651
|
Host immunity modulates the efficacy of microbiota transplantation for treatment of Clostridioides difficile infection. Nat Commun 2021; 12:755. [PMID: 33531483 PMCID: PMC7854624 DOI: 10.1038/s41467-020-20793-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 12/21/2020] [Indexed: 01/30/2023] Open
Abstract
Fecal microbiota transplantation (FMT) is a successful therapeutic strategy for treating recurrent Clostridioides difficile infection. Despite remarkable efficacy, implementation of FMT therapy is limited and the mechanism of action remains poorly understood. Here, we demonstrate a critical role for the immune system in supporting FMT using a murine C. difficile infection system. Following FMT, Rag1 heterozygote mice resolve C. difficile while littermate Rag1-/- mice fail to clear the infection. Targeted ablation of adaptive immune cell subsets reveal a necessary role for CD4+ Foxp3+ T-regulatory cells, but not B cells or CD8+ T cells, in FMT-mediated resolution of C. difficile infection. FMT non-responsive mice exhibit exacerbated inflammation, impaired engraftment of the FMT bacterial community and failed restoration of commensal bacteria-derived secondary bile acid metabolites in the large intestine. These data demonstrate that the host's inflammatory immune status can limit the efficacy of microbiota-based therapeutics to treat C. difficile infection.
Collapse
|
652
|
Huang M, Lu JJ, Ding J. Natural Products in Cancer Therapy: Past, Present and Future. NATURAL PRODUCTS AND BIOPROSPECTING 2021; 11:5-13. [PMID: 33389713 PMCID: PMC7933288 DOI: 10.1007/s13659-020-00293-7] [Citation(s) in RCA: 266] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/15/2020] [Indexed: 05/02/2023]
Abstract
Natural products, with remarkable chemical diversity, have been extensively investigated for their anticancer potential for more than a half-century. The collective efforts of the community have achieved the tremendous advancements, bringing natural products to clinical use and discovering new therapeutic opportunities, yet the challenges remain ahead. With remarkable changes in the landscape of cancer therapy and growing role of cutting-edge technologies, we may have come to a crossroads to revisit the strategies to understand nature products and to explore their therapeutic utility. This review summarizes the key advancements in nature product-centered cancer research and calls for the implementation of systematic approaches, new pharmacological models, and exploration of emerging directions to revitalize natural products search in cancer therapy.
Collapse
Affiliation(s)
- Min Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jian Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
653
|
Oliva M, Mulet-Margalef N, Ochoa-De-Olza M, Napoli S, Mas J, Laquente B, Alemany L, Duell EJ, Nuciforo P, Moreno V. Tumor-Associated Microbiome: Where Do We Stand? Int J Mol Sci 2021; 22:1446. [PMID: 33535583 PMCID: PMC7867144 DOI: 10.3390/ijms22031446] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
The study of the human microbiome in oncology is a growing and rapidly evolving field. In the past few years, there has been an exponential increase in the number of studies investigating associations of microbiome and cancer, from oncogenesis and cancer progression to resistance or sensitivity to specific anticancer therapies. The gut microbiome is now known to play a significant role in antitumor immune responses and in predicting the efficacy of immune-checkpoint inhibitors in cancer patients. Beyond the gut, the tumor-associated microbiome-microbe communities located either in the tumor or within its body compartment-seems to interact with the local microenvironment and the tumor immune contexture, ultimately impacting cancer progression and treatment outcome. However, pre-clinical research focusing on causality and mechanistic pathways as well as proof-of-concept studies are still needed to fully understand the potential clinical utility of microbiome in cancer patients. Moreover, there is a need for the standardization of methodology and the implementation of quality control across microbiome studies to allow for a better interpretation and greater comparability of the results reported between them. This review summarizes the accumulating evidence in the field and discusses the current and upcoming challenges of microbiome studies.
Collapse
Affiliation(s)
- Marc Oliva
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Nuria Mulet-Margalef
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Maria Ochoa-De-Olza
- Service of Immuno-Oncology, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland;
- Ludwig Institute for Cancer Research, University of Lausanne, 1066 Lausanne, Switzerland
| | - Stefania Napoli
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (S.N.); (P.N.)
| | - Joan Mas
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| | - Berta Laquente
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Laia Alemany
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
- Cancer Epidemiology Research Program, Catalan Institute of Oncology, L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- EPIBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
| | - Eric J. Duell
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (S.N.); (P.N.)
| | - Victor Moreno
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| |
Collapse
|
654
|
McPherson AC, Pandey SP, Bender MJ, Meisel M. Systemic Immunoregulatory Consequences of Gut Commensal Translocation. Trends Immunol 2021; 42:137-150. [PMID: 33422410 PMCID: PMC10110348 DOI: 10.1016/j.it.2020.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
One major determinant of systemic immunity during homeostasis and in certain complex multifactorial diseases (e.g. cancer and autoimmune conditions), is the gut microbiota. These commensals can shape systemic immune responses via translocation of metabolites, microbial cell wall components, and viable microbes. In the last few years, bacterial translocation has revealed itself as playing a key, and potentially causal role in mediating immunomodulatory processes in nongastrointestinal diseases. Moreover, recent observations regarding the presence of complex microbial communities and viable bacteria within gut-distal tissues during homeostasis challenge the current paradigm that healthy mammals are entirely sterile at nonmucosal sites. This review discusses our current understanding of how the gut microbiota orchestrates systemic immunity during noninfectious extraintestinal diseases and homeostasis, focusing on the translocation of viable bacteria to gut-distal sites.
Collapse
Affiliation(s)
- Alex C McPherson
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Surya P Pandey
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mackenzie J Bender
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marlies Meisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
655
|
Albracht-Schulte K, Islam T, Johnson P, Moustaid-Moussa N. Systematic Review of Beef Protein Effects on Gut Microbiota: Implications for Health. Adv Nutr 2021; 12:102-114. [PMID: 32761179 PMCID: PMC7850003 DOI: 10.1093/advances/nmaa085] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/10/2020] [Accepted: 06/24/2020] [Indexed: 01/07/2023] Open
Abstract
The influence of diet on the gut microbiota is an emerging research area with significant impact on human health and disease. However, the effects of beef, the most consumed red meat in the United States, on gut microbial profile are not well studied. Following Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols, the objective of this systematic review was to conduct a rigorous and thorough review of the current scientific literature regarding the effects of beef protein and the resulting bioactivity of beef protein and amino acids on the gut microbiota, with the goal of identifying gaps in the literature and guiding future research priorities. Utilizing MEDLINE Complete, PubMed, ScienceDirect, Scopus, and Google Scholar databases, we conducted searches including terms and combinations of the following: animal protein, amino acid, beef, bioactive compounds, diet, health, microbiome, peptide, processed beef, and protein. We identified 131 articles, from which 15 were included in our review. The effects of beef on mouse and rat models were mostly consistent for the bacterial phylum level. Short-term (1-4-wk) beef intakes had little to no effect on microbial profiles in humans. Most studies utilized high beef feeding (240-380 g/d), and no study examined recommended amounts of protein [∼3.71 oz/d (105 g/d) meats, poultry, and eggs, or ∼26 oz/week (737 g/wk) from these food sources] according to US dietary guidelines. Additionally, the majority of animal and human studies with adverse findings examined the impact of beef in the context of a diet high in fat or sugar. In conclusion, an extensive gap exists in the literature regarding beef and the microbiota. More studies are necessary to elucidate the role of the microbiota following the consumption of beef, especially in interaction with other dietary compounds, and how beef preparation, processing, and cooking methods differentially influence the biological effects of beef on human health.
Collapse
Affiliation(s)
- Kembra Albracht-Schulte
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Tariful Islam
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Paige Johnson
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
656
|
Yamamura R, Ooshio T, Sonoshita M. Tiny Drosophila makes giant strides in cancer research. Cancer Sci 2021; 112:505-514. [PMID: 33275812 PMCID: PMC7893992 DOI: 10.1111/cas.14747] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer burden has been increasing worldwide, making cancer the second leading cause of death in the world. Over the past decades, various experimental models have provided important insights into the nature of cancer. Among them, the fruit fly Drosophila as a whole-animal toolkit has made a decisive contribution to our understanding of fundamental mechanisms of cancer development including loss of cell polarity. In recent years, scalable Drosophila platforms have proven useful also in developing anti-cancer regimens that are effective not only in mammalian models but also in patients. Here, we review studies using Drosophila as a tool to advance cancer study by complementing other traditional research systems.
Collapse
Affiliation(s)
- Ryodai Yamamura
- Division of Biomedical OncologyInstitute for Genetic MedicineHokkaido UniversitySapporoJapan
| | - Takako Ooshio
- Division of Biomedical OncologyInstitute for Genetic MedicineHokkaido UniversitySapporoJapan
| | - Masahiro Sonoshita
- Division of Biomedical OncologyInstitute for Genetic MedicineHokkaido UniversitySapporoJapan
- Global Station for Biosurfaces and Drug DiscoveryHokkaido UniversitySapporoJapan
| |
Collapse
|
657
|
Han C, Song J, Hu J, Fu H, Feng Y, Mu R, Xing Z, Wang Z, Wang L, Zhang J, Wang C, Dong L. Smectite promotes probiotic biofilm formation in the gut for cancer immunotherapy. Cell Rep 2021; 34:108706. [PMID: 33567279 DOI: 10.1016/j.celrep.2021.108706] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/12/2020] [Accepted: 01/08/2021] [Indexed: 12/17/2022] Open
Abstract
Administration of probiotics to regulate the immune system is a potential anti-tumor strategy. However, oral administration of probiotics is ineffective because of the poor inhabitation of exogenous bacteria in host intestines. Here we report that smectite, a type of mineral clay and established anti-diarrhea drug, promotes expansion of probiotics (especially Lactobacillus) in the murine gut and subsequently elicits anti-tumor immune responses. The ion-exchangeable microstructure of smectite preferentially promotes lactic acid bacteria (LABs) to form biofilms on smectite in vitro and in vivo. In mouse models, smectite laden with LAB biofilms (Lactobacillus and Bifidobacterium) inhibits tumor growth (when used alone) and enhances the efficacy of chemotherapy or immunotherapy (when used in combination with either of them) by activating dendritic cells (DCs) via Toll-like receptor 2 (TLR2) signaling. Our findings suggest oral administration of smectite as a promising strategy to enrich probiotics in vivo for cancer immunotherapy.
Collapse
Affiliation(s)
- Congwei Han
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Jinji Song
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Junqing Hu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Huijie Fu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yanxian Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR
| | - Ruoyu Mu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR
| | - Zhen Xing
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Zhenzhen Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR; Medical School of Nanjing University, Nanjing, Jiangsu 21093, China
| | - Lintao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China; Medical School of Nanjing University, Nanjing, Jiangsu 21093, China.
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR.
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China; Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
658
|
Yu Q, Jobin C, Thomas RM. Implications of the microbiome in the development and treatment of pancreatic cancer: Thinking outside of the box by looking inside the gut. Neoplasia 2021; 23:246-256. [PMID: 33418277 PMCID: PMC7804346 DOI: 10.1016/j.neo.2020.12.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/19/2022]
Abstract
Pancreatic ductal adenocarcinoma is the third leading cause of cancer-related death in the United States. As one of the most lethal cancer types, the prognosis for patients diagnosed with pancreatic cancer remains dismal and novel investigations are urgently needed. Evidence for an association of microbes with pancreatic cancer risk, development, treatment response, and post-treatment survivorship is rapidly developing. Herein, we provide an overview on the role of the microbiome as it relates to the natural history of pancreatic cancer, including host immune interactions, alterations in metabolism, direct carcinogenic effect, and its role in treatment response.
Collapse
Affiliation(s)
- Qin Yu
- Department of Medicine, University of Florida College of Medicine, Gainesville, 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
| | - Ryan M Thomas
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA; Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
659
|
Impacts of gut microbiota on gestational diabetes mellitus: a comprehensive review. Eur J Nutr 2021; 60:2343-2360. [PMID: 33512587 DOI: 10.1007/s00394-021-02483-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is a condition that seriously threatens mother and child health. The incidence of GDM has increased worldwide in the past decades. In addition, the complications of GDM such as type 2 diabetes (T2DM) and neonatal malformations could negatively affect the living quality of mothers and their children. AIM It has been widely known that the imbalance of gut microbiota or called 'gut dysbiosis' plays a key role in the development of insulin resistance and chronic low-grade inflammation in T2DM patients. However, the impacts of gut microbiota on GDM remain controversial. Here, we aim to comprehensively review the alterations of gut microbiota in GDM mothers and their offspring. RESULTS The alterations of Firmicutes/Bacteroidetes (F/B) ratio, short-chain fatty acid (SCFA)-producing bacteria, bacteria with probiotics properties and gram-negative lipopolysaccharide (LPS)-producing bacteria play a vital role in the development of GDM. The beneficial roles of gut microbiota modification (probiotics, synbiotics and lifestyle modification) as a treatment of GDM were found in some, but not all studies. CONCLUSION In the near future, gut microbiota modification may be considered as one of the standard treatments for GDM. Moreover, further studies regarding the specific gut microbiota that are associated with the early development of GDM are required. This may contribute to the novel diagnostic markers for early stages of GDM.
Collapse
|
660
|
Saeedi-Boroujeni A, Mahmoudian-Sani MR. Anti-inflammatory potential of Quercetin in COVID-19 treatment. J Inflamm (Lond) 2021; 18:3. [PMID: 33509217 PMCID: PMC7840793 DOI: 10.1186/s12950-021-00268-6] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a betacoronavirus causing severe inflammatory pneumonia, so that excessive inflammation is considered a risk factor for the disease. According to reports, cytokine storm is strongly responsible for death in such patients. Some of the consequences of severe inflammation and cytokine storms include acute respiratory distress syndrome, acute lung injury, and multiple organ dysfunction syndromes. Phylogenetic findings show more similarity of the SARS-CoV-2 virus with bat coronaviruses, and less with SARS-CoV. Quercetin is a carbohydrate-free flavonoid that is the most abundant flavonoid in vegetables and fruits and has been the most studied to determine the biological effects of flavonoids. Inflammasomes are cytosolic multi-protein complexes assembling in response to cytosolic PAMP and DAMPs, whose function is to generate active forms of cytokines IL-1β and IL-18. Activation or inhibition of the NLRP3 inflammasome is affected by regulators such as TXNIP, SIRT1 and NRF2. Quercetin suppresses the NLRP3 inflammasome by affecting these regulators. Quercetin, as an anti-inflammatory, antioxidant, analgesic and inflammatory compound, is probably a potential treatment for severe inflammation and one of the main life-threatening conditions in patients with COVID-19.
Collapse
Affiliation(s)
- Ali Saeedi-Boroujeni
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical, Sciences, Ahvaz, Iran
- Abadan School of Medical Sciences, Abadan, Iran
- ImmunologyToday, Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad-Reza Mahmoudian-Sani
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
661
|
Masetti R, Zama D, Leardini D, Muratore E, Turroni S, Brigidi P, Pession A. Microbiome-Derived Metabolites in Allogeneic Hematopoietic Stem Cell Transplantation. Int J Mol Sci 2021; 22:1197. [PMID: 33530464 PMCID: PMC7865777 DOI: 10.3390/ijms22031197] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
The gut microbiome has emerged as a major character in the context of hematopoietic stem cell transplantation. The biology underpinning this relationship is still to be defined. Recently, mounting evidence has suggested a role for microbiome-derived metabolites in mediating crosstalk between intestinal microbial communities and the host. Some of these metabolites, such as fiber-derived short-chain fatty acids or amino acid-derived compounds, were found to have a role also in the transplant setting. New interesting data have been published on this topic, posing a new intriguing perspective on comprehension and treatment. This review provides an updated comprehensive overview of the available evidence in the field of gut microbiome-derived metabolites and hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Riccardo Masetti
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (R.M.); (D.Z.); (E.M.); (A.P.)
| | - Daniele Zama
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (R.M.); (D.Z.); (E.M.); (A.P.)
| | - Davide Leardini
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (R.M.); (D.Z.); (E.M.); (A.P.)
| | - Edoardo Muratore
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (R.M.); (D.Z.); (E.M.); (A.P.)
| | - Silvia Turroni
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy;
| | - Andrea Pession
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (R.M.); (D.Z.); (E.M.); (A.P.)
| |
Collapse
|
662
|
Chambers LM, Michener CM, Rose PG, Reizes O, Yao M, Vargas R. Impact of antibiotic treatment on immunotherapy response in women with recurrent gynecologic cancer. Gynecol Oncol 2021; 161:211-220. [PMID: 33504455 DOI: 10.1016/j.ygyno.2021.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/17/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE(S) To identify whether antibiotics (ABX) impact immunotherapy (ICI) response rate (RR), progression-free survival (PFS), and overall survival (OS) in women with recurrent endometrial (EC), cervical (CC) and ovarian cancer (OC). METHODS This retrospective cohort study included women with recurrent EC, CC, and OC treated with ICIs from 1/1/17-9/1/2020. ABX were defined as 30 days before (pABX) or concurrently (cABX) with ICI. The impact of ABX upon PFS and OS was assessed by univariate analysis and multivariable Cox regression. RESULTS Of 101 women, 52.5% (n = 53) had recurrent EC, 21.4% (n = 22) CC and 25.7% (n = 26) OC. 56.9% (n = 58) received ABX, with 22.8% (n = 23) pABX and 46.5% (n = 47) cABX. While no difference was observed in ICI RR for any ABX vs. none (p = 0.89) and cABX vs. none (p = 0.33), pABX (n = 23) were associated with decreased RR vs. none (n = 78) (Partial Response - 8.7% vs. 30.8%; Complete Response - 4.3% vs. 9.0%; p = 0.002). On univariate analysis, pABX were associated with worsened PFS (2.9 vs. 8.9 months; HR 2.53, 95% CI 1.48-4.31, p < 0.001) and OS (9.3 vs. 19.9 months; HR 2.29, 95% CI 1.22-4.32, p = 0.01). No PFS or OS difference was noted for cABX (PFS - 9.3 vs. 6.0 months; HR 0.70, 95% CI 0.43-1.12; p = 0.14; OS - 13.4 vs. 16.3 months; HR 0.89, 95% CI 0.51-1.54; p = 0.68). On multivariable analysis, pABX were associated with significantly decreased PFS (HR 3.10, 95% CI 1.75-5.49, p < 0.001) and OS (HR 3.03, 95% CI 1.50-6.10, p = 0.002). CONCLUSIONS In women with recurrent EC, OC, and CC receiving ICI, pABX, but not cABX, are associated with decreased RR, PFS, and OS. Further investigation is warranted to understand predictors of ICI response in women with gynecologic cancer.
Collapse
Affiliation(s)
- Laura M Chambers
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women's Health Institute, Cleveland Clinic, Desk A81, 9500 Euclid Avenue, Cleveland, OH 44195, United States of America.
| | - Chad M Michener
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women's Health Institute, Cleveland Clinic, Desk A81, 9500 Euclid Avenue, Cleveland, OH 44195, United States of America
| | - Peter G Rose
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women's Health Institute, Cleveland Clinic, Desk A81, 9500 Euclid Avenue, Cleveland, OH 44195, United States of America
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Science, Cleveland Clinic Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Meng Yao
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Roberto Vargas
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women's Health Institute, Cleveland Clinic, Desk A81, 9500 Euclid Avenue, Cleveland, OH 44195, United States of America
| |
Collapse
|
663
|
Khalyfa A, Ericsson A, Qiao Z, Almendros I, Farré R, Gozal D. Circulating exosomes and gut microbiome induced insulin resistance in mice exposed to intermittent hypoxia: Effects of physical activity. EBioMedicine 2021; 64:103208. [PMID: 33485839 PMCID: PMC7910674 DOI: 10.1016/j.ebiom.2021.103208] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/27/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023] Open
Abstract
Background Gut microbiota (GM) contribute to obesity and insulin resistance (IR). Obstructive sleep apnea (OSA), characterized by intermittent hypoxia (IH), promotes IR and alters GM. Since circulating exosomes are implicated in IR, we examined the effects of IH and physical activity (PA) in mice on GM, colonic epithelium permeability, systemic IR, and plasma exosome cargo, and exosome effects on visceral white adipose tissues (vWAT) IR. Methods C57BL/6 mice were exposed to IH or room air (RA) for 6 weeks with and without PA (n = 12/group), and GM and systemic IR changes were assessed, as well as the effects of plasma exosomes on naïve adipocyte insulin sensitivity. Fecal microbiota transfers (FMT) were performed in naïve mice (n = 5/group), followed by fecal 16S rRNA sequencing, and systemic IR and exosome-induced effects on adipocyte insulin sensitivity were evaluated. Findings Principal coordinate analysis (PCoA) ordinates revealed B-diversity among IH and FMT recipients that accounted for 64% principal component 1 (PC1) and 12.5% (PC2) of total variance. Dominant microbiota families and genera in IH-exposed and FMT-treated were preserved, and IH-exposed GM and IH-FMT induced increased gut permeability. Plasma exosomes from IH-exposed and IH-FMT mice decreased pAKT/AKT responses to exogenous insulin in adipocytes vs. IH+PA or RA FMT-treated mice (p = 0.001). Interpretation IH exposures mimicking OSA induce changes in GM, increase gut permeability, and alter plasma exosome cargo, the latter inducing adipocyte dysfunction (increased IR). Furthermore, these alterations improved with PA. Thus, IH leads to perturbations of a singular GM-circulating exosome pathway that disrupts adipocyte homeostasis resulting in metabolic dysfunction, as reflected by IR. Funding This study was supported by grants from the National Institutes of Health grants HL130984 and HL140548 and University of Missouri Tier 2 grant. The study has not received any funding or grants from pharmaceutical or other industrial corporations.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Department of Child Health and the Child Health Research Institute, University of Missouri, School of Medicine, Columbia, 400N. Keene Street, Suite 010, MO 65201, United States.
| | - Aaron Ericsson
- University of Missouri Metagenomics Center, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri at Columbia, Columbia, MO 65201, United States
| | - Zhuanghong Qiao
- Department of Child Health and the Child Health Research Institute, University of Missouri, School of Medicine, Columbia, 400N. Keene Street, Suite 010, MO 65201, United States
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER de Enfermedades Respiratorias, Madrid, Spain; Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER de Enfermedades Respiratorias, Madrid, Spain; Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, University of Missouri, School of Medicine, Columbia, 400N. Keene Street, Suite 010, MO 65201, United States.
| |
Collapse
|
664
|
Impact of Different Exercise Modalities on the Human Gut Microbiome. Sports (Basel) 2021; 9:sports9020014. [PMID: 33494210 PMCID: PMC7909775 DOI: 10.3390/sports9020014] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 12/18/2022] Open
Abstract
In this study we examined changes to the human gut microbiome resulting from an eight-week intervention of either cardiorespiratory exercise (CRE) or resistance training exercise (RTE). Twenty-eight subjects (21 F; aged 18-26) were recruited for our CRE study and 28 subjects (17 F; aged 18-33) were recruited for our RTE study. Fecal samples for gut microbiome profiling were collected twice weekly during the pre-intervention phase (three weeks), intervention phase (eight weeks), and post-intervention phase (three weeks). Pre/post VO2max, three repetition maximum (3RM), and body composition measurements were conducted. Heart rate ranges for CRE were determined by subjects' initial VO2max test. RTE weight ranges were established by subjects' initial 3RM testing for squat, bench press, and bent-over row. Gut microbiota were profiled using 16S rRNA gene sequencing. Microbiome sequence data were analyzed with QIIME 2. CRE resulted in initial changes to the gut microbiome which were not sustained through or after the intervention period, while RTE resulted in no detectable changes to the gut microbiota. For both CRE and RTE, we observe some evidence that the baseline microbiome composition may be predictive of exercise gains. This work suggests that the human gut microbiome can change in response to a new exercise program, but the type of exercise likely impacts whether a change occurs. The changes observed in our CRE intervention resemble a disturbance to the microbiome, where an initial shift is observed followed by a return to the baseline state. More work is needed to understand how sustained changes to the microbiome occur, resulting in differences that have been reported in cross sectional studies of athletes and non-athletes.
Collapse
|
665
|
Hu-Lieskovan S, Malouf GG, Jacobs I, Chou J, Liu L, Johnson ML. Addressing resistance to immune checkpoint inhibitor therapy: an urgent unmet need. Future Oncol 2021; 17:1401-1439. [PMID: 33475012 DOI: 10.2217/fon-2020-0967] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of various cancers by reversing the immunosuppressive mechanisms employed by tumors to restore anticancer immunity. Although ICIs have demonstrated substantial clinical efficacy, patient response can vary in depth and duration, and many do not respond at all or eventually develop resistance. ICI resistance mechanisms can be tumor-intrinsic, related to the tumor microenvironment or patient-specific factors. Multiple resistance mechanisms may be present within one tumor subtype, or heterogeneity exists among patients with the same tumor type. Consequently, designing effective combination treatment strategies is challenging. This review will discuss ICI resistance mechanisms, and summarize findings from key preclinical and clinical trials of ICIs, to identify potential treatment strategies or pathways to overcome ICI resistance.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Department of Medicine, Division of Oncology, Huntsman Cancer Institute / University of Utah, Salt Lake City, UT 84112, USA
| | - Gabriel G Malouf
- Department of Medical Oncology, Institut de Cancérologie de Strasbourg & Department of Functional Genomics & Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UNISTRA, Illkirch Cedex, Strasbourg, France
| | | | | | - Li Liu
- Pfizer Inc, San Diego, CA 92121, USA
| | - Melissa L Johnson
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN 37203, USA
| |
Collapse
|
666
|
|
667
|
The influence of gut microbiota in cardiovascular diseases-a brief review. Porto Biomed J 2021; 6:e106. [PMID: 33490701 PMCID: PMC7817281 DOI: 10.1097/j.pbj.0000000000000106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Lately, the gut microbiota has emerged as an important mediator of the development and the outcomes of certain diseases. It's well known that the gut microbiota plays an important role in maintaining human health. Still far from being completely understood and analyzed is the complexity of this ecosystem, although a close relationship between the gut microbiota and cardiovascular diseases (CVD) has been established. A loss of diversity in the microbiota will lead to physiological changes, which can improve inflammatory or infection states like atherosclerosis and hypertension, the basic pathological process of CVD. Targeting the gut microbiota and its metabolites are new and promising strategies for the treatment and prognosis of CVD.
Collapse
|
668
|
Wu Q, Liu J, Wu S, Xie X. The impact of antibiotics on efficacy of immune checkpoint inhibitors in malignancies: A study based on 44 cohorts. Int Immunopharmacol 2021; 92:107303. [PMID: 33465728 DOI: 10.1016/j.intimp.2020.107303] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Pre-clinical and clinical data had revealed the gut microbiome plays a critical role in immune checkpoint inhibitors (ICIs) efficacy. This study was designed to investigate whether antibiotics (ATBs) affect the prognosis of malignancies treated with ICIs. METHODS Electronic databases were searched to identify relevant trials that evaluated the impact of ATBs on ICIs efficacy. The primary endpoints were overall survival (OS) and progression-free survival (PFS) measured by HRs with corresponding 95%CIs. Subgroup analyses were performed based on cancer type, study design, ICIs agent, and time of ATBs administration. RESULTS Totally, 12,492 individuals in the 44 cohorts were recruited. Pooled results showed that ATBs administration was significantly correlated with a worse objective remission rate (ORR) (OR = 0.61, 95%CI (0.42-0.90), P = 0.0128), PFS (HR = 1.18, 95%CI (1.11-1.25), P < 0.0001), and OS (HR = 1.20, 95%CI (1.15-1.25), P < 0.0001) in patients treated with ICIs. In subgroup analyses, patients treated with ICIs exposed to ATBs suffered an evidently worse ORR in arms of renal cell carcinoma (RCC) (OR = 0.30, 95%CI (0.14-0.67), P = 0.0034), multiple (OR = 0.44, 95%CI (0.27-0.73), P = 0.0016), and before ICIs initiation (OR = 0.47, 95%CI (0.32-0.71), P = 0.0003) without heterogeneity; experienced a worse PFS and OS in arms of non-small cell lung cancer, melanoma, RCC, urothelial carcinoma, multiple, prospective, retrospective, PD-(L)1 alone, PD-(L)1 plus CTLA-4, before ICIs initiation, before ICIs initiation and concurrent, and before or after ICIs within 1 month, while a better PFS and OS in concurrent with ICIs arm. CONCLUSIONS ATBs administration was negatively associated with ORR, PFS and OS in malignancies treated with ICIs, while the time of ATBs exposure might impact ICIs efficacy.
Collapse
Affiliation(s)
- Qing Wu
- Department of Oncology, Molecular Oncology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Junjin Liu
- Department of Infectious Diseases, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Sumei Wu
- Department of Oncology, Molecular Oncology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Xianhe Xie
- Department of Oncology, Molecular Oncology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China.
| |
Collapse
|
669
|
Kaymak I, Williams KS, Cantor JR, Jones RG. Immunometabolic Interplay in the Tumor Microenvironment. Cancer Cell 2021; 39:28-37. [PMID: 33125860 PMCID: PMC7837268 DOI: 10.1016/j.ccell.2020.09.004] [Citation(s) in RCA: 204] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/22/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
Immune cells' metabolism influences their differentiation and function. Given that a complex interplay of environmental factors within the tumor microenvironment (TME) can have a profound impact on the metabolic activities of immune, stromal, and tumor cell types, there is emerging interest to advance understanding of these diverse metabolic phenotypes in the TME. Here, we discuss cell-extrinsic contributions to the metabolic activities of immune cells. Then, considering recent technical advances in experimental systems and metabolic profiling technologies, we propose future directions to better understand how immune cells meet their metabolic demands in the TME, which can be leveraged for therapeutic benefit.
Collapse
Affiliation(s)
- Irem Kaymak
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Kelsey S Williams
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Jason R Cantor
- Morgridge Institute for Research, Madison, WI 53715, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Russell G Jones
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
670
|
Parasympathetic influences in cancer pathogenesis: further insights. Clin Transl Oncol 2021; 23:1491-1493. [PMID: 33398710 DOI: 10.1007/s12094-020-02523-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 10/22/2022]
|
671
|
Choudhry H. The Microbiome and Its Implications in Cancer Immunotherapy. Molecules 2021; 26:E206. [PMID: 33401586 PMCID: PMC7795182 DOI: 10.3390/molecules26010206] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is responsible for ~18 million deaths globally each year, representing a major cause of death. Several types of therapy strategies such as radiotherapy, chemotherapy and more recently immunotherapy, have been implemented in treating various types of cancer. Microbes have recently been found to be both directly and indirectly involved in cancer progression and regulation, and studies have provided novel and clear insights into the microbiome-mediated emergence of cancers. Scientists around the globe are striving hard to identify and characterize these microbes and the underlying mechanisms by which they promote or suppress various kinds of cancer. Microbes may influence immunotherapy by blocking various cell cycle checkpoints and the production of certain metabolites. Hence, there is an urgent need to better understand the role of these microbes in the promotion and suppression of cancer. The identification of microbes may help in the development of future diagnostic tools to cure cancers possibly associated with the microbiome. This review mainly focuses on various microbes and their association with different types of cancer, responses to immunotherapeutic modulation, physiological responses, and prebiotic and postbiotic effects.
Collapse
Affiliation(s)
- Hani Choudhry
- Department of Biochemistry, Faculty of Sciences, Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
672
|
Li J, Liang L, Yang Y, Li X, Ma Y. N 6-methyladenosine as a biological and clinical determinant in colorectal cancer: progression and future direction. Theranostics 2021; 11:2581-2593. [PMID: 33456561 PMCID: PMC7806471 DOI: 10.7150/thno.52366] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers and one of the leading causes of cancer death. Recent studies have provided evidence that N6-methyladenosine (m6A), the most abundant RNA modifications in eukaryote, performs many functions in RNA metabolism including translation, splicing, storage, trafficking and degradation. Aberrant regulation of m6A modification in mRNAs and noncoding RNAs found in CRC tissues is crucial for cancer formation, progression, invasion and metastasis. Further, m6A regulators and m6A-related RNAs may become promising biomarkers, prognosis predictors as well as therapeutic targets. Here, we review the biological and clinical roles of m6A modification in CRC, and discuss the potential of m6A in clinical translation.
Collapse
|
673
|
Pradhan M, Chocry M, Gibbons DL, Sepesi B, Cascone T. Emerging biomarkers for neoadjuvant immune checkpoint inhibitors in operable non-small cell lung cancer. Transl Lung Cancer Res 2021; 10:590-606. [PMID: 33569339 PMCID: PMC7867746 DOI: 10.21037/tlcr-20-573] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The advent of immune checkpoint inhibitors (ICIs) has dramatically changed the treatment of patients with locally advanced unresectable and metastatic non-small cell lung cancer (NSCLC). Now, ICIs are undergoing evaluation as neoadjuvant therapy in patients with early-stage, resectable NSCLC using candidate surrogate endpoints of clinical efficacy, i.e., major pathologic response (MPR, ≤10% viable tumor cells in resected tumors). The initial results from early, small-scale trials are encouraging; however, they also reveal that a substantial number of patients with operable disease may not benefit from neoadjuvant ICIs. Consequently, much investigative effort is currently directed toward identifying mechanisms of resistance to ICI therapy in resectable NSCLC. There is also an urgent need for biomarkers that could be used to guide the clinical decision-making process and maximize the clinical benefit of ICIs in patients with early-stage, resectable NSCLC. Here, we summarize the initial results from the trials of neoadjuvant ICIs in patients with early-stage and locally advanced operable NSCLC and review the findings of studies investigating emerging biomarkers associated with those trials.
Collapse
Affiliation(s)
- Monika Pradhan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mathieu Chocry
- Aix-Marseille Université, Institut de Neurophysiopathologie (INP), CNRS, Marseille, France
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
674
|
Abstract
The functional diversity of the mammalian intestinal microbiome far exceeds that of the host organism, and microbial genes contribute substantially to the well-being of the host. However, beneficial gut organisms can also be pathogenic when present in the gut or other locations in the body. Among dominant beneficial bacteria are several species of Bacteroides, which metabolize polysaccharides and oligosaccharides, providing nutrition and vitamins to the host and other intestinal microbial residents. These topics and the specific organismal and molecular interactions that are known to be responsible for the beneficial and detrimental effects of Bacteroides species in humans comprise the focus of this review. The complexity of these interactions will be revealed.
Collapse
Affiliation(s)
- Hassan Zafar
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, USA
- Department of Microbiology and Molecular Genetics, Faculty of Life Sciences, University of Okara,Okara, PunjabPakistan
| | - Milton H. Saier
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, USA
| |
Collapse
|
675
|
Riaz Rajoka MS, Mehwish HM, Xiong Y, Song X, Hussain N, Zhu Q, He Z. Gut microbiota targeted nanomedicine for cancer therapy: Challenges and future considerations. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2020.10.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
676
|
Fink A, Doblhammer G, Tamgüney G. Recurring Gastrointestinal Infections Increase the Risk of Dementia. J Alzheimers Dis 2021; 84:797-806. [PMID: 34602468 PMCID: PMC8673498 DOI: 10.3233/jad-210316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Gastrointestinal infections cause significant health problems, including those affecting the immune, musculoskeletal, and nervous system, and are one of the leading causes for death worldwide. Recent findings suggest that microbiota of the gastrointestinal tract contribute to dementia. OBJECTIVE In this nested case-control study we investigated the role of common gastrointestinal infections on the subsequent risk of dementia. METHODS We used a longitudinal sample of 202,806 individuals from health claims data of the largest German health insurer and applied a nested case-control design with 23,354 initial dementia cases between 2006 and 2014 and 23,354 matched controls. We used conditional logistic regression to compute odds ratios (ORs) for dementia and corresponding 95%confidence intervals (CIs), adjusting for potential confounders. RESULTS The risk of dementia was increased in patients with recurring incidences of quarters with diagnosed gastrointestinal infections when compared to the unexposed population (one quarter: OR = 1.49, 95%CI = 1.40-1.58; two quarters: OR = 1.70, 95%CI = 1.51-1.91; three or more quarters: OR = 1.64, 95%CI = 1.40-1.93), adjusted for potential confounders. CONCLUSION Our findings suggest that recurring gastrointestinal infections are associated with an increased risk of subsequent dementia.
Collapse
Affiliation(s)
- Anne Fink
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gabriele Doblhammer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Sociology and Demography, University of Rostock, Rostock, Germany
| | - Gültekin Tamgüney
- Institut für Biologische Informationsprozesse, Strukturbiochemie (IBI-7), Forschungszentrum Jülich GmbH, Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
677
|
Atezolizumab (in Combination with Nab-Paclitaxel): A Review in Advanced Triple-Negative Breast Cancer. Drugs 2020; 80:601-607. [PMID: 32248356 DOI: 10.1007/s40265-020-01295-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Atezolizumab (Tecentriq®), an immune checkpoint inhibitor against programmed death ligand 1 (PD-L1), is the first immunotherapy agent to be approved (for use in combination with nab-paclitaxel) in the USA, the EU (as first-line) and Japan for the treatment of advanced triple-negative breast cancer (TNBC). Approval was based on the results of the phase III IMpassion130 trial in patients with unresectable locally advanced or metastatic TNBC, in which atezolizumab plus nab-paclitaxel significantly prolonged progression-free survival (PFS) when compared to placebo plus nab-paclitaxel in the intent-to-treat (ITT) population and the PD-L1+ subgroup. Statistically significant overall survival (OS) benefits were not seen in two interim analyses and final OS data are awaited. The tolerability and safety profile of atezolizumab plus nab-paclitaxel was consistent with those of each individual drug. The most common treatment-related adverse events included neutropenia, peripheral neuropathy and reduced neutrophil count. Adverse events of special interest occurred with higher frequency in patients who received atezolizumab plus nab-paclitaxel than placebo plus nab-paclitaxel, and were mostly immune-related (e.g. immune-related rash, hypothyroidism and hepatitis). Health-related quality of life was not significantly impacted by the addition of atezolizumab to nab-paclitaxel therapy. Thus, atezolizumab plus nab-paclitaxel is a useful immunochemotherapy option for patients with unresectable locally advanced or metastatic TNBC, including those whose tumours have PD-L1 expression ≥ 1%.
Collapse
|
678
|
Miglietta F, Cona MS, Dieci MV, Guarneri V, La Verde N. An overview of immune checkpoint inhibitors in breast cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:452-472. [PMID: 36046385 PMCID: PMC9400749 DOI: 10.37349/etat.2020.00029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Although breast cancer is not traditionally considered an immunogenic type of tumor, the combination of immunotherapy and chemotherapy has recently emerged as a novel treatment option in triple-negative subtype in the advanced setting and other similar combinations of immune checkpoint inhibitors with chemotherapy are expected to become part of the neoadjuvant management in the near future. In addition, encouraging results have been observed with the combination of immune checkpoint blockade with diverse biological agents, including anti-HER2 agents, CDK 4/6 inhibitors, PARP-inhibitors. The present review summarized the available evidence coming from clinical trials on the role of immune checkpoint inhibitors in the management of breast cancer, both in advanced and early setting.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Maria Silvia Cona
- Department of Oncology, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, Via G.B. Grassi 74, 20157 Milano, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Nicla La Verde
- Department of Oncology, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, Via G.B. Grassi 74, 20157 Milano, Italy
| |
Collapse
|
679
|
Qi X, Yang M, Stenberg J, Dey R, Fogwe L, Alam MS, Kimchi ET, Staveley-O'Carroll KF, Li G. Gut microbiota mediated molecular events and therapy in liver diseases. World J Gastroenterol 2020; 26:7603-7618. [PMID: 33505139 PMCID: PMC7789060 DOI: 10.3748/wjg.v26.i48.7603] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/24/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota is a community of microorganisms that reside in the gastrointestinal tract. An increasing number of studies has demonstrated that the gut-liver axis plays a critical role in liver homeostasis. Dysbiosis of gut microbiota can cause liver diseases, including nonalcoholic fatty liver disease and alcoholic liver disease. Preclinical and clinical investigations have substantiated that the metabolites and other molecules derived from gut microbiota and diet interaction function as mediators to cause liver fibrosis, cirrhosis, and final cancer. This effect has been demonstrated to be associated with dysregulation of intrahepatic immunity and liver metabolism. Targeting these findings have led to the development of novel preventive and therapeutic strategies. Here, we review the cellular and molecular mechanisms underlying gut microbiota-mediated impact on liver disease. We also summarize the advancement of gut microbiota-based therapeutic strategies in the control of liver diseases.
Collapse
Affiliation(s)
- Xiaoqiang Qi
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
| | - Joseph Stenberg
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
| | - Rahul Dey
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
| | - Leslie Fogwe
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
| | | | - Eric T Kimchi
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
| | - Kevin F Staveley-O'Carroll
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
| | - Guangfu Li
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65212, United States
| |
Collapse
|
680
|
Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, Rotin D, Anafi L, Avivi C, Melnichenko J, Steinberg-Silman Y, Mamtani R, Harati H, Asher N, Shapira-Frommer R, Brosh-Nissimov T, Eshet Y, Ben-Simon S, Ziv O, Khan MAW, Amit M, Ajami NJ, Barshack I, Schachter J, Wargo JA, Koren O, Markel G, Boursi B. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2020; 371:602-609. [PMID: 33303685 DOI: 10.1126/science.abb5920] [Citation(s) in RCA: 919] [Impact Index Per Article: 183.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022]
Abstract
The gut microbiome has been shown to influence the response of tumors to anti-PD-1 (programmed cell death-1) immunotherapy in preclinical mouse models and observational patient cohorts. However, modulation of gut microbiota in cancer patients has not been investigated in clinical trials. In this study, we performed a phase 1 clinical trial to assess the safety and feasibility of fecal microbiota transplantation (FMT) and reinduction of anti-PD-1 immunotherapy in 10 patients with anti-PD-1-refractory metastatic melanoma. We observed clinical responses in three patients, including two partial responses and one complete response. Notably, treatment with FMT was associated with favorable changes in immune cell infiltrates and gene expression profiles in both the gut lamina propria and the tumor microenvironment. These early findings have implications for modulating the gut microbiota in cancer treatment.
Collapse
Affiliation(s)
- Erez N Baruch
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel. .,Department of Clinical Immunology and Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Youngster
- Pediatric Division and the Microbiome Research Center, Shamir (Assaf Harofeh) Medical Center, Be'er Ya'akov, Israel.,School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Ben-Betzalel
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Rona Ortenberg
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Adi Lahat
- Department of Gastroenterology, Sheba Medical Center, Tel HaShomer, Israel
| | - Lior Katz
- Department of Gastroenterology, Hadassah Medical Center, Jerusalem, Israel
| | - Katerina Adler
- Department of Mathematics, Bar Ilan University, Ramat Gan, Israel
| | | | - Stephen Raskin
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Radiological Institute, Sheba Medical Center, Tel HaShomer, Israel
| | - Naamah Bloch
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Daniil Rotin
- Institute of Pathology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Liat Anafi
- Institute of Pathology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Camila Avivi
- Institute of Pathology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Jenny Melnichenko
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Yael Steinberg-Silman
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Ronac Mamtani
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Hagit Harati
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Nethanel Asher
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Ronnie Shapira-Frommer
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Tal Brosh-Nissimov
- Infectious Diseases Unit, Assuta Ashdod University Hospital, Ashdod, Israel
| | - Yael Eshet
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Pathology, Sheba Medical Center, Tel-HaShomer, Israel.,Department of Nuclear Medicine, Sheba Medical Center, Tel HaShomer, Israel
| | - Shira Ben-Simon
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Oren Ziv
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Md Abdul Wadud Khan
- Program for Innovative Microbiome and Translational Research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nadim J Ajami
- Program for Innovative Microbiome and Translational Research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Iris Barshack
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Pathology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Jacob Schachter
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel.,School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jennifer A Wargo
- Program for Innovative Microbiome and Translational Research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Gal Markel
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel. .,Department of Clinical Immunology and Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Talpiot Medical Leadership Program, Sheba Medical Center, Tel HaShomer, Israel
| | - Ben Boursi
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, USA.,Department of Oncology, Sheba Medical Center, Tel HaShomer, Israel
| |
Collapse
|
681
|
Liu NN, Ma Q, Ge Y, Yi CX, Wei LQ, Tan JC, Chu Q, Li JQ, Zhang P, Wang H. Microbiome dysbiosis in lung cancer: from composition to therapy. NPJ Precis Oncol 2020; 4:33. [PMID: 33303906 PMCID: PMC7730185 DOI: 10.1038/s41698-020-00138-z] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
The correlations between microbiota dysbiosis and cancer have gained extensive attention and been widely explored. As a leading cancer diagnosis worldwide, lung cancer poses a great threat to human health. The healthy human lungs are consistently exposed to external environment and harbor a specific pattern of microbiota, sharing many key pathological and physiological characteristics with the intestinal tract. Although previous findings uncovered the critical roles of microbiota in tumorigenesis and response to anticancer therapy, most of them were focused on the intestinal microbiota rather than lung microbiota. Notably, the considerable functions of microbiota in maintaining lung homeostasis should not be neglected as the microbiome dysbiosis may promote tumor development and progression through production of cytokines and toxins and multiple other pathways. Despite the fact that increasing studies have revealed the effect of microbiome on the induction of lung cancer and different disease status, the underlying mechanisms and potential therapeutic strategies remained unclear. Herein, we summarized the recent progresses about microbiome in lung cancer and further discussed the role of microbial communities in promoting lung cancer progression and the current status of therapeutic approaches targeting microbiome to alleviate and even cure lung cancer.
Collapse
Affiliation(s)
- Ning-Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Qiang Ma
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital Tongji University, Shanghai, China
| | - Yang Ge
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Cheng-Xiang Yi
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital Tongji University, Shanghai, China
| | - Lu-Qi Wei
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jing-Cong Tan
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Qiao Chu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jing-Quan Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital Tongji University, Shanghai, China.
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
682
|
Zheng L, Xu J, Sai B, Zhu Y, Wang L, Yin N, Yu F, Zhou W, Wu M, Tang J, Xiang J. Microbiome Related Cytotoxically Active CD8+ TIL Are Inversely Associated With Lung Cancer Development. Front Oncol 2020; 10:531131. [PMID: 33363002 PMCID: PMC7756114 DOI: 10.3389/fonc.2020.531131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the most common cancer type around the world. Although major advances in cancer therapy, lung cancer has been the largest proportion of all cancer-related deaths. The respiratory tract contains many types of bacteria and a distinct lung microbiome in lung cancer patients was described in many studies. The specific roles of these lung microorganisms in lung cancer progression remain unclear. In this study, we evaluated the effect of inhalation of bronchoalveolar fluid (BAL) in the lung cancer cell growth. The microbiome-based immune and carcinogenesis was examined in tumor-bearing mouse model. We found that inhalation of BAL collected from non-small cell lung cancer (NSCLC) patients altered the lung microbiota and inhibited tumor cell growth. The inhibitory effect was due to the infiltration of CD3 and CD8+ T cells and decrease of M2 macrophages in lungs. The microbial communities of NSCLC BAL inhalation group were dominated by Pasteurella, whereas the microbial communities of non-cancer control and PBS inhalation group were dominated by Delftia. Linear discriminant analysis (LDA) indicated that the genera Pasteurella, Pseudomonas, and Chryseobacterium were increased in NSCLC BAL inhalation group, while genera Delftia, Ezakiella, Blautia, Cloacibacterium, and Microvirga et al. were increased in PBS and Non-cancer group. We demonstrated a significant positive correlation between Pasteurella and cytotoxic CD8+ TIL and a negative correlation with M2 macrophages. Coriobacteriaceae was positively correlated with M2 macrophages and negatively correlated with CD8+ cells. The abundance of Pasteurella was negatively correlated with tumor cell growth. Our findings provide a promising strategy that can be used as a therapeutic vaccine for lung cancer patients.
Collapse
Affiliation(s)
- Leliang Zheng
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Jiaqi Xu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, China
| | - Buqing Sai
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, China
| | - Yinghong Zhu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, China
| | - Lujuan Wang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, China
| | - Na Yin
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, China
| | - Fenglei Yu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, Changsha, China
| | - Wen Zhou
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, China
| | - Minghua Wu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, China
| | - Jingqun Tang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, Changsha, China
| | - Juanjuan Xiang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, China
| |
Collapse
|
683
|
Gaissmaier L, Christopoulos P. Immune Modulation in Lung Cancer: Current Concepts and Future Strategies. Respiration 2020; 99:1-27. [PMID: 33291116 DOI: 10.1159/000510385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer immunotherapy represents the most dynamic field of biomedical research currently, with thoracic immuno-oncology as a forerunner. PD-(L)1 inhibitors are already part of standard first-line treatment for both non-small-cell and small-cell lung cancer, while unprecedented 5-year survival rates of 15-25% have been achieved in pretreated patients with metastatic disease. Evolving strategies are mainly aiming for improvement of T-cell function, increase of immune activation in the tumor microenvironment (TME), and supply of tumor-reactive lymphocytes. Several novel therapeutics have demonstrated preclinical efficacy and are increasingly used in rational combinations within clinical trials. Two overarching trends dominate: extension of immunotherapy to earlier disease stages, mainly as neoadjuvant treatment, and a shift of focus towards multivalent, individualized, mutatome-based antigen-specific modalities, mainly adoptive cell therapies and cancer vaccines. The former ensures ample availability of treated and untreated patient samples, the latter facilitates deeper mechanistic insights, and both in combination build an overwhelming force that is accelerating progress and driving the greatest revolution cancer medicine has seen so far. Today, immune modulation represents the most potent therapeutic modality in oncology, the most important topic in clinical and translational cancer research, and arguably our greatest, meanwhile justified hope for achieving cure of pulmonary neoplasms and other malignancies in the next future.
Collapse
Affiliation(s)
- Lena Gaissmaier
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany,
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany,
| |
Collapse
|
684
|
Wang R, Chen M, Ye X, Poon K. Role and potential clinical utility of ARID1A in gastrointestinal malignancy. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 787:108360. [PMID: 34083049 DOI: 10.1016/j.mrrev.2020.108360] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/26/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
ARID1A (AT-rich interactive domain 1A) is a newly discovered tumor suppressor gene, and its encoded product is an important component of the SWI/SNF chromatin remodeling complex. ARID1A plays an important role in cell proliferation, invasion and metastasis, apoptosis, cell cycle regulation, epithelial mesenchymal transition, and the regulation of other of biological behaviors. Recently, ARID1A mutations have been increasingly reported in esophageal adenocarcinoma, gastric cancer, colorectal cancer, hepatocellular carcinoma, cholangiocarcinoma, pancreatic cancer, and other malignant tumors of the digestive system. This article reviews the relationship between ARID1A mutation and the molecular mechanisms of carcinogenesis, including microsatellite instability and the PI3K/ATK signaling pathway, and relates these mechanisms to the prognostic assessment of digestive malignancy. Further, this review describes the potential for molecular pathologic epidemiology (MPE) to provide new insights into environment-tumor-host interactions.
Collapse
Affiliation(s)
- Ruihua Wang
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, Guangdong Province, China.
| | - Mei Chen
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, Guangdong Province, China.
| | - Xiaojun Ye
- Program of Food Science and Technology, Division of Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, 519085, Guangdong Province, China.
| | - Karen Poon
- Program of Food Science and Technology, Division of Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, 519085, Guangdong Province, China.
| |
Collapse
|
685
|
Limeta A, Ji B, Levin M, Gatto F, Nielsen J. Meta-analysis of the gut microbiota in predicting response to cancer immunotherapy in metastatic melanoma. JCI Insight 2020; 5:140940. [PMID: 33268597 PMCID: PMC7714408 DOI: 10.1172/jci.insight.140940] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/16/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Identifying factors conferring responses to therapy in cancer is critical to select the best treatment for patients. For immune checkpoint inhibition (ICI) therapy, mounting evidence suggests that the gut microbiome can determine patient treatment outcomes. However, the extent to which gut microbial features are applicable across different patient cohorts has not been extensively explored. METHODS We performed a meta-analysis of 4 published shotgun metagenomic studies (Ntot = 130 patients) investigating differential microbiome composition and imputed metabolic function between responders and nonresponders to ICI. RESULTS Our analysis identified both known microbial features enriched in responders, such as Faecalibacterium as the prevailing taxa, as well as additional features, including overrepresentation of Barnesiella intestinihominis and the components of vitamin B metabolism. A classifier designed to predict responders based on these features identified responders in an independent cohort of 27 patients with the area under the receiver operating characteristic curve of 0.625 (95% CI: 0.348–0.899) and was predictive of prognosis (HR = 0.35, P = 0.081). CONCLUSION These results suggest the existence of a fecal microbiome signature inherent across responders that may be exploited for diagnostic or therapeutic purposes. FUNDING This work was funded by the Knut and Alice Wallenberg Foundation, BioGaia AB, and Cancerfonden. A meta-analysis of studies investigating the impact of the gut microbiome on response to cancer immunotherapy in patients with metastatic melanoma
Collapse
Affiliation(s)
- Angelo Limeta
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Boyang Ji
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Max Levin
- Wallenberg Laboratory for Cardiovascular Research, Department of Molecular and Clinical Medicine, and.,Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Francesco Gatto
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.,Elypta AB, Stockholm, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark.,BioInnovation Institute, Copenhagen, Denmark
| |
Collapse
|
686
|
Masetti R, Zama D, Leardini D, Muratore E, Turroni S, Prete A, Brigidi P, Pession A. The gut microbiome in pediatric patients undergoing allogeneic hematopoietic stem cell transplantation. Pediatr Blood Cancer 2020; 67:e28711. [PMID: 32939928 DOI: 10.1002/pbc.28711] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022]
Abstract
The gut microbiome (GM) has been associated with different clinical outcomes in the context of allogeneic hematopoietic stem cell transplantation (HSCT). Large multicenter cohort studies in adults have found significant correlations with overall survival, relapse, and incidence of complications. Moreover, GM is already a promising target for therapeutic interventions. However, few data are available in children, a population presenting unique features and challenges. During childhood, the GM evolves rapidly with large structural fluctuations, alongside with the maturation of the immune system. Furthermore, the HSCT procedure presents significant differences in children. These considerations underline the importance of a specific focus on the pediatric setting, and the role of GM and its age-dependent trajectory in influencing the immunity reconstitution and clinical outcomes. This review provides a comprehensive overview of the available evidence in the field of GM and pediatric HSCT, highlighting age-specific issues and discussing GM-based therapeutic approaches.
Collapse
Affiliation(s)
- Riccardo Masetti
- Pediatric Oncology and Hematology Unit "Lalla Seràgnoli," Department of Pediatrics, University of Bologna, Sant'Orsola Malpighi Hospital, Bologna, Italy
| | - Daniele Zama
- Pediatric Oncology and Hematology Unit "Lalla Seràgnoli," Department of Pediatrics, University of Bologna, Sant'Orsola Malpighi Hospital, Bologna, Italy
| | - Davide Leardini
- Pediatric Oncology and Hematology Unit "Lalla Seràgnoli," Department of Pediatrics, University of Bologna, Sant'Orsola Malpighi Hospital, Bologna, Italy
| | - Edoardo Muratore
- Pediatric Oncology and Hematology Unit "Lalla Seràgnoli," Department of Pediatrics, University of Bologna, Sant'Orsola Malpighi Hospital, Bologna, Italy
| | - Silvia Turroni
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Arcangelo Prete
- Pediatric Oncology and Hematology Unit "Lalla Seràgnoli," Department of Pediatrics, University of Bologna, Sant'Orsola Malpighi Hospital, Bologna, Italy
| | - Patrizia Brigidi
- Department of Medicine and Surgery (DIMEC), University of Bologna, Bologna, Italy
| | - Andrea Pession
- Pediatric Oncology and Hematology Unit "Lalla Seràgnoli," Department of Pediatrics, University of Bologna, Sant'Orsola Malpighi Hospital, Bologna, Italy
| |
Collapse
|
687
|
Bandera-Merchan B, Boughanem H, Crujeiras AB, Macias-Gonzalez M, Tinahones FJ. Ketotherapy as an epigenetic modifier in cancer. Rev Endocr Metab Disord 2020; 21:509-519. [PMID: 32514818 DOI: 10.1007/s11154-020-09567-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epigenetic alterations in cancer play a variety of roles. Aberrant DNA methylation, as one of the epigenetic mechanisms, has been widely studied in both tumor and liquid biopsies and provide a useful bench mark for treatment response in cancer. Recently, several studies have reported an association between the type of diet and epigenetic modifications. Whereby there is a growing interest in finding the "anti-cancer diet formula", if such a thing exists. In this sense, ketogenic diets (KD) have reported potentially beneficial effects, which were able to prevent malignancies and decrease tumor growth. Some studies have even shown increased survival in cancer patients, reduced side effects of cytotoxic treatments, and intensified efficacy of cancer therapies. Although the biological mechanisms of KD are not well understood, it has been reported that KD may affect DNA methylation by modulating the expression of crucial genes involved in tumor survival and proliferation. However, there are many considerations to take into account to use ketotherapy in cancer, such as epigenetic mark, type of cancer, immunological and metabolic state or microbiota profile. In this review, we argue about ketotherapy as a potential strategy to consider as coadjuvant of cancer therapy. We will focus on mainly epigenetic mechanisms and dietary approach that could be included in the current clinical practice guidelines.
Collapse
Affiliation(s)
- Borja Bandera-Merchan
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010, Málaga, Spain
| | - Hatim Boughanem
- Biomedical Research Institute of Malaga (IBIMA). Faculty of Science, University of Malaga, 29010, Málaga, Spain
| | - Ana B Crujeiras
- Epigenomics in Endocrinology and Nutrition Group, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS/SERGAS), 15706, Santiago de Compostela, Spain
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Manuel Macias-Gonzalez
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010, Málaga, Spain.
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Francisco J Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010, Málaga, Spain
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
688
|
Newsome RC, Jobin C. Microbiome-Derived Liquid Biopsy: New Hope for Cancer Screening? Clin Chem 2020; 67:463-465. [PMID: 33277898 DOI: 10.1093/clinchem/hvaa240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 01/20/2023]
Affiliation(s)
| | - Christian Jobin
- Department of Medicine.,Department of Infectious Diseases and Pathology.,Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
689
|
Șurlin P, Nicolae FM, Șurlin VM, Pătrașcu Ș, Ungureanu BS, Didilescu AC, Gheonea DI. Could Periodontal Disease through Periopathogen Fusobacterium Nucleatum be an Aggravating Factor for Gastric Cancer? J Clin Med 2020; 9:jcm9123885. [PMID: 33260439 PMCID: PMC7761398 DOI: 10.3390/jcm9123885] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Periodontal disease affects the supporting tissues of the teeth, being a chronic inflammatory disease caused by specific microorganisms from subgingival biofilm. Fusobacterium nucleatum is a Gram-negative anaerobic bacterium that acts as a periodontal pathogen, being an important factor in linking Gram-positive and Gram-negative bacteria in the periodontal biofilm, but its involvement in systemic diseases has also been found. Several studies regarding the implication of Fusobacterium nucleatum in gastro-enterological cancers have been conducted. The present review aims to update and systematize the latest information about Fusobacterium nucleatum in order to evaluate the possibility of an association between periodontal disease and the evolution of gastroenterological cancers through the action of Fusobacterium nucleatum, highlighting gastric cancer. This would motivate future research on the negative influence of periodontal pathology on the evolution of gastric cancer in patients suffering from both pathologies.
Collapse
Affiliation(s)
- Petra Șurlin
- Department of Periodontology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Flavia Mirela Nicolae
- Department of Periodontology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Correspondence: (F.M.N.); (V.M.S.)
| | - Valeriu Marin Șurlin
- Department 1st of Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Correspondence: (F.M.N.); (V.M.S.)
| | - Ștefan Pătrașcu
- Department 1st of Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Bogdan Silviu Ungureanu
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (B.S.U.); (D.I.G.)
| | - Andreea Cristiana Didilescu
- Department of Embriology, University of Medicine and Pharmacy Carol Davila of Bucharest, 020021 Bucharest, Romania;
| | - Dan Ionuț Gheonea
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (B.S.U.); (D.I.G.)
| |
Collapse
|
690
|
Buti S, Bersanelli M, Perrone F, Tiseo M, Tucci M, Adamo V, Stucci LS, Russo A, Tanda ET, Spagnolo F, Rastelli F, Pergolesi F, Santini D, Russano M, Anesi C, Giusti R, Filetti M, Marchetti P, Botticelli A, Gelibter A, Occhipinti MA, Ferrari M, Vitale MG, Nicolardi L, Chiari R, Rijavec E, Nigro O, Tuzi A, De Tursi M, Di Marino P, Conforti F, Queirolo P, Bracarda S, Macrini S, Gori S, Zoratto F, Veltri E, Di Cocco B, Mallardo D, Vitale MG, Santoni M, Patruno L, Porzio G, Ficorella C, Pinato DJ, Ascierto PA, Cortellini A. Effect of concomitant medications with immune-modulatory properties on the outcomes of patients with advanced cancer treated with immune checkpoint inhibitors: development and validation of a novel prognostic index. Eur J Cancer 2020; 142:18-28. [PMID: 33212418 DOI: 10.1016/j.ejca.2020.09.033] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Concomitant medications are known to impact on clinical outcomes of patients treated with immune checkpoint inhibitors (ICIs). We aimed weighing the role of different concomitant baseline medications to create a drug-based prognostic score. METHODS We evaluated concomitant baseline medications at immunotherapy initiation for their impact on objective response rate (ORR), progression-free survival (PFS) and overall survival (OS) in a single-institution cohort of patients with advanced cancer treated with ICIs (training cohort, N = 217), and a drug-based prognostic score with the drugs resulting significantly impacting the OS was computed. Secondly, we externally validated the score in a large multicenter external cohort (n = 1012). RESULTS In the training cohort (n = 217), the median age was 69 years (range: 32-89), and the primary tumours were non-small-cell lung cancer (70%), melanoma (14.7%), renal cell carcinoma (9.2%) and others (6%). Among baseline medications, corticosteroids (hazard ratio [HR] = 2.3; 95% confidence interval [CI]: 1.60-3.30), systemic antibiotics (HR = 2.07; 95% CI: 1.31-3.25) and proton-pump inhibitors (PPIs) (HR = 1.57; 95% CI: 1.13-2.18) were significantly associated with OS. The prognostic score was calculated using these three drug classes, defining good, intermediate and poor prognosis patients. Within the training cohort, OS (p < 0.0001), PFS (p < 0.0001) and ORR (p = 0.0297) were significantly distinguished by the score stratification. The prognostic value of the score was also demonstrated in terms of OS (p < 0.0001), PFS (p < 0.0001) and ORR (p = 0.0006) within the external cohort. CONCLUSION Cumulative exposure to corticosteroids, antibiotics and PPIs (three likely microbiota-modulating drugs) leads to progressively worse outcomes after ICI therapy. We propose a simple score that can help stratifying patients in routine practice and clinical trials of ICIs.
Collapse
Affiliation(s)
- Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Melissa Bersanelli
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy; Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Fabiana Perrone
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy; Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marco Tucci
- Medical Oncology Unit, University of Bari, Department of Biomedical Sciences and Human Oncology, Bari, Italy; National Cancer Research Center, Tumori Institute IRCCS Giovanni Paolo II, Bari, Italy
| | - Vincenzo Adamo
- Medical Oncology, A.O. Papardo & Department of Human Pathology, University of Messina, Italy
| | - Luigia S Stucci
- Medical Oncology Unit, University of Bari, Department of Biomedical Sciences and Human Oncology, Bari, Italy
| | - Alessandro Russo
- Medical Oncology, A.O. Papardo & Department of Human Pathology, University of Messina, Italy
| | | | | | | | | | | | - Marco Russano
- Medical Oncology, Campus Bio-Medico University, Rome, Italy
| | - Cecilia Anesi
- Medical Oncology, Campus Bio-Medico University, Rome, Italy
| | - Raffaele Giusti
- Medical Oncology Unit, Sant' Andrea Hospital of Rome, Rome, Italy
| | - Marco Filetti
- Medical Oncology Unit, Sant' Andrea Hospital of Rome, Rome, Italy
| | - Paolo Marchetti
- Medical Oncology Unit, Sant' Andrea Hospital of Rome, Rome, Italy; Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy; Medical Oncology (B), Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy; Medical Oncology (B), Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Alain Gelibter
- Medical Oncology (B), Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | | | - Marco Ferrari
- Medical Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | | | | - Rita Chiari
- UOC Oncologia Padova Sud AULSS6 Euganea, Padova, Italy
| | - Erika Rijavec
- Medical Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Olga Nigro
- Medical Oncology, ASST Sette Laghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Alessandro Tuzi
- Medical Oncology, ASST Sette Laghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Michele De Tursi
- Department of Medical, Oral & Biotechnological Sciences, University G. D'Annunzio, Chieti-Pescara, Italy
| | | | - Fabio Conforti
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Paola Queirolo
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sergio Bracarda
- S.C. Medical Oncology, Azienda Ospedaliera S. Maria, Terni, Italy
| | - Serena Macrini
- S.C. Medical Oncology, Azienda Ospedaliera S. Maria, Terni, Italy
| | - Stefania Gori
- Oncology Unit, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar, VR, Italy
| | | | - Enzo Veltri
- Medical Oncology, Santa Maria Goretti Hospital, Latina, Italy
| | | | - Domenico Mallardo
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione "G. Pascale", Naples, Italy
| | - Maria Grazia Vitale
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione "G. Pascale", Naples, Italy
| | - Matteo Santoni
- Department of Oncology, Macerata Hospital, Macerata, Italy
| | - Leonardo Patruno
- Medical Oncology Unit, St. Salvatore Hospital, L'Aquila, Italy; Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Corrado Ficorella
- Medical Oncology Unit, St. Salvatore Hospital, L'Aquila, Italy; Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - David J Pinato
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione "G. Pascale", Naples, Italy
| | - Alessio Cortellini
- Medical Oncology Unit, St. Salvatore Hospital, L'Aquila, Italy; Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
691
|
Araujo DV, Watson GA, Oliva M, Heirali A, Coburn B, Spreafico A, Siu LL. Bugs as drugs: The role of microbiome in cancer focusing on immunotherapeutics. Cancer Treat Rev 2020; 92:102125. [PMID: 33227623 DOI: 10.1016/j.ctrv.2020.102125] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022]
Abstract
The human microbiome comprising microorganisms, their collective genomes and metabolic products has gained tremendous research interest in oncology, as multiple cohorts and case studies have demonstrated discernible interpatient differences in this ecosystem based on clinical variables including disease type, stage, diet, antibiotic usage, cancer treatments, therapeutic responses and toxicities. The modulation of the gut microbiome is the subject of many ongoing preclinical and clinical investigations, through the manipulation of diet, as well as the use of prebiotics, probiotics, specific antibiotics, fecal microbial transplantation, microbial consortia and stool substitutes. Standardization and quality control are needed to maximize the information being generated in this growing field, ranging from technical assays to measure microbiome composition, to methodological aspects in the analysis and reporting of results. Proof-of-mechanism and proof-of-concept clinical trials with appropriate controls are needed to confirm or refute the feasibility, safety and ultimately the clinical utility of human microbiome modulation in cancer patients.
Collapse
Affiliation(s)
- Daniel V Araujo
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, Canada
| | - Geoffrey A Watson
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, Canada
| | - Marc Oliva
- Catalan Institute of Oncology (Hospital Duran i Reynals), Department of Medical Oncology, IDIBELL, Barcelona, Spain
| | - Alya Heirali
- Toronto General Research Institute, Division of Infectious Diseases, University Health Network, Toronto, Canada
| | - Bryan Coburn
- Toronto General Research Institute, Division of Infectious Diseases, University Health Network, Toronto, Canada
| | - Anna Spreafico
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, Canada
| | - Lillian L Siu
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, Canada.
| |
Collapse
|
692
|
Forster S, Radpour R. Molecular Immunotherapy: Promising Approach to Treat Metastatic Colorectal Cancer by Targeting Resistant Cancer Cells or Cancer Stem Cells. Front Oncol 2020; 10:569017. [PMID: 33240813 PMCID: PMC7680905 DOI: 10.3389/fonc.2020.569017] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/08/2020] [Indexed: 12/17/2022] Open
Abstract
The immune system is able to recognize and eliminate tumor cells. Some tumors, including colorectal cancer (CRC), induce immune tolerance via different mechanisms of “immunoediting” and “immune evasion” and can thus escape immune surveillance. The impact of immunotherapy on cancer has been investigated for many years, but so far, the application was limited to few cancer types. Immuno-oncological therapeutic strategies against metastatic colorectal cancer (mCRC), the adaptive immune system activating approaches, offer a high potential for adaptation to the great heterogeneity of CRC. Moreover, novel treatment approaches are currently being tested that might specifically target the disease initiating and maintaining population of colorectal cancer stem cells (CSCs). In this review, we aim to summarize the current state of immune-oncology and tumor immunotherapy of patients with mCRC and discuss different therapeutic modalities that focus on the activation of tumor-specific T-cells and their perspectives such as tumor vaccination, checkpoint inhibition, and adoptive T-cell transfer or on the eradication of colorectal CSCs.
Collapse
Affiliation(s)
- Stefan Forster
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
693
|
Sui X, Chen Y, Liu B, Li L, Huang X, Wang M, Wang G, Gao X, Zhang L, Bao X, Yang D, Wang X, Zhong C. The relationship between KRAS gene mutation and intestinal flora in tumor tissues of colorectal cancer patients. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1085. [PMID: 33145304 PMCID: PMC7575961 DOI: 10.21037/atm-20-5622] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Colorectal cancer is among the most prominent malignant tumors endangering human health, with affected populations exhibiting an increasingly younger trend. The Kirsten ras (KRAS) gene acts as a crucial regulator in this disease and influences multiple signaling pathways. In the present study, the KRAS gene mutation-induced alteration of intestinal flora in colorectal cancer patients was explored, and the intestinal microbes that may be affected by the KRAS gene were examined to provide new insights into the diagnosis and treatment of colorectal cancer. Methods Deoxyribonucleic acid (DNA) was extracted from 177 colorectal cancer patients in our hospital. The mutation of the KRAS gene was subsequently detected using real-time fluorescence quantitative polymerase chain reaction (qPCR), and survival analysis was performed. Moreover, genomic DNA was extracted from the fecal microbes in 30 of these patients, and the differences in the intestinal flora between mutation and non-mutation groups were evaluated using linear discriminant analysis (LDA) Effect size (LEfSe) analysis. Results KRAS gene mutation substantially affected the distant metastasis of colorectal cancer, and the survival prognosis in the non-mutation group was significantly superior compared to the mutation group. The mutation group had a notably higher prevalence of microbes including Roseburia, Parabacteroides, Metascardovia, Staphylococcus, Staphylococcaceae, and Bacillales than the non-mutation group. The presence of microbes in the non-mutation group, such as Clostridiales, Bacteroidetes, Lachnospiraceae, Coprococcus, and Ruminococcaceae was markedly higher than in the mutation group. Firmicutes were negatively correlated with the presence of Actinomyces and Bacteroidetes, while Bacteroidetes were positively associated with the level of Actinomyces. Conclusions In colorectal cancer, KRAS gene mutation can remarkably affect the survival prognosis and change the composition and abundance of intestinal flora, such as Roseburia, Parabacteroides, Metascardovia, Staphylococcus, and Bacillales, thereby influencing tumor development.
Collapse
Affiliation(s)
- Xinke Sui
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Yan Chen
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Baojun Liu
- Department of Medical Oncology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Lianyong Li
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Xin Huang
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Min Wang
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Guodong Wang
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Xiaopei Gao
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Lu Zhang
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Xinwei Bao
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Dengfeng Yang
- Laboratory department, Mian County Hospital, Mian, China
| | - Xiaoying Wang
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Changqing Zhong
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
694
|
Cortellini A, Tucci M, Adamo V, Stucci LS, Russo A, Tanda ET, Spagnolo F, Rastelli F, Bisonni R, Santini D, Russano M, Anesi C, Giusti R, Filetti M, Marchetti P, Botticelli A, Gelibter A, Occhipinti MA, Marconcini R, Vitale MG, Nicolardi L, Chiari R, Bareggi C, Nigro O, Tuzi A, De Tursi M, Petragnani N, Pala L, Bracarda S, Macrini S, Inno A, Zoratto F, Veltri E, Di Cocco B, Mallardo D, Vitale MG, Pinato DJ, Porzio G, Ficorella C, Ascierto PA. Integrated analysis of concomitant medications and oncological outcomes from PD-1/PD-L1 checkpoint inhibitors in clinical practice. J Immunother Cancer 2020; 8:jitc-2020-001361. [PMID: 33154150 PMCID: PMC7646355 DOI: 10.1136/jitc-2020-001361] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background Concomitant medications, such as steroids, proton pump inhibitors (PPI) and antibiotics, might affect clinical outcomes with immune checkpoint inhibitors. Methods We conducted a multicenter observational retrospective study aimed at evaluating the impact of concomitant medications on clinical outcomes, by weighing their associations with baseline clinical characteristics (including performance status, burden of disease and body mass index) and the underlying causes for their prescription. This analysis included consecutive stage IV patients with cancer, who underwent treatment with single agent antiprogrammed death-1/programmed death ligand-1 (PD-1/PD-L1) with standard doses and schedules at the medical oncology departments of 20 Italian institutions. Each medication taken at the immunotherapy initiation was screened and collected into key categories as follows: corticosteroids, antibiotics, gastric acid suppressants (including proton pump inhibitors - PPIs), statins and other lipid-lowering agents, aspirin, anticoagulants, non-steroidal anti-inflammatory drugs (NSAIDs), ACE inhibitors/Angiotensin II receptor blockers, calcium antagonists, β-blockers, metformin and other oral antidiabetics, opioids. Results From June 2014 to March 2020, 1012 patients were included in the analysis. Primary tumors were: non-small cell lung cancer (52.2%), melanoma (26%), renal cell carcinoma (18.3%) and others (3.6%). Baseline statins (HR 1.60 (95% CI 1.14 to 2.25), p=0.0064), aspirin (HR 1.47 (95% CI 1.04 to 2.08, p=0.0267) and β-blockers (HR 1.76 (95% CI 1.16 to 2.69), p=0.0080) were confirmed to be independently related to an increased objective response rate. Patients receiving cancer-related steroids (HR 1.72 (95% CI 1.43 to 2.07), p<0.0001), prophylactic systemic antibiotics (HR 1.85 (95% CI 1.23 to 2.78), p=0.0030), prophylactic gastric acid suppressants (HR 1.29 (95% CI 1.09 to 1.53), p=0.0021), PPIs (HR 1.26 (95% CI 1.07 to 1.48), p=0.0050), anticoagulants (HR 1.43 (95% CI: 1.16 to 1.77), p=0.0007) and opioids (HR 1.71 (95% CI 1.28 to 2.28), p=0.0002) were confirmed to have a significantly higher risk of disease progression. Patients receiving cancer-related steroids (HR 2.16 (95% CI 1.76 to 2.65), p<0.0001), prophylactic systemic antibiotics (HR 1.93 (95% CI 1.25 to 2.98), p=0.0030), prophylactic gastric acid suppressants (HR 1.29 (95% CI 1.06 to 1.57), p=0.0091), PPI (HR 1.26 (95% CI 1.04 to 1.52), p=0.0172), anticoagulants (HR 1.45 (95% CI 1.14 to 1.84), p=0.0024) and opioids (HR 1.53 (95% CI 1.11 to 2.11), p=0.0098) were confirmed to have a significantly higher risk of death. Conclusion We confirmed the association between baseline steroids administered for cancer-related indication, systemic antibiotics, PPIs and worse clinical outcomes with PD-1/PD-L1 checkpoint inhibitors, which can be assumed to have immune-modulating detrimental effects.
Collapse
Affiliation(s)
- Alessio Cortellini
- Department of Biotechnology and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy .,Medical Oncology, St. Salvatore Hospital, L'Aquila, Italy
| | - Marco Tucci
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari, Bary, Italy.,National Cancer Research Center, Tumori Institute IRCCS Giovanni Paolo II, Bari, Italy
| | - Vincenzo Adamo
- Medical Oncology, Department of Human Pathology, A.O. Papardo, University of Messina, Messina, Italy
| | - Luigia Stefania Stucci
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari, Bary, Italy
| | - Alessandro Russo
- Medical Oncology, Department of Human Pathology, A.O. Papardo, University of Messina, Messina, Italy
| | | | | | | | - Renato Bisonni
- Medical Oncology, ASUR District Area 4 Fermo, Fermo, Italy
| | | | - Marco Russano
- Medical Oncology, Campus Bio-Medico University, Rome, Italy
| | - Cecilia Anesi
- Medical Oncology, Campus Bio-Medico University, Rome, Italy
| | - Raffaele Giusti
- Medical Oncology Unit, Sant'Andrea Hospital of Rome, Rome, Italy
| | - Marco Filetti
- Medical Oncology Unit, Sant'Andrea Hospital of Rome, Rome, Italy
| | - Paolo Marchetti
- Medical Oncology Unit, Sant'Andrea Hospital of Rome, Rome, Italy.,Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Medical Oncology Unit B, Policlinico Umberto I, Sapienza University of Rome, Roma, Italy
| | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alain Gelibter
- Medical Oncology Unit B, Policlinico Umberto I, Sapienza University of Rome, Roma, Italy
| | | | | | | | - Linda Nicolardi
- UOC Oncologia Padova Sud, Azienda ULSS 6 Euganea, Padova, Italy
| | - Rita Chiari
- UOC Oncologia Padova Sud, Azienda ULSS 6 Euganea, Padova, Italy
| | - Claudia Bareggi
- Medical Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore di Milano Policlinico, Milano, Italy
| | - Olga Nigro
- Medical Oncology, ASST Sette Laghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Alessandro Tuzi
- Medical Oncology, ASST Sette Laghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Michele De Tursi
- Department of Medical, Oral and Biotechnological Sciences, Gabriele d'Annunzio University of Chieti and Pescara, Chieti, Italy
| | - Nicola Petragnani
- Department of Psychological, Health and Territorial Sciences, University G. D'Annunzio of Chieti and Pescara, Chieti, Italy
| | - Laura Pala
- Division of Medical Oncology for Melanoma, Sarcoma and Rare Tumors, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Sergio Bracarda
- Medical Oncology, Azienda Ospedaliera S. Maria, Terni, Italy
| | - Serena Macrini
- Medical Oncology, Azienda Ospedaliera S. Maria, Terni, Italy
| | - Alessandro Inno
- Oncology Unit, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar, Italy
| | | | - Enzo Veltri
- Medical Oncology, Santa Maria Goretti Hospital, Latina, Italy
| | | | - Domenico Mallardo
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| | - Maria Grazia Vitale
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| | - David James Pinato
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | | | - Corrado Ficorella
- Department of Biotechnology and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.,Medical Oncology, St. Salvatore Hospital, L'Aquila, Italy
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| |
Collapse
|
695
|
Gastman B, Agarwal PK, Berger A, Boland G, Broderick S, Butterfield LH, Byrd D, Fecci PE, Ferris RL, Fong Y, Goff SL, Grabowski MM, Ito F, Lim M, Lotze MT, Mahdi H, Malafa M, Morris CD, Murthy P, Neves RI, Odunsi A, Pai SI, Prabhakaran S, Rosenberg SA, Saoud R, Sethuraman J, Skitzki J, Slingluff CL, Sondak VK, Sunwoo JB, Turcotte S, Yeung CC, Kaufman HL. Defining best practices for tissue procurement in immuno-oncology clinical trials: consensus statement from the Society for Immunotherapy of Cancer Surgery Committee. J Immunother Cancer 2020; 8:e001583. [PMID: 33199512 PMCID: PMC7670953 DOI: 10.1136/jitc-2020-001583] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy is now a cornerstone for cancer treatment, and much attention has been placed on the identification of prognostic and predictive biomarkers. The success of biomarker development is dependent on accurate and timely collection of biospecimens and high-quality processing, storage and shipping. Tumors are also increasingly used as source material for the generation of therapeutic T cells. There have been few guidelines or consensus statements on how to optimally collect and manage biospecimens and source material being used for immunotherapy and related research. The Society for Immunotherapy of Cancer Surgery Committee has brought together surgical experts from multiple subspecialty disciplines to identify best practices and to provide consensus on how best to access and manage specific tissues for immuno-oncology treatments and clinical investigation. In addition, the committee recommends early integration of surgeons and other interventional physicians with expertise in biospecimen collection, especially in clinical trials, to optimize the quality of tissue and the validity of correlative clinical studies in cancer immunotherapy.
Collapse
Affiliation(s)
- Brian Gastman
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | - Piyush K Agarwal
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Adam Berger
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Genevieve Boland
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephen Broderick
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
- Department of Surgery, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Lisa H Butterfield
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | - David Byrd
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Peter E Fecci
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Robert L Ferris
- Departments of Otolaryngology, Immunology, and Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | | | - Matthew M Grabowski
- Department of Neurosurgery, Duke Center for Brain and Spine Metastasis, Durham, North Carolina, USA
| | - Fumito Ito
- Center for Immunotherapy, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Michael Lim
- Departments of Neurosurgery, Oncology, Radiation Oncology, and Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Haider Mahdi
- OBGYN and Women's Health Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Carol D Morris
- Division of Orthopaedic Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pranav Murthy
- Department of Surgery, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rogerio I Neves
- Department of Surgery, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Adekunle Odunsi
- Departments of Immunology and Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Sara I Pai
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sangeetha Prabhakaran
- Division of Surgical Oncology, Department of Surgery, UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | | | - Ragheed Saoud
- Department of Surgery, University of Chicago Hospitals, Chicago, Illinois, United States
| | | | - Joseph Skitzki
- Departments of Surgical Oncology and Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Craig L Slingluff
- Department of Surgery, Division of Surgical Oncology, Breast and Melanoma Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - John B Sunwoo
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA
| | - Simon Turcotte
- Surgery Department, Centre Hospitalier de l'Universite de Montreal, Montreal, Quebec, Canada
| | - Cecilia Cs Yeung
- Department of Pathology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Howard L Kaufman
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Immuneering Corp, Cambridge, Massachusetts, USA
| |
Collapse
|
696
|
Caenepeel C, Sadat Seyed Tabib N, Vieira-Silva S, Vermeire S. Review article: how the intestinal microbiota may reflect disease activity and influence therapeutic outcome in inflammatory bowel disease. Aliment Pharmacol Ther 2020; 52:1453-1468. [PMID: 32969507 DOI: 10.1111/apt.16096] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/08/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal bacteria produce metabolites and by-products necessary for homeostasis. Imbalance in this equilibrium is linked to multiple pathologies including inflammatory bowel disease (IBD). The role of the gut microbiota in determining treatment response is becoming apparent, and may act as biomarker for efficacy. AIM To describe knowledge about the intestinal microbiota on disease severity and treatment outcomes in IBD METHODS: Descriptive review using PubMed to identify literature on the intestinal microbiota in IBD RESULTS: Severe IBD has a less diverse microbiota with fewer commensal microbiota communities and more opportunistic pathogenic bacteria originating from the oral cavity or respiratory tract. IBD treatments can alter gut microbiota composition, but in vitro/in vivo studies are needed to prove causation. A diversification of the microbiota is observed during remission. Patients with a more diverse baseline microbiome and higher microbial diversity show better response to anti-tumour necrosis factor-α, vedolizumab and ustekinumab therapy. Higher abundance of short chain fatty acid-producing bacteria, fewer mucus-colonising bacteria and lower abundance of pro-inflammatory bacteria have also been associated with a favourable outcome. Predictive models, based on a combination of microbiota, clinical data and serological markers, have good accuracy for treatment outcome and disease severity. CONCLUSION The intestinal microbiota in IBD carries a set of promising biomarkers of disease activity and prediction of therapeutic outcome. Current insights may also help in designing microbiota modulation strategies to improve outcomes in IBD.
Collapse
Affiliation(s)
| | | | - Sara Vieira-Silva
- Department of Microbiology and Immunology, Laboratory of Molecular Bacteriology, Rega Institute for Medical Research, VIB, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases & Metabolism, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium.,Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
697
|
Yang L, Hou K, Zhang B, Ouyang C, Lin A, Xu S, Ke D, Fang L, Chen Q, Wu J, Yan C, Lian Y, Jiang T, He J, Wang H, Fu Y, Xiao C, Chen Z. Preservation of the fecal samples at ambient temperature for microbiota analysis with a cost-effective and reliable stabilizer EffcGut. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 741:140423. [PMID: 32615432 DOI: 10.1016/j.scitotenv.2020.140423] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/31/2020] [Accepted: 06/20/2020] [Indexed: 02/05/2023]
Abstract
With the increasing researches on the role of gut microbiota in human health and disease, appropriate storage method of fecal samples at ambient temperature would conveniently guarantee the precise and reliable microbiota results. Nevertheless, less choice of stabilizer that is cost-efficient and feasible to be used in longer preservation period obstructed the large-scale metagenomics studies. Here, we evaluated the efficacy of a guanidine isothiocyanate-based reagent method EffcGut and compared it with the other already used storage method by means of 16S rRNA gene sequencing technology. We found that guanidine isothiocyanate-based reagent method at ambient temperature was not inferior to OMNIgene·GUT OM-200 and it could retain the similar bacterial community as that of -80 °C within 24 weeks. Furthermore, bacterial diversity and community structure difference were compared among different sample fraction (supernatant, suspension and precipitate) preserved in EffcGut and -80 °C. We found that supernatant under the preservation of EffcGut retained the similar community structure and composition as that of the low temperature preservation method.
Collapse
Affiliation(s)
- Luxi Yang
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystem, School of Life Sciences, Xiamen University, Xiamen, China; Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Kaijian Hou
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, the First Affiliated Hospital of Shantou University Medical College, China
| | - Bangzhou Zhang
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystem, School of Life Sciences, Xiamen University, Xiamen, China; Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Cong Ouyang
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Aiqiang Lin
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Shuangbin Xu
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Dongxian Ke
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Lujing Fang
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Qiongyun Chen
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Jingtong Wu
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Changsheng Yan
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Yifan Lian
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China
| | - Tao Jiang
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Jianquan He
- Department of Rehabilitation, Zhongshan Hospital, Xiamen University, China
| | - Han Wang
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Yousi Fu
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystem, School of Life Sciences, Xiamen University, Xiamen, China
| | | | - Zhangran Chen
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystem, School of Life Sciences, Xiamen University, Xiamen, China; Institute for Microbial Ecology, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
698
|
Yu Q, Wu L, Ji J, Feng J, Dai W, Li J, Wu J, Guo C. Gut Microbiota, Peroxisome Proliferator-Activated Receptors, and Hepatocellular Carcinoma. J Hepatocell Carcinoma 2020; 7:271-288. [PMID: 33150145 PMCID: PMC7605923 DOI: 10.2147/jhc.s277870] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/10/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world. HCC incidence rate is sixth and mortality is fourth worldwide. However, HCC pathogenesis and molecular mechanisms remain unclear. The incidence of HCC is associated with genetic, environmental, and metabolic factors. The role of gut microbiota in the pathogenesis of HCC has attracted researchers’ attention because of anatomical and functional interactions between liver and intestine. Studies have demonstrated the involvement of gut microbiota in the development of HCC and chronic liver diseases, such as alcoholic liver disease (ALD), nonalcoholic fatty liver disease (NAFLD), and liver cirrhosis. Peroxisome proliferator-activated receptors (PPARs) are a group of receptors with diverse biological functions. Natural and synthetic PPAR agonists show potential for treatment of NAFLD, liver fibrosis, and HCC. Recent studies have demonstrated that PPARs take part in gut microbiota inhabitation and adaptation. This manuscript reviews the role of gut microbiota in the development of HCC and precancerous diseases, the role of PPARs in modulation of gut microbiota and HCC, and potential of gut microbiota for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, People's Republic of China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, People's Republic of China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, People's Republic of China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, People's Republic of China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, People's Republic of China
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, People's Republic of China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, People's Republic of China.,Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200336, People's Republic of China
| | - Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, People's Republic of China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, People's Republic of China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, People's Republic of China
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, People's Republic of China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, People's Republic of China
| |
Collapse
|
699
|
Zhao Z, Fei K, Bai H, Wang Z, Duan J, Wang J. Metagenome association study of the gut microbiome revealed biomarkers linked to chemotherapy outcomes in locally advanced and advanced lung cancer. Thorac Cancer 2020; 12:66-78. [PMID: 33111503 PMCID: PMC7779204 DOI: 10.1111/1759-7714.13711] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 12/24/2022] Open
Abstract
Background The gut microbiome is important in the development and immunotherapy efficacy of lung cancer. However, the relationship between the intestinal flora and chemotherapy outcomes remains unclear and was investigated in this study. Methods We analyzed baseline stool samples from patients with locally advanced and advanced lung cancer before chemotherapy treatment, through metagenomics of the gut microbiota. The composition, diversity, function, and metabolic pathway analysis were compared among patients with different clinical outcomes. Results From 64 patients, 33 responded to treatment (responders) and 31 did not (nonresponders). Streptococcus mutans and Enterococcus casseliflavus were enriched in responders (P < 0.05), while 11 bacteria including Leuconostoc lactis and Eubacterium siraeum were enriched in nonresponders (P < 0.05) by variance analysis. Responders were associated with significantly higher Acidobacteria and Granulicella, while Streptococcus oligofermentans, Megasphaera micronuciformis, and Eubacterium siraeum were more abundant in nonresponders by Lefse analysis. Streptococcus mutans and Enterococcus casseliflavus were further identified as bacterial markers relevant to responders using unsupervised clustering, and Leuconostoc lactis and Eubacterium siraeum were related to nonresponders. The L‐glutamate degradation VIII pathway was enriched in responders (P = 0.014), and the C4 photosynthetic carbon assimilation cycle, reductive TCA cycle I, and hexitol fermentation to lactate, formate, ethanol, and acetate were enriched in nonresponders (P < 0.05). Additionally, significant associations of bacterial species with clinical phenotypes were observed by Spearman correlation analysis. Conclusions The specific gut microbiome of patients with lung cancer might be connected to the clinical outcomes of chemotherapy. Key points
Collapse
Affiliation(s)
- Zhe Zhao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kailun Fei
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Bai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhijie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Duan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
700
|
Irfan M, Delgado RZR, Frias-Lopez J. The Oral Microbiome and Cancer. Front Immunol 2020; 11:591088. [PMID: 33193429 PMCID: PMC7645040 DOI: 10.3389/fimmu.2020.591088] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/29/2020] [Indexed: 12/24/2022] Open
Abstract
There is mounting evidence that members of the human microbiome are highly associated with a wide variety of cancer types. Among oral cancers, oral squamous cell carcinoma (OSCC) is the most prevalent and most commonly studied, and it is the most common malignancy of the head and neck worldwide. However, there is a void regarding the role that the oral microbiome may play in OSCC. Previous studies have not consistently found a characteristic oral microbiome composition associated with OSCC. Although a direct causality has not been proven, individual members of the oral microbiome are capable of promoting various tumorigenic functions related to cancer development. Two prominent oral pathogens, Porphyromonas gingivalis, and Fusobacterium nucleatum can promote tumor progression in mice. P. gingivalis infection has been associated with oro-digestive cancer, increased oral cancer invasion, and proliferation of oral cancer stem cells. The microbiome can influence the evolution of the disease by directly interacting with the human body and significantly altering the response and toxicity to various forms of cancer therapy. Recent studies have shown an association of certain phylogenetic groups with the immunotherapy treatment outcomes of certain tumors. On the other side of the coin, recently it has been a resurgence in interest on the potential use of bacteria to cure cancer. These kinds of treatments were used in the late nineteenth and early twentieth centuries as the first line of defense against cancer in some hospitals but later displaced by other types of treatments such as radiotherapy. Currently, organisms such as Salmonella typhimurium and Clostridium spp. have been used for targeted strategies as potential vectors to treat cancer. In this review, we briefly summarize our current knowledge of the role of the oral microbiome, focusing on its bacterial fraction, in cancer in general and in OSCC more precisely, and a brief description of the potential use of bacteria to target tumors.
Collapse
Affiliation(s)
- Muhammad Irfan
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, United States
| | | | - Jorge Frias-Lopez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, United States
| |
Collapse
|