and/or anti-CTLA-4) in melanoma patients. METHODS We assessed blood and stool samples of 29 cutaneous melanoma patients who received immune checkpoint inhibitor therapy. For functional and phenotypical immune analysis, 12-color flow cytometry and FluoroSpot assays were conducted. Gut microbiome was analyzed with shotgun metagenomics sequencing. To combine clinical, microbiome and immune variables, we applied the Random Forest algorithm. RESULTS A total of 29 patients was analyzed in this study, among whom 51.7% (n = 15) reached a durable clinical benefit. The Immune receptor TIGIT is significantly upregulated in T cells (p = 0.0139) and CD56high NK cells (p = 0.0037) of responders. Several bacterial taxa were associated with response (e.g. Ruminococcus torques) or failure (e.g. Barnesiella intestinihominis) to immune therapy. A combination of two microbiome features (Barnesiella intestinihominis and the Enterobacteriaceae family) and one immune feature (TIGIT+ CD56high NK cells) was able to predict response to ICI already at baseline (AUC = 0.85; 95% CI: 0.841-0.853). CONCLUSIONS Our results reconfirm a link between intestinal microbiota and response to ICI therapy in melanoma patients and furthermore point to TIGIT as a promising target for future immunotherapies.
Collapse
Affiliation(s)
- Anastasia Tsakmaklis
- Department I of Internal Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, Cologne, Germany
| | - Fedja Farowski
- Department I of Internal Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, Cologne, Germany
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt, 60590, Germany
| | - Rafael Zenner
- Department I of Internal Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Till Robin Lesker
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Till Strowig
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Hans Schlößer
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Interventional Immunology, University of Cologne, Cologne, Germany
| | - Jonas Lehmann
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Interventional Immunology, University of Cologne, Cologne, Germany
| | | | - Cornelia Mauch
- Department of Dermatology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Max Schlaak
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Jana Knuever
- Department of Dermatology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Viola Schweinsberg
- Department of Dermatology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lucie M Heinzerling
- Department of Dermatology and Allergy, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Maria J G T Vehreschild
- Department I of Internal Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, Cologne, Germany.
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt, 60590, Germany.
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine & Pharmacology ITMP, Frankfurt am Main, 60596, Germany.
| |
Collapse
|
14
|
Tu SM, Aydin AM, Maraboyina S, Chen Z, Singh S, Gokden N, Langford T. Stem Cell Origin of Cancer: Clinical Implications for Cancer Immunity and Immunotherapy. Cancers (Basel) 2023; 15:5385. [PMID: 38001645 PMCID: PMC10670143 DOI: 10.3390/cancers15225385] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
A simple way to understand the immune system is to separate the self from non-self. If it is self, the immune system tolerates and spares. If it is non-self, the immune system attacks and destroys. Consequently, if cancer has a stem cell origin and is a stem cell disease, we have a serious problem and a major dilemma with immunotherapy. Because many refractory cancers are more self than non-self, immunotherapy may become an uphill battle and pyrrhic victory in cancer care. In this article, we elucidate cancer immunity. We demonstrate for whom, with what, as well as when and how to apply immunotherapy in cancer care. We illustrate that a stem cell theory of cancer affects our perspectives and narratives of cancer. Without a pertinent theory about cancer's origin and nature, we may unwittingly perform misdirected cancer research and prescribe misguided cancer treatments. In the ongoing saga of immunotherapy, we are at a critical juncture. Because of the allure and promises of immunotherapy, we will be treating more patients not immediately threatened by their cancer. They may have more to lose than to gain, if we have a misconception and if we are on a wrong mission with immunotherapy. According to the stem cell theory of cancer, we should be careful with immunotherapy. When we do not know or realize that cancer originates from a stem cell and has stem-ness capabilities, we may cause more harm than good in some patients and fail to separate the truth from the myth about immunotherapy in cancer care.
Collapse
Affiliation(s)
- Shi-Ming Tu
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (Z.C.); (S.S.)
| | - Ahmet Murat Aydin
- Department of Urology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.M.A.); (T.L.)
| | - Sanjay Maraboyina
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Zhongning Chen
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (Z.C.); (S.S.)
| | - Sunny Singh
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (Z.C.); (S.S.)
| | - Neriman Gokden
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Timothy Langford
- Department of Urology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.M.A.); (T.L.)
| |
Collapse
|
15
|
Zhou X, You L, Xin Z, Su H, Zhou J, Ma Y. Leveraging circulating microbiome signatures to predict tumor immune microenvironment and prognosis of patients with non-small cell lung cancer. J Transl Med 2023; 21:800. [PMID: 37950236 PMCID: PMC10636862 DOI: 10.1186/s12967-023-04582-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/29/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Accumulating evidence supports the significant role of human microbiome in development and therapeutic response of tumors. Circulating microbial DNA is non-invasive and could show a general view of the microbiome of host, making it a promising biomarker for cancers. However, whether circulating microbiome is associated with prognosis of non-small cell lung cancer (NSCLC) and its potential mechanisms on tumor immune microenvironment still remains unknown. METHODS The blood microbiome data and matching tumor RNA-seq data of TCGA NSCLC patients were obtained from Poore's study and UCSC Xena. Univariate and multivariate Cox regression analysis were used to identify circulating microbiome signatures associated with overall survival (OS) and construct the circulating microbial abundance prognostic scoring (MAPS) model. Nomograms integrating clinical characteristics and circulating MAPS scores were established to predict OS rate of NSCLC patients. Joint analysis of blood microbiome data and matching tumor RNA-seq data was used to deciphered the tumor microenvironment landscape of patients in circulating MAPS-high and MAPS-low groups. Finally, the predictive value of circulating MAPS on the efficacy of immunotherapy and chemotherapy were assessed. RESULTS A circulating MAPS prediction model consisting of 14 circulating microbes was constructed and had an independent prognostic value for NSCLC. The integration of circulating MAPS into nomograms may improve the prognosis predictive power. Joint analysis revealed potential interactions between prognostic circulating microbiome and tumor immune microenvironment. Especially, intratumor plasma cells and humoral immune response were enriched in circulating MAPS-low group, while intratumor CD4 + Th2 cells and proliferative related pathways were enriched in MAPS-high group. Finally, drug sensitivity analysis indicated the potential of circulating MAPS as a predictor of chemotherapy efficacy. CONCLUSION A circulating MAPS prediction model was constructed successfully and showed great prognostic value for NSCLC. Our study provides new insights of interactions between microbes, tumors and immunity, and may further contribute to precision medicine for NSCLC.
Collapse
Affiliation(s)
- Xiaohan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Liting You
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Zhaodan Xin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Huiting Su
- Department of Laboratory Medicine, Guang 'an People's Hospital, Guang 'an, 638000, Sichuan, People's Republic of China
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Ying Ma
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
16
|
Joachim L, Göttert S, Sax A, Steiger K, Neuhaus K, Heinrich P, Fan K, Orberg ET, Kleigrewe K, Ruland J, Bassermann F, Herr W, Posch C, Heidegger S, Poeck H. The microbial metabolite desaminotyrosine enhances T-cell priming and cancer immunotherapy with immune checkpoint inhibitors. EBioMedicine 2023; 97:104834. [PMID: 37865045 PMCID: PMC10597767 DOI: 10.1016/j.ebiom.2023.104834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/23/2023] Open
Abstract
BACKGROUND Inter-individual differences in response to immune checkpoint inhibitors (ICI) remain a major challenge in cancer treatment. The composition of the gut microbiome has been associated with differential ICI outcome, but the underlying molecular mechanisms remain unclear, and therapeutic modulation challenging. METHODS We established an in vivo model to treat C57Bl/6j mice with the type-I interferon (IFN-I)-modulating, bacterial-derived metabolite desaminotyrosine (DAT) to improve ICI therapy. Broad spectrum antibiotics were used to mimic gut microbial dysbiosis and associated ICI resistance. We utilized genetic mouse models to address the role of host IFN-I in DAT-modulated antitumour immunity. Changes in gut microbiota were assessed using 16S-rRNA sequencing analyses. FINDINGS We found that oral supplementation of mice with the microbial metabolite DAT delays tumour growth and promotes ICI immunotherapy with anti-CTLA-4 or anti-PD-1. DAT-enhanced antitumour immunity was associated with more activated T cells and natural killer cells in the tumour microenvironment and was dependent on host IFN-I signalling. Consistent with this, DAT potently enhanced expansion of antigen-specific T cells following vaccination with an IFN-I-inducing adjuvant. DAT supplementation in mice compensated for the negative effects of broad-spectrum antibiotic-induced dysbiosis on anti-CTLA-4-mediated antitumour immunity. Oral administration of DAT altered the gut microbial composition in mice with increased abundance of bacterial taxa that are associated with beneficial response to ICI immunotherapy. INTERPRETATION We introduce the therapeutic use of an IFN-I-modulating bacterial-derived metabolite to overcome resistance to ICI. This approach is a promising strategy particularly for patients with a history of broad-spectrum antibiotic use and associated loss of gut microbial diversity. FUNDING Melanoma Research Alliance, Deutsche Forschungsgemeinschaft, German Cancer Aid, Wilhelm Sander Foundation, Novartis Foundation.
Collapse
Affiliation(s)
- Laura Joachim
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Centre for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Sascha Göttert
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Anna Sax
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Centre for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome, ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Paul Heinrich
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Kaiji Fan
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Erik Thiele Orberg
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Centre for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Karin Kleigrewe
- Bavarian Centre for Biomolecular Mass Spectrometry, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Jürgen Ruland
- Centre for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
| | - Florian Bassermann
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Centre for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Christian Posch
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany; Faculty of Medicine, Sigmund Freud University Vienna, Vienna, Austria
| | - Simon Heidegger
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany; Centre for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.
| | - Hendrik Poeck
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany; Centre for Immunomedicine in Transplantation and Oncology (CITO), Regensburg, Germany; Bavarian Cancer Research Centre (BZKF), Regensburg, Germany.
| |
Collapse
|
17
|
Schorr L, Mathies M, Elinav E, Puschhof J. Intracellular bacteria in cancer-prospects and debates. NPJ Biofilms Microbiomes 2023; 9:76. [PMID: 37813921 PMCID: PMC10562400 DOI: 10.1038/s41522-023-00446-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023] Open
Abstract
Recent evidence suggests that some human cancers may harbor low-biomass microbial ecosystems, spanning bacteria, viruses, and fungi. Bacteria, the most-studied kingdom in this context, are suggested by these studies to localize within cancer cells, immune cells and other tumor microenvironment cell types, where they are postulated to impact multiple cancer-related functions. Herein, we provide an overview of intratumoral bacteria, while focusing on intracellular bacteria, their suggested molecular activities, communication networks, host invasion and evasion strategies, and long-term colonization capacity. We highlight how the integration of sequencing-based and spatial techniques may enable the recognition of bacterial tumor niches. We discuss pitfalls, debates and challenges in decisively proving the existence and function of intratumoral microbes, while reaching a mechanistic elucidation of their impacts on tumor behavior and treatment responses. Together, a causative understanding of possible roles played by intracellular bacteria in cancer may enable their future utilization in diagnosis, patient stratification, and treatment.
Collapse
Affiliation(s)
- Lena Schorr
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Marius Mathies
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany.
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| | - Jens Puschhof
- Microbiome and Cancer Division, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
18
|
Simpson RC, Shanahan ER, Scolyer RA, Long GV. Towards modulating the gut microbiota to enhance the efficacy of immune-checkpoint inhibitors. Nat Rev Clin Oncol 2023; 20:697-715. [PMID: 37488231 DOI: 10.1038/s41571-023-00803-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2023] [Indexed: 07/26/2023]
Abstract
The gut microbiota modulates immune processes both locally and systemically. This includes whether and how the immune system reacts to emerging tumours, whether antitumour immune responses are reactivated during treatment with immune-checkpoint inhibitors (ICIs), and whether unintended destructive immune pathologies accompany such treatment. Advances over the past decade have established that the gut microbiota is a promising target and that modulation of the microbiota might overcome resistance to ICIs and/or improve the safety of treatment. However, the specific mechanisms through which the microbiota modulates antitumour immunity remain unclear. Understanding the biology underpinning microbial associations with clinical outcomes in patients receiving ICIs, as well as the landscape of a 'healthy' microbiota would provide a critical foundation to facilitate opportunities to effectively manipulate the microbiota and thus improve patient outcomes. In this Review, we explore the role of diet and the gut microbiota in shaping immune responses during treatment with ICIs and highlight the key challenges in attempting to leverage the gut microbiome as a practical tool for the clinical management of patients with cancer.
Collapse
Affiliation(s)
- Rebecca C Simpson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Erin R Shanahan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.
- Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia.
| |
Collapse
|
19
|
Zhuang YP, Zhou HL, Chen HB, Zheng MY, Liang YW, Gu YT, Li WT, Qiu WL, Zhou HG. Gut microbiota interactions with antitumor immunity in colorectal cancer: From understanding to application. Biomed Pharmacother 2023; 165:115040. [PMID: 37364479 DOI: 10.1016/j.biopha.2023.115040] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Colorectal cancer (CRC) is one of highly prevalent cancer. Immunotherapy with immune checkpoint inhibitors (ICIs) has dramatically changed the landscape of treatment for many advanced cancers, but CRC still exhibits suboptimal response to immunotherapy. The gut microbiota can affect both anti-tumor and pro-tumor immune responses, and further modulate the efficacy of cancer immunotherapy, particularly in the context of therapy with ICIs. Therefore, a deeper understanding of how the gut microbiota modulates immune responses is crucial to improve the outcomes of CRC patients receiving immunotherapy and to overcome resistance in nonresponders. The present review aims to describe the relationship between the gut microbiota, CRC, and antitumor immune responses, with a particular focus on key studies and recent findings on the effect of the gut microbiota on the antitumor immune activity. We also discuss the potential mechanisms by which the gut microbiota influences host antitumor immune responses as well as the prospective role of intestinal flora in CRC treatment. Furthermore, the therapeutic potential and limitations of different modulation strategies for the gut microbiota are also discussed. These insights may facilitate to better comprehend the interplay between the gut microbiota and the antitumor immune responses of CRC patients and provide new research pathways to enhance immunotherapy efficacy and expand the patient population that could be benefited by immunotherapy.
Collapse
Affiliation(s)
- Yu-Pei Zhuang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong-Li Zhou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hai-Bin Chen
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming-Yue Zheng
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu-Wei Liang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu-Tian Gu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen-Ting Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Wen-Li Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Hong-Guang Zhou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
20
|
Pandey P, Khan F. Gut microbiome in cancer immunotherapy: Current trends, translational challenges and future possibilities. Biochim Biophys Acta Gen Subj 2023; 1867:130401. [PMID: 37307905 DOI: 10.1016/j.bbagen.2023.130401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
Gut microbiota is regarded as a crucial regulator of the immune system. Healthy gut microbiota plays a specialized role in host xenobiotics, nutrition, drug metabolism, regulation of the structural integrity of the gut mucosal barrier, defense against infections, and immunomodulation. It is now understood that any imbalance in gut microbiota composition from that present in a healthy state is linked to genetic susceptibility to a number of metabolic disorders, including diabetes, autoimmunity, and cancer. Recent research has suggested that immunotherapy can treat many different cancer types with fewer side effects and better ability to eradicate tumors than conventional chemotherapy or radiotherapy. However, a significant number of patients eventually develop immunotherapy resistance. A strong correlation was observed between the composition of the gut microbiome and the effectiveness of treatment by examining the variations between populations that responded to immunotherapy and those that did not. Therefore, we suggest that modulating the microbiome could be a potential adjuvant therapy for cancer immunotherapy and that the architecture of the gut microbiota may be helpful in explaining the variation in treatment response. Herein, we focus on recent research on the interactions among the gut microbiome, host immunity, and cancer immunotherapy. In addition, we highlighted the clinical manifestations, future opportunities, and limitations of microbiome manipulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida 201306, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida 201306, India.
| |
Collapse
|
21
|
Fortman DD, Hurd D, Davar D. The Microbiome in Advanced Melanoma: Where Are We Now? Curr Oncol Rep 2023; 25:997-1016. [PMID: 37269504 PMCID: PMC11090495 DOI: 10.1007/s11912-023-01431-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE OF REVIEW This review summarizes recent data linking gut microbiota composition to ICI outcomes and gut microbiota-specific interventional clinical trials in melanoma. RECENT FINDINGS Preclinical and clinical studies have demonstrated the effects of the gut microbiome modulation upon ICI response in advanced melanoma, with growing evidence supporting the ability of the gut microbiome to restore or improve ICI response in advanced melanoma through dietary fiber, probiotics, and FMT. Immune checkpoint inhibitors (ICI) targeting the PD-1, CTLA-4, and LAG-3 negative regulatory checkpoints have transformed the management of melanoma. ICIs are FDA-approved in advanced metastatic disease, stage III resected melanoma, and high-risk stage II melanoma and are being investigated more recently in the management of high-risk resectable melanoma in the peri-operative setting. The gut microbiome has emerged as an important tumor-extrinsic modulator of both response and immune-related adverse event (irAE) development in ICI-treated cancer in general, and melanoma in particular.
Collapse
Affiliation(s)
- Dylan D Fortman
- Division of General Internal Medicine, Department of Medicine, University of Pittsburgh Medical Center (UPMC) and University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Drew Hurd
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pavilion, Suite 1.32d, 5115, Center Avenue, Pittsburgh, PA, 15213, USA
| | - Diwakar Davar
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pavilion, Suite 1.32d, 5115, Center Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
22
|
Qu R, Zhang Y, Ma Y, Zhou X, Sun L, Jiang C, Zhang Z, Fu W. Role of the Gut Microbiota and Its Metabolites in Tumorigenesis or Development of Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205563. [PMID: 37263983 PMCID: PMC10427379 DOI: 10.1002/advs.202205563] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/20/2023] [Indexed: 06/03/2023]
Abstract
Colorectal cancer (CRC) is the most common cancer of the digestive system with high mortality and morbidity rates. Gut microbiota is found in the intestines, especially the colorectum, and has structured crosstalk interactions with the host that affect several physiological processes. The gut microbiota include CRC-promoting bacterial species, such as Fusobacterium nucleatum, Escherichia coli, and Bacteroides fragilis, and CRC-protecting bacterial species, such as Clostridium butyricum, Streptococcus thermophilus, and Lacticaseibacillus paracasei, which along with other microorganisms, such as viruses and fungi, play critical roles in the development of CRC. Different bacterial features are identified in patients with early-onset CRC, combined with different patterns between fecal and intratumoral microbiota. The gut microbiota may be beneficial in the diagnosis and treatment of CRC; some bacteria may serve as biomarkers while others as regulators of chemotherapy and immunotherapy. Furthermore, metabolites produced by the gut microbiota play essential roles in the crosstalk with CRC cells. Harmful metabolites include some primary bile acids and short-chain fatty acids, whereas others, including ursodeoxycholic acid and butyrate, are beneficial and impede tumor development and progression. This review focuses on the gut microbiota and its metabolites, and their potential roles in the development, diagnosis, and treatment of CRC.
Collapse
Affiliation(s)
- Ruize Qu
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Yi Zhang
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Yanpeng Ma
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Xin Zhou
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Lulu Sun
- State Key Laboratory of Women's Reproductive Health and Fertility PromotionPeking UniversityBeijing100191P. R. China
- Department of Endocrinology and MetabolismPeking University Third HospitalBeijing100191P. R. China
| | - Changtao Jiang
- Center of Basic Medical ResearchInstitute of Medical Innovation and ResearchThird HospitalPeking UniversityBeijing100191P. R. China
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesPeking University and the Key Laboratory of Molecular Cardiovascular Science (Peking University)Ministry of EducationBeijing100191P. R. China
- Center for Obesity and Metabolic Disease ResearchSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Zhipeng Zhang
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Wei Fu
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| |
Collapse
|
23
|
Abstract
After decades of research, our knowledge of the complexity of cancer mechanisms, elegantly summarized as 'hallmarks of cancer', is expanding, as are the therapeutic opportunities that this knowledge brings. However, cancer still needs intense research to diminish its tremendous impact. In this context, the use of simple model organisms such as Caenorhabditis elegans, in which the genetics of the apoptotic pathway was discovered, can facilitate the investigation of several cancer hallmarks. Amenable for genetic and drug screens, convenient for fast and efficient genome editing, and aligned with the 3Rs ('Replacement, Reduction and Refinement') principles for ethical animal research, C. elegans plays a significant role in unravelling the intricate network of cancer mechanisms and presents a promising option in clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Julián Cerón
- Modeling Human Diseases in C. elegans Group – Genes, Disease and Therapy Program, Bellvitge Biomedical Research Institute – IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
24
|
Wong CC, Yu J. Gut microbiota in colorectal cancer development and therapy. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00766-x. [PMID: 37169888 DOI: 10.1038/s41571-023-00766-x] [Citation(s) in RCA: 141] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2023] [Indexed: 05/13/2023]
Abstract
Colorectal cancer (CRC) is one of the commonest cancers globally. A unique aspect of CRC is its intimate association with the gut microbiota, which forms an essential part of the tumour microenvironment. Research over the past decade has established that dysbiosis of gut bacteria, fungi, viruses and Archaea accompanies colorectal tumorigenesis, and these changes might be causative. Data from mechanistic studies demonstrate the ability of the gut microbiota to interact with the colonic epithelia and immune cells of the host via the release of a diverse range of metabolites, proteins and macromolecules that regulate CRC development. Preclinical and some clinical evidence also underscores the role of the gut microbiota in modifying the therapeutic responses of patients with CRC to chemotherapy and immunotherapy. Herein, we summarize our current understanding of the role of gut microbiota in CRC and outline the potential translational and clinical implications for CRC diagnosis, prevention and treatment. Emphasis is placed on how the gut microbiota could now be better harnessed by developing targeted microbial therapeutics as chemopreventive agents against colorectal tumorigenesis, as adjuvants for chemotherapy and immunotherapy to boost drug efficacy and safety, and as non-invasive biomarkers for CRC screening and patient stratification. Finally, we highlight the hurdles and potential solutions to translating our knowledge of the gut microbiota into clinical practice.
Collapse
Affiliation(s)
- Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
25
|
Xia C, Su J, Liu C, Mai Z, Yin S, Yang C, Fu L. Human microbiomes in cancer development and therapy. MedComm (Beijing) 2023; 4:e221. [PMID: 36860568 PMCID: PMC9969057 DOI: 10.1002/mco2.221] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/25/2023] [Accepted: 02/01/2023] [Indexed: 03/03/2023] Open
Abstract
Colonies formed by bacteria, archaea, fungi, and viral groups and their genomes, metabolites, and expressed proteins constitute complex human microbiomes. An increasing evidences showed that carcinogenesis and disease progression were link to microbiomes. Different organ sources, their microbial species, and their metabolites are different; the mechanisms of carcinogenic or procancerous are also different. Here, we summarize how microbiomes contribute to carcinogenesis and disease progression in cancers of the skin, mouth, esophagus, lung, gastrointestinal, genital, blood, and lymph malignancy. We also insight into the molecular mechanisms of triggering, promoting, or inhibiting carcinogenesis and disease progress induced by microbiomes or/and their secretions of bioactive metabolites. And then, the strategies of application of microorganisms in cancer treatment were discussed in detail. However, the mechanisms by which human microbiomes function are still poorly understood. The bidirectional interactions between microbiotas and endocrine systems need to be clarified. Probiotics and prebiotics are believed to benefit human health via a variety of mechanisms, in particular, in tumor inhibition. It is largely unknown how microbial agents cause cancer or how cancer progresses. We expect this review may open new perspectives on possible therapeutic approaches of patients with cancer.
Collapse
Affiliation(s)
- Chenglai Xia
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Jiyan Su
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Can Liu
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zhikai Mai
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Shuanghong Yin
- Affiliated Foshan Maternity and Chlid Healthcare HospitalSouthern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical UniversityGuangzhouChina
| | - Chuansheng Yang
- Department of Head‐Neck and Breast SurgeryYuebei People's Hospital of Shantou UniversityShaoguanChina
| | - Liwu Fu
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Sun Yat‐sen University Cancer CenterGuangzhouPeople's Republic of China
| |
Collapse
|
26
|
Wang L, Tang L, Zhai D, Song M, Li W, Xu S, Jiang S, Meng H, Liang J, Wang Y, Zhang B. The role of the sex hormone-gut microbiome axis in tumor immunotherapy. Gut Microbes 2023; 15:2185035. [PMID: 36880651 PMCID: PMC10012946 DOI: 10.1080/19490976.2023.2185035] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
Accumulating evidence suggested that both gut microbiome and sex play a critical role in the efficacy of immune checkpoint blockade therapy. Considering the reciprocal relationship between sex hormones and gut microbiome, the sex hormone-gut microbiome axis may participate in the regulation of the response to immune checkpoint inhibitors (ICIs). In this review, it was attempted to summarize the current knowledge about the influences of both sex and gut microbiome on the antitumor efficacy of ICIs and describe the interaction between sex hormones and gut microbiome. Accordingly, this review discussed the potential of enhancing the antitumor efficacy of ICIs through regulating the levels of sex hormones through manipulation of gut microbiome. Collectively, this review provided reliable evidence concerning the role of the sex hormone-gut microbiome axis in tumor immunotherapy.
Collapse
Affiliation(s)
- Luoyang Wang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lei Tang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Dongchang Zhai
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Meiying Song
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wei Li
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Shuo Xu
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Suli Jiang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Haining Meng
- School of Emergency Medicine, Qingdao University, Qingdao, China
| | - Jie Liang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yingying Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Abstract
Trillions of microbes are indigenous to the human gastrointestinal tract, together forming an ecological community known as the gut microbiota. The gut microbiota is involved in dietary digestion to produce various metabolites. In healthy condition, microbial metabolites have unneglectable roles in regulating host physiology and intestinal homeostasis. However, increasing studies have reported the correlation between metabolites and the development of colorectal cancer (CRC), with the identification of oncometabolites. Meanwhile, metabolites can also influence the efficacy of cancer treatments. In this review, metabolites derived from microbes-mediated metabolism of dietary carbohydrates, proteins, and cholesterol, are introduced. The roles of pro-tumorigenic (secondary bile acids and polyamines) and anti-tumorigenic (short-chain fatty acids and indole derivatives) metabolites in CRC development are then discussed. The impacts of metabolites on chemotherapy and immunotherapy are further elucidated. Collectively, given the importance of microbial metabolites in CRC, therapeutic approaches that target metabolites may be promising to improve patient outcome.
Collapse
Affiliation(s)
- Yali Liu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| |
Collapse
|
28
|
Villemin C, Six A, Neville BA, Lawley TD, Robinson MJ, Bakdash G. The heightened importance of the microbiome in cancer immunotherapy. Trends Immunol 2023; 44:44-59. [PMID: 36464584 DOI: 10.1016/j.it.2022.11.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
The human microbiome is recognized as a key factor in health and disease. This has been further corroborated by identifying changes in microbiome composition and function as a novel hallmark in cancer. These effects are exerted through microbiome interactions with host cells, impacting a wide variety of developmental and physiological processes. In this review, we discuss some of the latest findings on how the bacterial component of the microbiome can influence outcomes for different cancer immunotherapy modalities, highlighting identified mechanisms of action. We also address the clinical efforts to utilize this knowledge to achieve better responses to immunotherapy. A refined understanding of microbiome variations in patients and microbiome-host interactions with cancer therapies is essential to realize optimal clinical responses.
Collapse
Affiliation(s)
| | - Anne Six
- Microbiotica Ltd., Cambridge, UK
| | | | - Trevor D Lawley
- Microbiotica Ltd., Cambridge, UK; Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | | | | |
Collapse
|
29
|
Fernandes MR, Aggarwal P, Costa RGF, Cole AM, Trinchieri G. Targeting the gut microbiota for cancer therapy. Nat Rev Cancer 2022; 22:703-722. [PMID: 36253536 DOI: 10.1038/s41568-022-00513-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 02/06/2023]
Abstract
Growing evidence suggests that the gut microbiota modulates the efficacy and toxicity of cancer therapy, most notably immunotherapy and its immune-related adverse effects. The poor response to immunotherapy in patients treated with antibiotics supports this influential role of the microbiota. Until recently, results pertaining to the identification of the microbial species responsible for these effects were incongruent, and relatively few studies analysed the underlying mechanisms. A better understanding of the taxonomy of the species involved and of the mechanisms of action has since been achieved. Defined bacterial species have been shown to promote an improved response to immune-checkpoint inhibitors by producing different products or metabolites. However, a suppressive effect of Gram-negative bacteria may be dominant in some unresponsive patients. Machine learning approaches trained on the microbiota composition of patients can predict the ability of patients to respond to immunotherapy with some accuracy. Thus, interest in modulating the microbiota composition to improve patient responsiveness to therapy has been mounting. Clinical proof-of-concept studies have demonstrated that faecal microbiota transplantation or dietary interventions might be utilized clinically to improve the success rate of immunotherapy in patients with cancer. Here, we review recent advances and discuss emerging strategies for microbiota-based cancer therapies.
Collapse
Affiliation(s)
- Miriam R Fernandes
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Poonam Aggarwal
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Raquel G F Costa
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Alicia M Cole
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
30
|
Hou X, Zheng Z, Wei J, Zhao L. Effects of gut microbiota on immune responses and immunotherapy in colorectal cancer. Front Immunol 2022; 13:1030745. [PMID: 36426359 PMCID: PMC9681148 DOI: 10.3389/fimmu.2022.1030745] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/24/2022] [Indexed: 08/13/2023] Open
Abstract
Accumulating evidence suggests that gut microbial dysbiosis is implicated in colorectal cancer (CRC) initiation and progression through interaction with host immune system. Given the intimate relationship between the gut microbiota and the antitumor immune responses, the microbiota has proven to be effective targets in modulating immunotherapy responses of preclinical CRC models. However, the proposed putative mechanisms of how these bacteria affect immune responses and immunotherapy efficacy remains obscure. In this review, we summarize recent findings of clinical gut microbial dysbiosis in CRC patients, the reciprocal interactions between gut microbiota and the innate and/or the adaptive immune system, as well as the effect of gut microbiota on immunotherapy response in CRC. Increased understanding of the gut microbiota-immune system interactions will benefit the rational application of microbiota to the clinical promising biomarker or therapeutic strategy as a cancer immunotherapy adjuvant.
Collapse
Affiliation(s)
| | | | | | - Ling Zhao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
31
|
Diet-driven microbial ecology underpins associations between cancer immunotherapy outcomes and the gut microbiome. Nat Med 2022; 28:2344-2352. [PMID: 36138151 DOI: 10.1038/s41591-022-01965-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/22/2022] [Indexed: 01/14/2023]
Abstract
The gut microbiota shapes the response to immune checkpoint inhibitors (ICIs) in cancer, however dietary and geographic influences have not been well-studied in prospective trials. To address this, we prospectively profiled baseline gut (fecal) microbiota signatures and dietary patterns of 103 trial patients from Australia and the Netherlands treated with neoadjuvant ICIs for high risk resectable metastatic melanoma and performed an integrated analysis with data from 115 patients with melanoma treated with ICIs in the United States. We observed geographically distinct microbial signatures of response and immune-related adverse events (irAEs). Overall, response rates were higher in Ruminococcaceae-dominated microbiomes than in Bacteroidaceae-dominated microbiomes. Poor response was associated with lower fiber and omega 3 fatty acid consumption and elevated levels of C-reactive protein in the peripheral circulation at baseline. Together, these data provide insight into the relevance of native gut microbiota signatures, dietary intake and systemic inflammation in shaping the response to and toxicity from ICIs, prompting the need for further studies in this area.
Collapse
|
32
|
Davar D, Zarour HM. Facts and Hopes for Gut Microbiota Interventions in Cancer Immunotherapy. Clin Cancer Res 2022; 28:4370-4384. [PMID: 35748749 PMCID: PMC9561605 DOI: 10.1158/1078-0432.ccr-21-1129] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/06/2022] [Accepted: 06/06/2022] [Indexed: 01/07/2023]
Abstract
Immune checkpoint inhibitors (ICI) targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD-1) proteins transformed the management of advanced cancers. Many tumor-intrinsic factors modulate immunological and clinical responses to such therapies, but ample evidence also implicates the gut microbiome in responses. The gut microbiome, comprising the bacteria, archaea, fungi, and viruses that live in the human digestive tract, is an established determinant of host immunity, but its impact on response to ICI therapy in mice and humans with cancer has only recently been appreciated. Therapeutic interventions to optimize microbiota composition to improve immunotherapy outcomes show promise in mice and humans with cancer. In this review, we discuss the rationale for gut microbiome-based cancer therapies, the results from early-phase clinical trials, and possible future developments.
Collapse
Affiliation(s)
- Diwakar Davar
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hassane M. Zarour
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
33
|
Soleimani N, Javadi MM. Future prospects of bacteria-mediated cancer therapies: Affliction or opportunity? Microb Pathog 2022; 172:105795. [PMID: 36155065 DOI: 10.1016/j.micpath.2022.105795] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/19/2022] [Indexed: 01/10/2023]
Abstract
Cancer, as a disease characterized by uncontrolled growth of cells, is recognized as one of the significant challenges in the field of health and medicine. There are various treatments for cancer like surgery, hormone therapy, chemotherapy, etc., but they have negative effects on the patient's lifestyle. Numerous side effects, and recently the emergence of drug resistance to these methods are weaknesses of these treatments. The utilization of bacteria as a treatment for cancer has attracted scientists' attention in the last decade. There are various methods of using bacteria to treat cancer, including the use of live, attenuated, or genetically engineered microbes, the use of bacterial toxins as an immunotoxin or conjugated to tumor antigens, bacteria-based cancer immunotherapy, bacterial vectors for gene-directed enzyme prodrug, and also the undeniable role of probiotics in treatment, are the cases that today are used for treatment. Bacterial therapy has shown a greater promise in cancer treatment due to its ability to lyse the tumor cells and deliver therapeutic products. However, the potential cytotoxicity of bacteria for healthy tissues, their inability to entirely lyse cancerous cells, and the possibility of mutations in their genomes are among the challenges of bacteriotherapy for cancer. Herein, we summarize the mechanism of bacteria, their potential benefits and harms, and the future of research in this field.
Collapse
Affiliation(s)
- Neda Soleimani
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mahtab Moshref Javadi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
34
|
Ibal JC, Park YJ, Park MK, Lee J, Kim MC, Shin JH. Review of the Current State of Freely Accessible Web Tools for the Analysis of 16S rRNA Sequencing of the Gut Microbiome. Int J Mol Sci 2022; 23:10865. [PMID: 36142775 PMCID: PMC9501225 DOI: 10.3390/ijms231810865] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Owing to the emergence and improvement of high-throughput technology and the associated reduction in costs, next-generation sequencing (NGS) technology has made large-scale sampling and sequencing possible. With the large volume of data produced, the processing and downstream analysis of data are important for ensuring meaningful results and interpretation. Problems in data analysis may be encountered if researchers have little experience in using programming languages, especially if they are clinicians and beginners in the field. A strategy for solving this problem involves ensuring easy access to commercial software and tools. Here, we observed the current status of free web-based tools for microbiome analysis that can help users analyze and handle microbiome data effortlessly. We limited our search to freely available web-based tools and identified MicrobiomeAnalyst, Mian, gcMeta, VAMPS, and Microbiome Toolbox. We also highlighted the various analyses that each web tool offers, how users can analyze their data using each web tool, and noted some of their limitations. From the abovementioned list, gcMeta, VAMPS, and Microbiome Toolbox had several issues that made the analysis more difficult. Over time, as more data are generated and accessed, more users will analyze microbiome data. Thus, the availability of free and easily accessible web tools can enable the easy use and analysis of microbiome data, especially for those users with less experience in using command-line interfaces.
Collapse
Affiliation(s)
- Jerald Conrad Ibal
- NGS Core Facility, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
| | - Yeong-Jun Park
- NGS Core Facility, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
| | - Min-Kyu Park
- NGS Core Facility, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
- Department of Applied Biosciences, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
| | - Jooeun Lee
- NGS Core Facility, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
| | - Min-Chul Kim
- NGS Core Facility, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
- Department of Applied Biosciences, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
| | - Jae-Ho Shin
- NGS Core Facility, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
- Department of Applied Biosciences, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
- Department of Integrative Biotechnology, Kyungpook National University, Daehak-ro 80, Daegu 41566, Korea
| |
Collapse
|
35
|
Liang H, Jo JH, Zhang Z, MacGibeny MA, Han J, Proctor DM, Taylor ME, Che Y, Juneau P, Apolo AB, McCulloch JA, Davar D, Zarour HM, Dzutsev AK, Brownell I, Trinchieri G, Gulley JL, Kong HH. Predicting cancer immunotherapy response from gut microbiomes using machine learning models. Oncotarget 2022; 13:876-889. [PMID: 35875611 PMCID: PMC9295706 DOI: 10.18632/oncotarget.28252] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/20/2022] [Indexed: 01/04/2023] Open
Abstract
Cancer immunotherapy has significantly improved patient survival. Yet, half of patients do not respond to immunotherapy. Gut microbiomes have been linked to clinical responsiveness of melanoma patients on immunotherapies; however, different taxa have been associated with response status with implicated taxa inconsistent between studies. We used a tumor-agnostic approach to find common gut microbiome features of response among immunotherapy patients with different advanced stage cancers. A combined meta-analysis of 16S rRNA gene sequencing data from our mixed tumor cohort and three published immunotherapy gut microbiome datasets from different melanoma patient cohorts found certain gut bacterial taxa correlated with immunotherapy response status regardless of tumor type. Using multivariate selbal analysis, we identified two separate groups of bacterial genera associated with responders versus non-responders. Statistical models of gut microbiome community features showed robust prediction accuracy of immunotherapy response in amplicon sequencing datasets and in cross-sequencing platform validation with shotgun metagenomic datasets. Results suggest baseline gut microbiome features may be predictive of clinical outcomes in oncology patients on immunotherapies, and some of these features may be generalizable across different tumor types, patient cohorts, and sequencing platforms. Findings demonstrate how machine learning models can reveal microbiome-immunotherapy interactions that may ultimately improve cancer patient outcomes.
Collapse
Affiliation(s)
- Hai Liang
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jay-Hyun Jo
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhiwei Zhang
- Biostatistics Branch, Division of Cancer Treatment and Diagnostics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Margaret A. MacGibeny
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Medical Education, West Virginia University, Morgantown, WV 26506, USA
| | - Jungmin Han
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M. Proctor
- Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Monica E. Taylor
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - You Che
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul Juneau
- NIH Library, Division of Library Services, Office of Research Services, NIH, Bethesda, MD 20892, USA
- Zimmerman Associates Inc., Fairfax, VA 22030, USA
| | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - John A. McCulloch
- Genetics and Microbiome Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Diwakar Davar
- Department of Medicine and UPMC Hillman Cancer Center University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hassane M. Zarour
- Department of Medicine and UPMC Hillman Cancer Center University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Amiran K. Dzutsev
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Isaac Brownell
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Center for Immuno-Oncology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - James L. Gulley
- Center for Immuno-Oncology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Heidi H. Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Ting NLN, Lau HCH, Yu J. Cancer pharmacomicrobiomics: targeting microbiota to optimise cancer therapy outcomes. Gut 2022; 71:1412-1425. [PMID: 35277453 PMCID: PMC9185832 DOI: 10.1136/gutjnl-2021-326264] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/25/2022] [Indexed: 02/06/2023]
Abstract
Despite the promising advances in novel cancer therapy such as immune checkpoint inhibitors (ICIs), limitations including therapeutic resistance and toxicity remain. In recent years, the relationship between gut microbiota and cancer has been extensively studied. Accumulating evidence reveals the role of microbiota in defining cancer therapeutic efficacy and toxicity. Unlike host genetics, microbiota can be easily modified via multiple strategies, including faecal microbiota transplantation (FMT), probiotics and antibiotics. Preclinical studies have identified the mechanisms on how microbes influence cancer treatment outcomes. Clinical trials have also demonstrated the potential of microbiota modulation in cancer treatments. Herein, we review the mechanistic insights of gut microbial interactions with chemotherapy and ICIs, particularly focusing on the interplay between gut bacteria and the pharmacokinetics (eg, metabolism, enzymatic degradation) or pharmacodynamics (eg, immunomodulation) of cancer treatment. The translational potential of basic findings in clinical settings is then explored, including using microbes as predictive biomarkers and microbial modulation by antibiotics, probiotics, prebiotics, dietary modulations and FMT. We further discuss the current limitations of gut microbiota modulation in patients with cancer and suggest essential directions for future study. In the era of personalised medicine, it is crucial to understand the microbiota and its interactions with cancer. Manipulating the gut microbiota to augment cancer therapeutic responses can provide new insights into cancer treatment.
Collapse
Affiliation(s)
- Nick Lung-Ngai Ting
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
37
|
Gunjur A, Manrique‐Rincón AJ, Klein O, Behren A, Lawley TD, Welsh SJ, Adams DJ. 'Know thyself' - host factors influencing cancer response to immune checkpoint inhibitors. J Pathol 2022; 257:513-525. [PMID: 35394069 PMCID: PMC9320825 DOI: 10.1002/path.5907] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 11/30/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionised oncology and are now standard-of-care for the treatment of a wide variety of solid neoplasms. However, tumour responses remain unpredictable, experienced by only a minority of ICI recipients across malignancy types. Therefore, there is an urgent need for better predictive biomarkers to identify a priori the patients most likely to benefit from these therapies. Despite considerable efforts, only three such biomarkers are FDA-approved for clinical use, and all rely on the availability of tumour tissue for immunohistochemical staining or genomic assays. There is emerging evidence that host factors - for example, genetic, metabolic, and immune factors, as well as the composition of one's gut microbiota - influence the response of a patient's cancer to ICIs. Tantalisingly, some of these factors are modifiable, paving the way for co-therapies that may enhance the therapeutic index of these treatments. Herein, we review key host factors that are of potential biomarker value for response to ICI therapy, with a particular focus on the proposed mechanisms for these influences. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ashray Gunjur
- Experimental Cancer Genetics, Wellcome Sanger InstituteHinxtonUK,Olivia Newton‐John Cancer Research InstituteLa Trobe University School of Cancer MedicineHeidelbergAustralia
| | - Andrea J Manrique‐Rincón
- Experimental Cancer Genetics, Wellcome Sanger InstituteHinxtonUK,Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of MedicineUniversity of CambridgeCambridgeUK
| | - Oliver Klein
- Olivia Newton‐John Cancer Research InstituteLa Trobe University School of Cancer MedicineHeidelbergAustralia,Department of Medical OncologyAustin HealthHeidelbergAustralia
| | - Andreas Behren
- Olivia Newton‐John Cancer Research InstituteLa Trobe University School of Cancer MedicineHeidelbergAustralia,Department of MedicineUniversity of MelbourneParkvilleAustralia
| | | | - Sarah J Welsh
- Department of SurgeryUniversity of CambridgeCambridgeUK,Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Sanger InstituteHinxtonUK
| |
Collapse
|
38
|
Leung H, Long X, Ni Y, Qian L, Nychas E, Siliceo SL, Pohl D, Hanhineva K, Liu Y, Xu A, Nielsen HB, Belda E, Clément K, Loomba R, Li H, Jia W, Panagiotou G. Risk assessment with gut microbiome and metabolite markers in NAFLD development. Sci Transl Med 2022; 14:eabk0855. [PMID: 35675435 PMCID: PMC9746350 DOI: 10.1126/scitranslmed.abk0855] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A growing body of evidence suggests interplay between the gut microbiota and the pathogenesis of nonalcoholic fatty liver disease (NAFLD). However, the role of the gut microbiome in early detection of NAFLD is unclear. Prospective studies are necessary for identifying reliable, microbiome markers for early NAFLD. We evaluated 2487 individuals in a community-based cohort who were followed up 4.6 years after initial clinical examination and biospecimen sampling. Metagenomic and metabolomic characterizations using stool and serum samples taken at baseline were performed for 90 participants who progressed to NAFLD and 90 controls who remained NAFLD free at the follow-up visit. Cases and controls were matched for gender, age, body mass index (BMI) at baseline and follow-up, and 4-year BMI change. Machine learning models integrating baseline microbial signatures (14 features) correctly classified participants (auROCs of 0.72 to 0.80) based on their NAFLD status and liver fat accumulation at the 4-year follow up, outperforming other prognostic clinical models (auROCs of 0.58 to 0.60). We confirmed the biological relevance of the microbiome features by testing their diagnostic ability in four external NAFLD case-control cohorts examined by biopsy or magnetic resonance spectroscopy, from Asia, Europe, and the United States. Our findings raise the possibility of using gut microbiota for early clinical warning of NAFLD development.
Collapse
Affiliation(s)
- Howell Leung
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Xiaoxue Long
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, 200233 Shanghai, China
| | - Yueqiong Ni
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Beutenbergstraße 11A, 07745 Jena, Germany.,Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, 200233 Shanghai, China.,Corresponding author. (Y.N.); (H.L.); (W.J.); (G.P.)
| | - Lingling Qian
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, 200233 Shanghai, China
| | - Emmanouil Nychas
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Sara Leal Siliceo
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Dennis Pohl
- Clinical Microbiomics, Fruebjergvej 3, 2100 Copenhagen, Denmark.,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Kati Hanhineva
- Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, 20014 Turku, Finland.,Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, 412 96 Gothenburg, Sweden.,School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Yan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China.,Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China.,Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | | | - Eugeni Belda
- Sorbonne Université, INSERM, NutriOmics Research Unit, Nutrition Department, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, 75013 Paris, France
| | - Karine Clément
- Sorbonne Université, INSERM, NutriOmics Research Unit, Nutrition Department, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, 75013 Paris, France
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Huating Li
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, 200233 Shanghai, China.,Corresponding author. (Y.N.); (H.L.); (W.J.); (G.P.)
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, 200233 Shanghai, China.,Corresponding author. (Y.N.); (H.L.); (W.J.); (G.P.)
| | - Gianni Panagiotou
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knöll Institute, Beutenbergstraße 11A, 07745 Jena, Germany.,The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China.,Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Corresponding author. (Y.N.); (H.L.); (W.J.); (G.P.)
| |
Collapse
|
39
|
Rommasi F. Bacterial-Based Methods for Cancer Treatment: What We Know and Where We Are. Oncol Ther 2022; 10:23-54. [PMID: 34780046 PMCID: PMC9098760 DOI: 10.1007/s40487-021-00177-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
A severe disease, cancer is caused by the exponential and uncontrolled growth of cells, leading to organ dysfunction as well as disorders. This disease has been recognized as one of the significant challenges to health and medicine. Various treatment procedures for cancer are associated with diverse side effects; the most conventional cancer treatments include chemotherapy, surgery, and radiotherapy, among others. Numerous adverse and side effects, low specificity and sensitivity, narrow therapeutic windows, and, recently, the emergence of tumor cells resistant to such treatments have been documented as the shortcomings of conventional treatment strategies. As a group of prokaryotic microorganisms, bacteria have great potential for use in cancer therapy. Currently, utilizing bacteria for cancer treatment has attracted the attention of scientists. The high potential of bacteria to become non-pathogenic by genetic manipulation, their distinguished virulence factors (which can be used as weapons against tumors), their ability to proliferate in tissues, and the contingency to control their population by administrating antibiotics, etc., have made bacteria viable candidates and live micro-medication for cancer therapies. However, the possible cytotoxicity impacts of bacteria, their inability to entirely lyse cancerous cells, as well as the probability of mutations in their genomes are among the significant challenges of bacteria-based methods for cancer treatment. In this article, various available data on bacterial therapeutics, along with their pros and cons, are discussed.
Collapse
Affiliation(s)
- Foad Rommasi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
40
|
Rizzo A, Ricci AD. Predictors of response for hepatocellular carcinoma immunotherapy: is there anything on the horizon? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022. [DOI: 10.1080/23808993.2022.2075724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello,” I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Angela Dalia Ricci
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
41
|
Fang C, Fang W, Xu L, Gao F, Hou Y, Zou H, Ma Y, Moll JM, Yang Y, Wang D, Huang Y, Ren H, Zhao H, Qin S, Zhong H, Li J, Liu S, Yang H, Wang J, Brix S, Kristiansen K, Zhang L. Distinct Functional Metagenomic Markers Predict the Responsiveness to Anti-PD-1 Therapy in Chinese Non-Small Cell Lung Cancer Patients. Front Oncol 2022; 12:837525. [PMID: 35530307 PMCID: PMC9069064 DOI: 10.3389/fonc.2022.837525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/21/2022] [Indexed: 12/24/2022] Open
Abstract
Background Programmed death 1 (PD-1) and the ligand of PD-1 (PD-L1) are central targets for immune-checkpoint therapy (ICT) blocking immune evasion-related pathways elicited by tumor cells. A number of PD-1 inhibitors have been developed, but the efficacy of these inhibitors varies considerably and is typically below 50%. The efficacy of ICT has been shown to be dependent on the gut microbiota, and experiments using mouse models have even demonstrated that modulation of the gut microbiota may improve efficacy of ICT. Methods We followed a Han Chinese cohort of 85 advanced non-small cell lung cancer (NSCLC) patients, who received anti-PD-1 antibodies. Tumor biopsies were collected before treatment initiation for whole exon sequencing and variant detection. Fecal samples collected biweekly during the period of anti-PD-1 antibody administration were used for metagenomic sequencing. We established gut microbiome abundance profiles for identification of significant associations between specific microbial taxa, potential functionality, and treatment responses. A prediction model based on random forest was trained using selected markers discriminating between the different response groups. Results NSCLC patients treated with antibiotics exhibited the shortest survival time. Low level of tumor-mutation burden and high expression level of HLA-E significantly reduced progression-free survival. We identified metagenomic species and functional pathways that differed in abundance in relation to responses to ICT. Data on differential enrichment of taxa and predicted microbial functions in NSCLC patients responding or non-responding to ICT allowed the establishment of random forest algorithm-adopted models robustly predicting the probability of whether or not a given patient would benefit from ICT. Conclusions Overall, our results identified links between gut microbial composition and immunotherapy efficacy in Chinese NSCLC patients indicating the potential for such analyses to predict outcome prior to ICT.
Collapse
Affiliation(s)
- Chao Fang
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- BGI-Shenzhen, Shenzhen, China
| | - Wenfeng Fang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Liqin Xu
- BGI-Shenzhen, Shenzhen, China
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Fangfang Gao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yong Hou
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- BGI-Shenzhen, Shenzhen, China
| | - Hua Zou
- BGI-Shenzhen, Shenzhen, China
| | - Yuxiang Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Janne Marie Moll
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Yunpeng Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | - Yan Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Huahui Ren
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- BGI-Shenzhen, Shenzhen, China
| | - Hongyun Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | - Huanzi Zhong
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- BGI-Shenzhen, Shenzhen, China
| | - Junhua Li
- BGI-Shenzhen, Shenzhen, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | | | - Huanming Yang
- BGI-Shenzhen, Shenzhen, China
- James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, China
- James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
- *Correspondence: Susanne Brix, ; Karsten Kristiansen, ; Li Zhang,
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- BGI-Shenzhen, Shenzhen, China
- Institute of Metagenomics, Qingdao-Europe Advance Institute for Life Sciences, BGI-Qingdao, Qingdao, China
- *Correspondence: Susanne Brix, ; Karsten Kristiansen, ; Li Zhang,
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- *Correspondence: Susanne Brix, ; Karsten Kristiansen, ; Li Zhang,
| |
Collapse
|
42
|
Abstract
Microorganisms within the gut and other niches may contribute to carcinogenesis, as well as shaping cancer immunosurveillance and response to immunotherapy. Our understanding of the complex relationship between different host-intrinsic microorganisms, as well as the multifaceted mechanisms by which they influence health and disease, has grown tremendously-hastening development of novel therapeutic strategies that target the microbiota to improve treatment outcomes in cancer. Accordingly, the evaluation of a patient's microbial composition and function and its subsequent targeted modulation represent key elements of future multidisciplinary and precision-medicine approaches. In this Review, we outline the current state of research toward harnessing the microbiome to better prevent and treat cancer.
Collapse
|
43
|
The Implication of Gastric Microbiome in the Treatment of Gastric Cancer. Cancers (Basel) 2022; 14:cancers14082039. [PMID: 35454944 PMCID: PMC9028069 DOI: 10.3390/cancers14082039] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Gastric cancer (GC) represents the fifth most common cancer worldwide. Recent developments in PCR and metagenomics clarify that the stomach contains a powerful microbiota. Conventional treatments for GC that include surgery, chemotherapy, and radiotherapy are not very effective. That’s why new therapeutic strategies are needed. The intestinal microbiota is involved in oncogenesis and cancer prevention, and the effectiveness of chemotherapy. Recent studies have shown that certain bacteria may enhance the effect of some traditional antineoplastic drugs and immunotherapies. Abstract Gastric cancer (GC) is one of the most common and deadly malignancies worldwide. Helicobacter pylori have been documented as a risk factor for GC. The development of sequencing technology has broadened the knowledge of the gastric microbiome, which is essential in maintaining homeostasis. Recent studies have demonstrated the involvement of the gastric microbiome in the development of GC. Therefore, the elucidation of the mechanism by which the gastric microbiome contributes to the development and progression of GC may improve GC’s prevention, diagnosis, and treatment. In this review, we discuss the current knowledge about changes in gastric microbial composition in GC patients, their role in carcinogenesis, the possible therapeutic role of the gastric microbiome, and its implications for current GC therapy.
Collapse
|
44
|
Lin Y, Kong DX, Zhang YN. Does the Microbiota Composition Influence the Efficacy of Colorectal Cancer Immunotherapy? Front Oncol 2022; 12:852194. [PMID: 35463305 PMCID: PMC9023803 DOI: 10.3389/fonc.2022.852194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the second most common malignancy globally, and many people with CRC suffer the fate of death. Due to the importance of CRC and its negative impact on communities, treatment strategies to control it or increase patient survival are being studied. Traditional therapies, including surgery and chemotherapy, have treated CRC patients. However, with the advancement of science, we are witnessing the emergence of novel therapeutic approaches such as immunotherapy for CRC treatment, which have had relatively satisfactory clinical outcomes. Evidence shows that gastrointestinal (GI) microbiota, including various bacterial species, viruses, and fungi, can affect various biological events, regulate the immune system, and even treat diseases like human malignancies. CRC has recently shown that the gut microorganism pattern can alter both antitumor and pro-tumor responses, as well as cancer immunotherapy. Of course, this is also true of traditional therapies because it has been revealed that gut microbiota can also reduce the side effects of chemotherapy. Therefore, this review summarized the effects of gut microbiota on CRC immunotherapy.
Collapse
Affiliation(s)
- Yan Lin
- Health Management Center, Department of General Practice, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Yan Lin, ; You-Ni Zhang,
| | - De-Xia Kong
- Health Management Center, Department of General Practice, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - You-Ni Zhang
- Department of Laboratory Medicine, Tiantai People’s Hospital, Taizhou, China
- *Correspondence: Yan Lin, ; You-Ni Zhang,
| |
Collapse
|
45
|
Cascone T, Chelvanambi M, Wargo JA. Immunotherapy response-associated Akkermansia: canary in a coal mine? Trends Immunol 2022; 43:337-339. [DOI: 10.1016/j.it.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
|
46
|
Rizzo A, Cusmai A, Gadaleta-Caldarola G, Palmiotti G. Which role for predictors of response to immune checkpoint inhibitors in hepatocellular carcinoma? Expert Rev Gastroenterol Hepatol 2022; 16:333-339. [PMID: 35403533 DOI: 10.1080/17474124.2022.2064273] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) remains a frequently diagnosed malignancy worldwide, still representing an important cause of cancer-related death. Recent years have seen the emergence of novel systemic treatments for HCC patients, including immune checkpoint inhibitors (ICIs). Nonetheless, several questions regarding HCC immunotherapy remain unanswered, especially in terms of biochemical predictors of response. AREAS COVERED In the current paper, we will discuss available evidence regarding predictive biomarkers of response to HCC immunotherapy. A literature search was conducted in January 2022 of Pubmed/Medline, Cochrane library, and Scopus databases. EXPERT OPINION The identification of predictive biomarkers represents an unmet need in HCC patients receiving ICIs. The HCC medical community is called to further efforts aimed to elucidate the effective role of PD-L1 expression, TMB, MSI, gut microbiota, and other emerging biomarkers.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico "Don Tonino Bello," I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Antonio Cusmai
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico "Don Tonino Bello," I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Gennaro Gadaleta-Caldarola
- Medical Oncology Unit, 'Mons. R. Dimiccoli' Hospital, Barletta (BT), Azienda Sanitaria Locale Barletta, 76121, Italy
| | - Gennaro Palmiotti
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico "Don Tonino Bello," I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| |
Collapse
|
47
|
Newsome RC, Gharaibeh RZ, Pierce CM, da Silva WV, Paul S, Hogue SR, Yu Q, Antonia S, Conejo-Garcia JR, Robinson LA, Jobin C. Interaction of bacterial genera associated with therapeutic response to immune checkpoint PD-1 blockade in a United States cohort. Genome Med 2022; 14:35. [PMID: 35346337 PMCID: PMC8961902 DOI: 10.1186/s13073-022-01037-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/08/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Recent studies show that human gut microbial composition can determine whether a patient is a responder or non-responder to immunotherapy but have not identified a common microbial signal shared by responding patients. The functional relationship between immunity, intestinal microbiota, and NSCLC response to immune checkpoint inhibitor/inhibition (ICI) in an American cohort remains unexplored. METHODS RNAlater-preserved fecal samples were collected from 65 pre-treatment (baseline) and post-treatment stage III/IV NSCLC patients undergoing ICI therapy, categorized as responders or non-responders according to RECIST criteria. Pooled and individual responder and non-responder microbiota were transplanted into a gnotobiotic mouse model of lung cancer and treated with ICIs. 16S rDNA and RNA sequencing was performed on patient fecal samples, 16S rDNA sequencing on mouse fecal samples, and flow cytometric analysis on mouse tumor tissue. RESULTS Responder patients have both a different microbial community structure than non-responders (P = 0.004) and a different bacterial transcriptome (PC2 = 0.03) at baseline. Taxa significantly enriched in responders include amplicon sequence variants (ASVs) belonging to the genera Ruminococcus, Akkermansia, and Faecalibacterium. Pooled and individual responder microbiota transplantation into gnotobiotic mice decreased tumor growth compared to non-responder colonized mice following ICI (P = 0.023, P = 0.019, P = 0.008, respectively). Responder tumors showed an increased anti-tumor cellular phenotype following ICI treatment. Responder mice are enriched with ASVs belonging to the genera Bacteroides, Blautia, Akkermansia, and Faecalibacterium. Overlapping taxa mapping between human and mouse cohorts correlated with tumor size and weight revealed a network highlighting responder-associated ASVs belonging to the genera Colidextribacter, Frisingicoccus, Marvinbryantia, and Blautia which have not yet been reported. CONCLUSIONS The role of isolate-specific function and bacterial gene expression in gut microbial-driven responsiveness to ICI has been underappreciated. This work supports further investigation using isolate-driven models to characterize the mechanisms underlying this phenomenon.
Collapse
Affiliation(s)
- Rachel C Newsome
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Raad Z Gharaibeh
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christine M Pierce
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
- Present Address: Department of Epidemiology, BARDS, MRL, Merck Sharp & Dohme Corp., a Subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Shirlene Paul
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Stephanie R Hogue
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Qin Yu
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Scott Antonia
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Lary A Robinson
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Christian Jobin
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA.
- Department of Infectious Diseases and Immunology, University of Florida College of Medicine, Gainesville, FL, USA.
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
48
|
You L, Zhou J, Xin Z, Hauck JS, Na F, Tang J, Zhou X, Lei Z, Ying B. Novel directions of precision oncology: circulating microbial DNA emerging in cancer-microbiome areas. PRECISION CLINICAL MEDICINE 2022; 5:pbac005. [PMID: 35692444 PMCID: PMC9026200 DOI: 10.1093/pcmedi/pbac005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Microbiome research has extended into the cancer area in the past decades. Microbes can affect oncogenesis, progression, and treatment response through various mechanisms, including direct regulation and indirect impacts. Microbiota-associated detection methods and agents have been developed to facilitate cancer diagnosis and therapy. Additionally, the cancer microbiome has recently been redefined. The identification of intra-tumoral microbes and cancer-related circulating microbial DNA (cmDNA) has promoted novel research in the cancer-microbiome area. In this review, we define the human system of commensal microbes and the cancer microbiome from a brand-new perspective and emphasize the potential value of cmDNA as a promising biomarker in cancer liquid biopsy. We outline all existing studies on the relationship between cmDNA and cancer and the outlook for potential preclinical and clinical applications of cmDNA in cancer precision medicine, as well as critical problems to be overcome in this burgeoning field.
Collapse
Affiliation(s)
- Liting You
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhaodan Xin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - J Spencer Hauck
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Feifei Na
- Department of Thoracic Cancer, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Tang
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000,China
| | - Xiaohan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zichen Lei
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
49
|
McCulloch JA, Davar D, Rodrigues RR, Badger JH, Fang JR, Cole AM, Balaji AK, Vetizou M, Prescott SM, Fernandes MR, Costa RGF, Yuan W, Salcedo R, Bahadiroglu E, Roy S, DeBlasio RN, Morrison RM, Chauvin JM, Ding Q, Zidi B, Lowin A, Chakka S, Gao W, Pagliano O, Ernst SJ, Rose A, Newman NK, Morgun A, Zarour HM, Trinchieri G, Dzutsev AK. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med 2022; 28:545-556. [PMID: 35228752 PMCID: PMC10246505 DOI: 10.1038/s41591-022-01698-2] [Citation(s) in RCA: 209] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 01/13/2022] [Indexed: 12/12/2022]
Abstract
Ample evidence indicates that the gut microbiome is a tumor-extrinsic factor associated with antitumor response to anti-programmed cell death protein-1 (PD-1) therapy, but inconsistencies exist between published microbial signatures associated with clinical outcomes. To resolve this, we evaluated a new melanoma cohort, along with four published datasets. Time-to-event analysis showed that baseline microbiota composition was optimally associated with clinical outcome at approximately 1 year after initiation of treatment. Meta-analysis and other bioinformatic analyses of the combined data show that bacteria associated with favorable response are confined within the Actinobacteria phylum and the Lachnospiraceae/Ruminococcaceae families of Firmicutes. Conversely, Gram-negative bacteria were associated with an inflammatory host intestinal gene signature, increased blood neutrophil-to-lymphocyte ratio, and unfavorable outcome. Two microbial signatures, enriched for Lachnospiraceae spp. and Streptococcaceae spp., were associated with favorable and unfavorable clinical response, respectively, and with distinct immune-related adverse effects. Despite between-cohort heterogeneity, optimized all-minus-one supervised learning algorithms trained on batch-corrected microbiome data consistently predicted outcomes to programmed cell death protein-1 therapy in all cohorts. Gut microbial communities (microbiotypes) with nonuniform geographical distribution were associated with favorable and unfavorable outcomes, contributing to discrepancies between cohorts. Our findings shed new light on the complex interaction between the gut microbiome and response to cancer immunotherapy, providing a roadmap for future studies.
Collapse
Affiliation(s)
- John A McCulloch
- Genetics and Microbiome Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Diwakar Davar
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Richard R Rodrigues
- Genetics and Microbiome Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jonathan H Badger
- Genetics and Microbiome Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jennifer R Fang
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alicia M Cole
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ascharya K Balaji
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Marie Vetizou
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Stephanie M Prescott
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Miriam R Fernandes
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Raquel G F Costa
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wuxing Yuan
- Genetics and Microbiome Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Rosalba Salcedo
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Erol Bahadiroglu
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Soumen Roy
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Richelle N DeBlasio
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert M Morrison
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joe-Marc Chauvin
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Quanquan Ding
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bochra Zidi
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ava Lowin
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Saranya Chakka
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wentao Gao
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ornella Pagliano
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scarlett J Ernst
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy Rose
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nolan K Newman
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Hassane M Zarour
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Giorgio Trinchieri
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Amiran K Dzutsev
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
50
|
Newsome RC, Yang Y, Jobin C. The microbiome, gastrointestinal cancer, and immunotherapy. J Gastroenterol Hepatol 2022; 37:263-272. [PMID: 34820895 PMCID: PMC9922516 DOI: 10.1111/jgh.15742] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract greatly contributes to global cancer burden and cancer-related deaths. The microbiota represents the population of microorganisms that live in and around the body, located primarily in the gastrointestinal tract. The microbiota has been implicated in colorectal cancer development and progression, but its role in cancer therapy for the gastrointestinal tract is less defined, especially for extra-intestinal cancers. In this review, we discuss the past 5 years of research into microbial involvement in immune-related therapies for colorectal, pancreatic, hepatic, and gastric cancers, with the goal of highlighting recent advances and new areas for investigation in this field.
Collapse
Affiliation(s)
- Rachel C Newsome
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ye Yang
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
| | - Christian Jobin
- Departments of Medicine, University of Florida, Gainesville, Florida, USA
- Infectious Diseases and Immunology, University of Florida, Gainesville, Florida, USA
- Anatomy and Cell Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|