601
|
Fan Z, Ross RP, Stanton C, Hou B, Zhao J, Zhang H, Yang B, Chen W. Lactobacillus casei CCFM1074 Alleviates Collagen-Induced Arthritis in Rats via Balancing Treg/Th17 and Modulating the Metabolites and Gut Microbiota. Front Immunol 2021; 12:680073. [PMID: 34079556 PMCID: PMC8165437 DOI: 10.3389/fimmu.2021.680073] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 04/14/2021] [Indexed: 01/04/2023] Open
Abstract
Gut microbiota and their influence on metabolites are receiving increasing attentions in autoimmune diseases including rheumatoid arthritis (RA). Probiotics become a promising manipulator to prevent or attenuate the progression of arthritis, some evidences suggesting that lactobacilli treatment influence the responses to RA therapy but the underlying mechanisms are limited. By using a collagen-induced arthritis (CIA) rats, the study assessed the effects of two L. casei strains (CCFM1074, CCFM1075) on the immune responses, gut microbiota and plasma metabolites via an integrated cross-omics approach including fecal 16S rRNA high-throughput sequencing and plasma metabolomics. The genome of the two strains was analyzed and compared using whole-genome sequencing approach to further confirm biology functions. CCFM1074 reduced arthritic symptoms while CCFM1075 did not, though both strains down-regulated the plasma IL-6 and Th17 cells proportion. CCFM1074 enhanced the proportion of Treg cells in mesenteric lymph nodes which was significantly associated with SCFAs upregulation, as well as with genomic evidence that CCFM1074 possesses more functional genes involved in carbohydrate metabolism. Moreover, CCFM1074 regulated the gut microbiota, including modulating community structure, decreasing the abundance of Alistipes and Parabacteroides and increasing the abundance of Oscillibacter. The differential metabolites modulated by CCFM1074 including eicosapentaenoic acid and docosapentaenoic acid which involved in unsaturated fatty acids metabolism. Furthermore, alterations of gut microbial community were correlated with the plasma metabolome. In summary, L. casei CCFM1074 alleviated arthritis via rebalancing gut microbiota, immune responses and plasma metabolites.
Collapse
Affiliation(s)
- Zhexin Fan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - R Paul Ross
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China.,Teagasc Food Research Centre, Moorepark, Co., Cork, Ireland
| | - Bao Hou
- Department of Basic Medicine, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China.,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
602
|
Borowsky J, Haruki K, Lau MC, Dias Costa A, Väyrynen JP, Ugai T, Arima K, da Silva A, Felt KD, Zhao M, Gurjao C, Twombly TS, Fujiyoshi K, Väyrynen SA, Hamada T, Mima K, Bullman S, Harrison TA, Phipps AI, Peters U, Ng K, Meyerhardt JA, Song M, Giovannucci EL, Wu K, Zhang X, Freeman GJ, Huttenhower C, Garrett WS, Chan AT, Leggett BA, Whitehall VLJ, Walker N, Brown I, Bettington M, Nishihara R, Fuchs CS, Lennerz JK, Giannakis M, Nowak JA, Ogino S. Association of Fusobacterium nucleatum with Specific T-cell Subsets in the Colorectal Carcinoma Microenvironment. Clin Cancer Res 2021; 27:2816-2826. [PMID: 33632927 PMCID: PMC8127352 DOI: 10.1158/1078-0432.ccr-20-4009] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/09/2021] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE While evidence indicates that Fusobacterium nucleatum (F. nucleatum) may promote colorectal carcinogenesis through its suppressive effect on T-cell-mediated antitumor immunity, the specific T-cell subsets involved remain uncertain. EXPERIMENTAL DESIGN We measured F. nucleatum DNA within tumor tissue by quantitative PCR on 933 cases (including 128 F. nucleatum-positive cases) among 4,465 incident colorectal carcinoma cases in two prospective cohorts. Multiplex immunofluorescence combined with digital image analysis and machine learning algorithms for CD3, CD4, CD8, CD45RO (PTPRC isoform), and FOXP3 measured various T-cell subsets. We leveraged data on Bifidobacterium, microsatellite instability (MSI), tumor whole-exome sequencing, and M1/M2-type tumor-associated macrophages [TAM; by CD68, CD86, IRF5, MAF, and MRC1 (CD206) multimarker assay]. Using the 4,465 cancer cases and inverse probability weighting method to control for selection bias due to tissue availability, multivariable-adjusted logistic regression analysis assessed the association between F. nucleatum and T-cell subsets. RESULTS The amount of F. nucleatum was inversely associated with tumor stromal CD3+ lymphocytes [multivariable OR, 0.47; 95% confidence interval (CI), 0.28-0.79, for F. nucleatum-high vs. -negative category; P trend = 0.0004] and specifically stromal CD3+CD4+CD45RO+ cells (corresponding multivariable OR, 0.52; 95% CI, 0.32-0.85; P trend = 0.003). These relationships did not substantially differ by MSI status, neoantigen load, or exome-wide tumor mutational burden. F. nucleatum was not significantly associated with tumor intraepithelial T cells or with M1 or M2 TAMs. CONCLUSIONS The amount of tissue F. nucleatum is associated with lower density of stromal memory helper T cells. Our findings provide evidence for the interactive pathogenic roles of microbiota and specific immune cells.
Collapse
Affiliation(s)
- Jennifer Borowsky
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Conjoint Gastroenterology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Koichiro Haruki
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mai Chan Lau
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Juha P Väyrynen
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Tomotaka Ugai
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kota Arima
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Annacarolina da Silva
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kristen D Felt
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Melissa Zhao
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Carino Gurjao
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Tyler S Twombly
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kenji Fujiyoshi
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sara A Väyrynen
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kosuke Mima
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Susan Bullman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Tabitha A Harrison
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Amanda I Phipps
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kana Wu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Wendy S Garrett
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Barbara A Leggett
- Conjoint Gastroenterology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- The Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Vicki L J Whitehall
- Conjoint Gastroenterology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Conjoint Internal Medicine Laboratory, Pathology Queensland, Queensland Health, Brisbane, Queensland, Australia
| | - Neal Walker
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Envoi Specialist Pathologists, Brisbane, Queensland, Australia
| | - Ian Brown
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Envoi Specialist Pathologists, Brisbane, Queensland, Australia
| | - Mark Bettington
- Conjoint Gastroenterology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Envoi Specialist Pathologists, Brisbane, Queensland, Australia
| | - Reiko Nishihara
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, Connecticut
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
- Smilow Cancer Hospital, New Haven, Connecticut
| | - Jochen K Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, Massachusetts
| |
Collapse
|
603
|
Geum MJ, Kim C, Kang JE, Choi JH, Kim JS, Son ES, Lim SM, Rhie SJ. Broad-Spectrum Antibiotic Regimen Affects Survival in Patients Receiving Nivolumab for Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2021; 14:ph14050445. [PMID: 34066877 PMCID: PMC8151442 DOI: 10.3390/ph14050445] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 01/17/2023] Open
Abstract
Antibiotic-induced dysbiosis may affect the efficacy of immune checkpoint inhibitors. We investigated the impact of antibiotics on the clinical outcomes of nivolumab in patients with non-small cell lung cancer (NSCLC). Patients who received nivolumab for NSCLC between July 2015 and June 2018 and who were followed up until June 2020 were included in a retrospective cohort analysis. Of 140 eligible patients, 70 were on antibiotics. Overall survival (OS) was shorter in patients on antibiotics (ABX) compared to those not on antibiotics (NoABX) (p = 0.014). OS was negatively associated with piperacillin/tazobactam (PTZ) (HR = 3.31, 95% CI: 1.77–6.18), days of therapy (DOT) ≥ 2 weeks (HR = 2.56, 95% CI: 1.30–5.22) and DOT of PTZ. The defined daily dose (DDD) in PTZ (r = 0.27) and glycopeptides (r = 0.21) showed weak correlations with mortality. There was no difference in progression-free survival (PFS) between ABX and NoABX; however, PFS was negatively associated with the antibiotic class PTZ and DOT of PTZ. Therefore, the use of a broad-spectrum antibiotic, such as PTZ, the long-term use of antibiotics more than 2 weeks in total and the large amount of defined daily dose of specific antibiotics were associated with decreased survival in patients receiving nivolumab for NSCLC.
Collapse
Affiliation(s)
- Min Jung Geum
- The Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
- Department of Pharmacy, Severance Hospital, Yonsei University Health System, Seoul 03722, Korea; (J.S.K.); (E.S.S.)
| | - Chungsoo Kim
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea;
| | - Ji Eun Kang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea;
- Department of Pharmacy, National Medical Center, Seoul 04564, Korea
| | - Jae Hee Choi
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
- Department of Pharmacy, Konkuk University Medical Center, Seoul 05030, Korea
| | - Jae Song Kim
- Department of Pharmacy, Severance Hospital, Yonsei University Health System, Seoul 03722, Korea; (J.S.K.); (E.S.S.)
| | - Eun Sun Son
- Department of Pharmacy, Severance Hospital, Yonsei University Health System, Seoul 03722, Korea; (J.S.K.); (E.S.S.)
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Cancer Center, Seoul 03722, Korea;
| | - Sandy Jeong Rhie
- The Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea;
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
- Correspondence:
| |
Collapse
|
604
|
Corral-Jara KF, Rosas da Silva G, Fierro NA, Soumelis V. Modeling the Th17 and Tregs Paradigm: Implications for Cancer Immunotherapy. Front Cell Dev Biol 2021; 9:675099. [PMID: 34026764 PMCID: PMC8137995 DOI: 10.3389/fcell.2021.675099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
CD4 + T cell differentiation is governed by gene regulatory and metabolic networks, with both networks being highly interconnected and able to adapt to external stimuli. Th17 and Tregs differentiation networks play a critical role in cancer, and their balance is affected by the tumor microenvironment (TME). Factors from the TME mediate recruitment and expansion of Th17 cells, but these cells can act with pro or anti-tumor immunity. Tregs cells are also involved in tumor development and progression by inhibiting antitumor immunity and promoting immunoevasion. Due to the complexity of the underlying molecular pathways, the modeling of biological systems has emerged as a promising solution for better understanding both CD4 + T cell differentiation and cancer cell behavior. In this review, we present a context-dependent vision of CD4 + T cell transcriptomic and metabolic network adaptability. We then discuss CD4 + T cell knowledge-based models to extract the regulatory elements of Th17 and Tregs differentiation in multiple CD4 + T cell levels. We highlight the importance of complementing these models with data from omics technologies such as transcriptomics and metabolomics, in order to better delineate existing Th17 and Tregs bifurcation mechanisms. We were able to recompilate promising regulatory components and mechanisms of Th17 and Tregs differentiation under normal conditions, which we then connected with biological evidence in the context of the TME to better understand CD4 + T cell behavior in cancer. From the integration of mechanistic models with omics data, the transcriptomic and metabolomic reprograming of Th17 and Tregs cells can be predicted in new models with potential clinical applications, with special relevance to cancer immunotherapy.
Collapse
Affiliation(s)
- Karla F. Corral-Jara
- Computational Systems Biology Team, Institut de Biologie de l’Ecole Normale Supérieure, CNRS UMR 8197, INSERM U1024, Ecole Normale Supérieure, PSL Research University, Paris, France
| | | | - Nora A. Fierro
- Department of Immunology, Biomedical Research Institute, National Autonomous University of Mexico, Mexico City, Mexico
| | - Vassili Soumelis
- Université de Paris, INSERM U976, France and AP-HP, Hôpital Saint-Louis, Immunology-Histocompatibility Department, Paris, France
| |
Collapse
|
605
|
Lyu Y, Yang H, Chen L. Metabolic regulation on the immune environment of glioma through gut microbiota. Semin Cancer Biol 2021; 86:990-997. [PMID: 33971263 DOI: 10.1016/j.semcancer.2021.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/08/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
The gut-brain axis has paved our way in understanding varieties of disease. The gut microbiota especially the bacterial population plays critical roles in immune system development and function. Glioma comprises 80 percent of malignant brain cancer and glioblastoma (GBM) is the most malignant kind. GBM has a reputation for its suppressive immune environment and poor patient prognosis. Moreover, altered metabolites from gut microbiota affect both systemic immune and central nervous system (CNS) immunity. Here we will focus on the crosstalk between gut microbiota and GBM, and further explore how this communication contributes to glioma initiation and development. Finally, we highlight the latest insights on the metabolic regulation of immunity through gut microbiota, which provides a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Yingying Lyu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, PR China; Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| |
Collapse
|
606
|
Zhu G, Su H, Johnson CH, Khan SA, Kluger H, Lu L. Intratumour microbiome associated with the infiltration of cytotoxic CD8+ T cells and patient survival in cutaneous melanoma. Eur J Cancer 2021; 151:25-34. [PMID: 33962358 DOI: 10.1016/j.ejca.2021.03.053] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/11/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The gut microbiome plays an important role in systemic inflammation and immune response. Microbes can translocate and reside in tumour niches. However, it is unclear how the intratumour microbiome affects immunity in human cancer. The purpose of this study was to investigate the association between intratumour bacteria, infiltrating CD8+ T cells and patient survival in cutaneous melanoma. METHODS Using The Cancer Genome Altas's cutaneous melanoma RNA sequencing data, levels of intratumour bacteria and infiltrating CD8+ T cells were determined. Correlation between intratumour bacteria and infiltrating CD8+ T cells or chemokine gene expression and survival analysis of infiltrating CD8+ T cells and Lachnoclostridium in cutaneous melanoma were performed. RESULTS Patients with low levels of CD8+ T cells have significantly shorter survival than those with high levels. The adjusted hazard ratio was 1.57 (low vs high) (95% confidence interval: 1.17-2.10, p = 0.002). Intratumour bacteria of the Lachnoclostridium genus ranked top in a positive association with infiltrating CD8+ T cells (correlation coefficient = 0.38, p = 9.4 × 10-14), followed by Gelidibacter (0.31, p = 1.13 × 10-9), Flammeovirga (0.29, p = 1.96 × 10-8) and Acinetobacter (0.28, p = 8.94 × 10-8). These intratumour genera positively correlated with chemokine CXCL9, CXCL10 and CCL5 expression. The high Lachnoclostridium load significantly reduced the mortality risk (p = 0.0003). However, no statistically significant correlation was observed between intratumour Lachnoclostridium abundance and the levels of either NK, B or CD4+ T cells. CONCLUSION Intratumour-residing gut microbiota could modulate chemokine levels and affect CD8+ T-cell infiltration, consequently influencing patient survival in cutaneous melanoma. Manipulating the intratumour gut microbiome may benefit patient outcomes for those undergoing immunotherapy.
Collapse
Affiliation(s)
- Gongjian Zhu
- Gansu Provincial Academy of Medical Science, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China; Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Haixiang Su
- Gansu Provincial Academy of Medical Science, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Sajid A Khan
- Department of Surgery, Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Harriet Kluger
- Department of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Yale Cancer Center, Yale University, New Haven, CT, USA.
| |
Collapse
|
607
|
Sims TT, Colbert LE, Klopp AH. The Role of the Cervicovaginal and Gut Microbiome in Cervical Intraepithelial Neoplasia and Cervical Cancer. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2021; 4:72-78. [PMID: 35663536 PMCID: PMC9153260 DOI: 10.36401/jipo-20-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 04/19/2023]
Abstract
The microbiome, which refers to the microbiota within a host and their collective genomes, has recently been demonstrated to play a critical role in cancer progression, metastasis, and therapeutic response. The microbiome is known to affect host immunity, but its influence on human papilloma virus (HPV) gynecologic malignancies remains limited and poorly understood. To date, studies have largely focused on the cervicovaginal microbiome; however, there is growing evidence that the gut microbiome may interact and substantially affect therapeutic response in gynecologic cancers. Importantly, new developments in microbiome sequencing and advanced bioinformatics technologies have enabled rapid advances in our understanding of the gut and local tumor microbiota. In this review, we examine the evidence supporting the role of the microbiome in HPV-associated cervical intraepithelial neoplasia (CIN) and cervical cancer, explore characteristics that influence and shape the host microbiota that impact HPV-driven carcinogenesis, and highlight potential approaches and considerations for future and ongoing research of the microbiome's effect on HPV-associated cancer.
Collapse
Affiliation(s)
- Travis T. Sims
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren E. Colbert
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ann H. Klopp
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
608
|
Sehrawat N, Yadav M, Singh M, Kumar V, Sharma VR, Sharma AK. Probiotics in microbiome ecological balance providing a therapeutic window against cancer. Semin Cancer Biol 2021; 70:24-36. [PMID: 32574811 DOI: 10.1016/j.semcancer.2020.06.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/25/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
The gut microbiota composition and dietary factors in our food along with the use of prebiotics and probiotics play an important role in the maintenance of human health. A well-balanced gut microbial population is necessary for the host and the microbiota to coexist in a mutually beneficial relationship maintaining homeostasis. Considering the potential of modern technological tools, it is possible nowadays to engineer prebiotic bacteria having a positive influence on the microbiome on one hand while on the other one may have the ease to get rid of the pathogenic proinflammatory microbes or elements causing dysbiosis. Past studies have seen that in cancer there is a loss of inter-microbial relationship cum interactions within microbiota members, the metabolic products produced by them and the host immune system in a microbial ecosystem, leading to dysbiosis. Current review highlights the importance of probiotics in the management of cancer by bringing together majority of the studies together at a single platform and moreover, stresses upon the need to maintain eubiosis in order to evade and inhibit the progression of cancer. Continuous expansion in knowledge about probiotics, their effect on various cancers and the underlying mechanism of action has raised the global scientific interest towards their possible use against different cancers. Furthermore, the article emphasizes upon the need to explore newer therapeutic targets comprising of the microbiome which could further pave the way to the concept of personalized medicines for various kinds of malignancies so as to derive maximum benefits of a treatment modality and to preserve the microbial homeostasis.
Collapse
Affiliation(s)
- Nirmala Sehrawat
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana, 133207, India
| | - Mukesh Yadav
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana, 133207, India
| | - Manoj Singh
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana, 133207, India
| | - Vikas Kumar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana, 133207, India
| | - Var Ruchi Sharma
- Department of Biotechnology, Sri Guru Gobind Singh College Sector-26, Chandigarh, UT 160019, India
| | - Anil K Sharma
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana, 133207, India.
| |
Collapse
|
609
|
Abstract
Introduction: Biliary tract cancer (BTC) is a heterogeneous group of aggressive malignancies comprising intrahepatic cholangiocarcinoma, extrahepatic cholangiocarcinoma, gallbladder cancer, and ampulla of Vater cancer. Although recent years have witnessed the emergence of novel treatment targets, medical therapy remains a compelling challenge in these hepatobiliary malignancies. In order to provide more effective treatment options, immune checkpoint inhibitors (ICIs) are currently under investigation in advanced BTC, with controversial results reported so far.Areas covered: This review provides an overview regarding current scenario of ICIs and immune-based combinations in advanced BTC, where several novel treatments are currently being developed, some of which have suggested interesting efficacy in recent clinical trials. In addition, we provide a report of ongoing Phase I to III clinical trials assessing ICIs and new immunotherapeutic strategies for advanced BTC.Expert opinion: Although immunotherapy has revolutionized the treatment landscape of several hematological and solid tumors, the role of ICIs and immune-based combinations in advanced BTC is still unclear. Despite ICI monotherapy has reported limited efficacy in this setting, the durable responses observed in sporadic cases suggest that testing patients for MMR, MSI, TMB, and PD-L1 expression is warranted. Results of currently ongoing trials are highly awaited.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy.,Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Angela Dalia Ricci
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy.,Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy.,Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
610
|
Isibor PO, Akinduti PA, Aworunse OS, Oyewale JO, Oshamika O, Ugboko HU, Taiwo OS, Ahuekwe EF, Obafemi YD, Onibokun EA, Oziegbe O, Oniha MI, Olopade BK, Atolagbe OM, Adekeye BT, Ajiboye IB, Bello OA, Popoola JO, Ayanda OI, Akinnola OO, Olasehinde GI, Eni AO, Nwinyi OC, Omonhinmin CA, Oranusi SU, Obembe OO. Significance of African Diets in Biotherapeutic Modulation of the Gut Microbiome. Bioinform Biol Insights 2021; 15:11779322211012697. [PMID: 33994782 PMCID: PMC8107938 DOI: 10.1177/11779322211012697] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/04/2021] [Indexed: 12/19/2022] Open
Abstract
Diet plays an essential role in human development and growth, contributing to health and well-being. The socio-economic values, cultural perspectives, and dietary formulation in sub-Saharan Africa can influence gut health and disease prevention. The vast microbial ecosystems in the human gut frequently interrelate to maintain a healthy, well-coordinated cellular and humoral immune signalling to prevent metabolic dysfunction, pathogen dominance, and induction of systemic diseases. The diverse indigenous diets could differentially act as biotherapeutics to modulate microbial abundance and population characteristics. Such modulation could prevent stunted growth, malnutrition, induction of bowel diseases, attenuated immune responses, and mortality, particularly among infants. Understanding the associations between specific indigenous African diets and the predictability of the dynamics of gut bacteria genera promises potential biotherapeutics towards improving the prevention, control, and treatment of microbiome-associated diseases such as cancer, inflammatory bowel disease, obesity, type 2 diabetes, and cardiovascular disease. The dietary influence of many African diets (especially grain-base such as millet, maize, brown rice, sorghum, soya, and tapioca) promotes gut lining integrity, immune tolerance towards the microbiota, and its associated immune and inflammatory responses. A fibre-rich diet is a promising biotherapeutic candidate that could effectively modulate inflammatory mediators' expression associated with immune cell migration, lymphoid tissue maturation, and signalling pathways. It could also modulate the stimulation of cytokines and chemokines involved in ensuring balance for long-term microbiome programming. The interplay between host and gut microbial digestion is complex; microbes using and competing for dietary and endogenous proteins are often attributable to variances in the comparative abundances of Enterobacteriaceae taxa. Many auto-inducers could initiate the process of quorum sensing and mammalian epinephrine host cell signalling system. It could also downregulate inflammatory signals with microbiota tumour taxa that could trigger colorectal cancer initiation, metabolic type 2 diabetes, and inflammatory bowel diseases. The exploitation of essential biotherapeutic molecules derived from fibre-rich indigenous diet promises food substances for the downregulation of inflammatory signalling that could be harmful to gut microbiota ecological balance and improved immune response modulation.
Collapse
Affiliation(s)
- PO Isibor
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - PA Akinduti
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - OS Aworunse
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - JO Oyewale
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - O Oshamika
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - HU Ugboko
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - OS Taiwo
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - EF Ahuekwe
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - YD Obafemi
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - EA Onibokun
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - O Oziegbe
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - MI Oniha
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - BK Olopade
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - OM Atolagbe
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - BT Adekeye
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - IB Ajiboye
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - OA Bello
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - JO Popoola
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - OI Ayanda
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - OO Akinnola
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - GI Olasehinde
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - AO Eni
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - OC Nwinyi
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - CA Omonhinmin
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - SU Oranusi
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| | - OO Obembe
- Biotechnology Research Centre, Department of
Biological Sciences, Covenant University, Ota, Nigeria
| |
Collapse
|
611
|
Sharma V, Fernando V, Letson J, Walia Y, Zheng X, Fackelman D, Furuta S. S-Nitrosylation in Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22094600. [PMID: 33925645 PMCID: PMC8124305 DOI: 10.3390/ijms22094600] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
S-nitrosylation is a selective and reversible post-translational modification of protein thiols by nitric oxide (NO), which is a bioactive signaling molecule, to exert a variety of effects. These effects include the modulation of protein conformation, activity, stability, and protein-protein interactions. S-nitrosylation plays a central role in propagating NO signals within a cell, tissue, and tissue microenvironment, as the nitrosyl moiety can rapidly be transferred from one protein to another upon contact. This modification has also been reported to confer either tumor-suppressing or tumor-promoting effects and is portrayed as a process involved in every stage of cancer progression. In particular, S-nitrosylation has recently been found as an essential regulator of the tumor microenvironment (TME), the environment around a tumor governing the disease pathogenesis. This review aims to outline the effects of S-nitrosylation on different resident cells in the TME and the diverse outcomes in a context-dependent manner. Furthermore, we will discuss the therapeutic potentials of modulating S-nitrosylation levels in tumors.
Collapse
|
612
|
Rajasekaran K, Carey RM, Lin X, Seckar TD, Wei Z, Chorath K, Newman JG, O'Malley BW, Weinstein GS, Feldman MD, Robertson E. The microbiome of HPV-positive tonsil squamous cell carcinoma and neck metastasis. Oral Oncol 2021; 117:105305. [PMID: 33905914 DOI: 10.1016/j.oraloncology.2021.105305] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/01/2021] [Accepted: 04/12/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Oropharyngeal squamous cell carcinoma (OPSCC) has now surpassed cervical cancer as the most common site of HPV-related cancer in the United States. HPV-positive OPSCCs behave differently from HPV-negative tumors and often present with early lymph node involvement. The bacterial microbiome of HPV-associated OPSCC may contribute to carcinogenesis, and certain bacteria may influence the spread of cancer from the primary site to regional lymphatics. OBJECTIVE To determine the bacterial microbiome in patients with HPV-associated, early tonsil SCC and compare them to benign tonsil specimens. METHOD The microbiome of primary tumor specimens and lymph nodes was compared to benign tonsillectomy specimens with pan-pathogen microarray (PathoChip). RESULTS A total of 114 patients were enrolled in the study. Patients with OPSCC had a microbiome that shifted towards more gram-negative. Numerous signatures of bacterial family and species were associated with the primary tumors and lymph nodes of cancer patients, including the urogenital pathogens Proteus mirabilis and Chlamydia trachomatis, Neisseria gonorrhoeae, Shigella dysenteriae, and Orientia tsutsugamushi. CONCLUSION Our results suggest that detection of urogenital pathogens is associated with lymph node metastasis for patients with HPV-positive OPSCCs. Additional studies are necessary to determine the effects of the OPSCC microbiome on disease progression and clinical outcomes.
Collapse
Affiliation(s)
- Karthik Rajasekaran
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States; Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, United States.
| | - Ryan M Carey
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Xiang Lin
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States; Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, United States
| | - Tyler D Seckar
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhi Wei
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States; Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, United States
| | - Kevin Chorath
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Jason G Newman
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Bert W O'Malley
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Gregory S Weinstein
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Erle Robertson
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
613
|
Borchmann S. An atlas of the tissue and blood metagenome in cancer reveals novel links between bacteria, viruses and cancer. MICROBIOME 2021; 9:94. [PMID: 33888160 PMCID: PMC8063312 DOI: 10.1186/s40168-021-01039-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 02/18/2021] [Indexed: 05/02/2023]
Abstract
BACKGROUND Host tissue infections by bacteria and viruses can cause cancer. Known viral carcinogenic mechanisms are disruption of the host genome via genomic integration and expression of oncogenic viral proteins. An important bacterial carcinogenic mechanism is chronic inflammation. Massively parallel sequencing now routinely generates datasets large enough to contain detectable traces of bacterial and viral nucleic acids of taxa that colonize the examined tissue or are integrated into the host genome. However, this hidden resource has not been comprehensively studied in large patient cohorts. METHODS In the present study, 3025 whole genome sequencing datasets and, where available, corresponding RNA-seq datasets are leveraged to gain insight into novel links between viruses, bacteria, and cancer. Datasets were obtained from multiple International Cancer Genome Consortium studies, with additional controls added from the 1000 genome project. A customized pipeline based on KRAKEN was developed and validated to identify bacterial and viral sequences in the datasets. Raw results were stringently filtered to reduce false positives and remove likely contaminants. RESULTS The resulting map confirms known links and expands current knowledge by identifying novel associations. Moreover, the detection of certain bacteria or viruses is associated with profound differences in patient and tumor phenotypes, such as patient age, tumor stage, survival, and somatic mutations in cancer genes or gene expression profiles. CONCLUSIONS Overall, these results provide a detailed, unprecedented map of links between viruses, bacteria, and cancer that can serve as a reference for future studies and further experimental validation. Video Abstract.
Collapse
Affiliation(s)
- Sven Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.
- Cancer Center Cologne Essen - Partner Site Cologne, CIO Cologne, University of Cologne, Cologne, Germany.
- German Hodgkin Study Group, Cologne, Germany.
| |
Collapse
|
614
|
Deeg HJ. Chimerism, the Microenvironment and Control of Leukemia. Front Immunol 2021; 12:652105. [PMID: 33968052 PMCID: PMC8100309 DOI: 10.3389/fimmu.2021.652105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/17/2021] [Indexed: 12/25/2022] Open
Abstract
Transplantation of allogeneic hematopoietic cells faces two barriers: failure of engraftment due to a host versus graft reaction, and the attack of donor cells against the patient, the graft versus host (GVH) reaction. This reaction may lead to GVH disease (GVHD), but in patients transplanted due to leukemia or other malignant disorders, this may also convey the benefit of a graft versus leukemia (GVL) effect. The interplay of transplant conditioning with donor and host cells and the environment in the patient is complex. The microbiome, particularly in the intestinal tract, profoundly affects these interactions, directly and via soluble mediators, which also reach other host organs. The microenvironment is further altered by the modifying effect of malignant cells on marrow niches, favoring the propagation of the malignant cells. The development of stable mixed donor/host chimerism has the potential of GVHD prevention without necessarily increasing the risk of relapse. There has been remarkable progress with novel conditioning regimens and selective T-cell manipulation aimed at securing engraftment while preventing GVHD without ablating the GVL effect. Interventions to alter the microenvironment and change the composition of the microbiome and its metabolic products may modify graft/host interactions, thereby further reducing GVHD, while enhancing the GVL effect. The result should be improved transplant outcome.
Collapse
Affiliation(s)
- H. Joachim Deeg
- Fred Hutchinson Cancer Research Center and the University of Washington, Seattle, WA, United States
| |
Collapse
|
615
|
A Review of Colorectal Cancer in Terms of Epidemiology, Risk Factors, Development, Symptoms and Diagnosis. Cancers (Basel) 2021; 13:cancers13092025. [PMID: 33922197 PMCID: PMC8122718 DOI: 10.3390/cancers13092025] [Citation(s) in RCA: 411] [Impact Index Per Article: 102.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
This review article contains a concise consideration of genetic and environmental risk factors for colorectal cancer. Known risk factors associated with colorectal cancer include familial and hereditary factors and lifestyle-related and ecological factors. Lifestyle factors are significant because of the potential for improving our understanding of the disease. Physical inactivity, obesity, smoking and alcohol consumption can also be addressed through therapeutic interventions. We also made efforts to systematize available literature and data on epidemiology, diagnosis, type and nature of symptoms and disease stages. Further study of colorectal cancer and progress made globally is crucial to inform future strategies in controlling the disease's burden through population-based preventative initiatives.
Collapse
|
616
|
Probiotic Supplement Preparation Relieves Test Anxiety by Regulating Intestinal Microbiota in College Students. DISEASE MARKERS 2021; 2021:5597401. [PMID: 33981369 PMCID: PMC8087991 DOI: 10.1155/2021/5597401] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/18/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Test anxiety creates barriers to learning and performance, which further affects students' social, behavioural, and emotional development. Currently, the medication to treat test anxiety has not been reported yet. Here, we enrolled 120 students to evaluate the effect of probiotic supplement preparation (PSP) on test anxiety from the aspect of the intestinal microbiota. We found that the intake of PSP alleviated the symptoms of depression and anxiety in students with test anxiety by evaluating their mental state using the Hamilton Depression Rating Scale and Hamilton Anxiety Scale. High-throughput sequencing results indicated that the consumption of PSP increased the abundance of Streptococcus and Akkermansia that was lowered by the anxiety state in the intestinal microbiota of students. Meanwhile, taking PSP reduced the level of intestinal pathogens of Fusobacterium and Clostridium as well. In conclusion, our work shows that PSP can reduce test anxiety and restore the disturbed microbiota to the standard level in Chinese college students, rendering the use of PSP a promising strategy for test anxiety.
Collapse
|
617
|
Lee J, Kay K, Troike K, Ahluwalia MS, Lathia JD. Sex Differences in Glioblastoma Immunotherapy Response. Neuromolecular Med 2021; 24:50-55. [PMID: 33864598 DOI: 10.1007/s12017-021-08659-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/09/2021] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM), the most common primary malignant brain tumor, remains difficult to treat and shares phenotypes, including an aberrant immune response, with other neurological disorders. Understanding the cellular and molecular mechanisms underlying this pathological immune response remains a priority, particularly as standard of care for advanced cancers evolves to include immunotherapies, which have yet to show strong clinical efficacy in GBM. Epidemiological evidence supports a sex difference in GBM, with increased prevalence in males, and recent studies identified differences between males and females ranging from genetic aberrations to cellular programs. Sex differences have also been identified in immune response, and in this mini-review, we present these differences to highlight potential sex-specific cellular and molecular mechanisms that underly GBM growth and response to immunotherapies. These sex differences offer an opportunity to understand GBM pathogenesis and extend beyond GBM to other tumors and neurological disorders to inform the development of next-generation therapies.
Collapse
Affiliation(s)
- Juyeun Lee
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, USA
| | - Kristen Kay
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, USA
| | - Katie Troike
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, USA
| | - Manmeet S Ahluwalia
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, USA.,Case Comprehensive Cancer Center, Cleveland, USA
| | - Justin D Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, USA. .,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, USA. .,Case Comprehensive Cancer Center, Cleveland, USA.
| |
Collapse
|
618
|
Tzeng A, Sangwan N, Jia M, Liu CC, Keslar KS, Downs-Kelly E, Fairchild RL, Al-Hilli Z, Grobmyer SR, Eng C. Human breast microbiome correlates with prognostic features and immunological signatures in breast cancer. Genome Med 2021; 13:60. [PMID: 33863341 PMCID: PMC8052771 DOI: 10.1186/s13073-021-00874-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Currently, over half of breast cancer cases are unrelated to known risk factors, highlighting the importance of discovering other cancer-promoting factors. Since crosstalk between gut microbes and host immunity contributes to many diseases, we hypothesized that similar interactions could occur between the recently described breast microbiome and local immune responses to influence breast cancer pathogenesis. METHODS Using 16S rRNA gene sequencing, we characterized the microbiome of human breast tissue in a total of 221 patients with breast cancer, 18 individuals predisposed to breast cancer, and 69 controls. We performed bioinformatic analyses using a DADA2-based pipeline and applied linear models with White's t or Kruskal-Wallis H-tests with Benjamini-Hochberg multiple testing correction to identify taxonomic groups associated with prognostic clinicopathologic features. We then used network analysis based on Spearman coefficients to correlate specific bacterial taxa with immunological data from NanoString gene expression and 65-plex cytokine assays. RESULTS Multiple bacterial genera exhibited significant differences in relative abundance when stratifying by breast tissue type (tumor, tumor adjacent normal, high-risk, healthy control), cancer stage, grade, histologic subtype, receptor status, lymphovascular invasion, or node-positive status, even after adjusting for confounding variables. Microbiome-immune networks within the breast tended to be bacteria-centric, with sparse structure in tumors and more interconnected structure in benign tissues. Notably, Anaerococcus, Caulobacter, and Streptococcus, which were major bacterial hubs in benign tissue networks, were absent from cancer-associated tissue networks. In addition, Propionibacterium and Staphylococcus, which were depleted in tumors, showed negative associations with oncogenic immune features; Streptococcus and Propionibacterium also correlated positively with T-cell activation-related genes. CONCLUSIONS This study, the largest to date comparing healthy versus cancer-associated breast microbiomes using fresh-frozen surgical specimens and immune correlates, provides insight into microbial profiles that correspond with prognostic clinicopathologic features in breast cancer. It additionally presents evidence for local microbial-immune interplay in breast cancer that merits further investigation and has preventative, diagnostic, and therapeutic potential.
Collapse
Affiliation(s)
- Alice Tzeng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Naseer Sangwan
- Microbiome Composition and Analytics Core, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Margaret Jia
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Chin-Chih Liu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Karen S Keslar
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Erinn Downs-Kelly
- Department of Anatomic Pathology, Cleveland Clinic, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland, OH, 44195, USA
| | - Robert L Fairchild
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Zahraa Al-Hilli
- Department of General Surgery, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Stephen R Grobmyer
- Cleveland Clinic Abu Dhabi, Oncology Institute, Abu Dhabi, United Arab Emirates
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA.
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, 44195, USA.
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Germline High Risk Focus Group, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
619
|
Luo Y, Zhou T. Connecting the dots: Targeting the microbiome in drug toxicity. Med Res Rev 2021; 42:83-111. [PMID: 33856076 DOI: 10.1002/med.21805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/22/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022]
Abstract
The gut microbiota has a vast influence on human health and its role in initiating, aggravating, or ameliorating diseases is beginning to emerge. Recently, its contribution to heterogeneous toxicological responses is also gaining attention, especially in drug-induced toxicity. Whether they are orally administered or not, drugs may interact with the gut microbiota directly or indirectly, which leads to altered toxicity. Present studies focus more on the unidirectional influence of how xenobiotics disturb intestinal microbial composition and functions, and thus induce altered homeostasis. However, interactions between the gut microbiota and xenobiotics are bidirectional and the impact of the gut microbiota on xenobiotics, especially on drugs, should not be neglected. Thus, in this review, we focus on how the gut microbiota modulates drug toxicity by highlighting the microbiome, microbial enzyme, and microbial metabolites. We connect the dots between drugs, the microbiome, microbial enzymes or metabolites, drug metabolites, and host toxicological responses to facilitate the discovery of microbial targets and mechanisms associated with drug toxicity. Besides this, current mainstream strategies to manipulate drug toxicity by targeting the microbiome are summarized and discussed. The review provides technical reference for the evaluation of medicinal properties in the research and development of innovative drugs, and for the future exploitation of strategies to reduce drug toxicity by targeting the microbiome.
Collapse
Affiliation(s)
- Yusha Luo
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China.,Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China.,Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
620
|
Piccioni A, Franza L, Brigida M, Zanza C, Torelli E, Petrucci M, Nicolò R, Covino M, Candelli M, Saviano A, Ojetti V, Franceschi F. Gut Microbiota and Acute Diverticulitis: Role of Probiotics in Management of This Delicate Pathophysiological Balance. J Pers Med 2021; 11:jpm11040298. [PMID: 33919818 PMCID: PMC8070761 DOI: 10.3390/jpm11040298] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/05/2021] [Accepted: 04/11/2021] [Indexed: 02/05/2023] Open
Abstract
How can the knowledge of probiotics and their mechanisms of action be translated into clinical practice when treating patients with diverticular disease and acute diverticulitis? Changes in microbiota composition have been observed in patients who were developing acute diverticulitis, with a reduction of taxa with anti-inflammatory activity, such as Clostridium cluster IV, Lactobacilli and Bacteroides. Recent observations supported that a dysbiosis characterised by decreased presence of anti-inflammatory bacterial species might be linked to mucosal inflammation, and a vicious cycle results from a mucosal inflammation driving dysbiosis at the same time. An alteration in gut microbiota can lead to an altered activation of nerve fibres, and subsequent neuronal and muscular dysfunction, thus favoring abdominal symptoms' development. The possible role of dysbiosis and mucosal inflammation in leading to dysmotility is linked, in turn, to bacterial translocation from the lumen of the diverticulum to perivisceral area. There, a possible activation of Toll-like receptors has been described, with a subsequent inflammatory reaction at the level of the perivisceral tissues. Being aware that bacterial colonisation of diverticula is involved in the pathogenesis of acute diverticulitis, the rationale for the potential role of probiotics in the treatment of this disease becomes clearer. For this review, articles were identified using the electronic PubMed database through a comprehensive search conducted by combining key terms such as "gut microbiota", "probiotics and gut disease", "probiotics and acute diverticulitis", "probiotics and diverticular disease", "probiotics mechanism of action". However, the amount of data present on this matter is not sufficient to draw robust conclusions on the efficacy of probiotics for symptoms' management in diverticular disease.
Collapse
Affiliation(s)
- Andrea Piccioni
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
- Correspondence:
| | - Laura Franza
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Mattia Brigida
- Unit of Gastroenterology, Department of Systems Medicine, Tor Vergata University, 2-00133 Rome, Italy;
| | - Christian Zanza
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Enrico Torelli
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Martina Petrucci
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Rebecca Nicolò
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Marcello Covino
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
| | - Marcello Candelli
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
| | - Angela Saviano
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Veronica Ojetti
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Francesco Franceschi
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| |
Collapse
|
621
|
Li W, Deng X, Chen T. Exploring the Modulatory Effects of Gut Microbiota in Anti-Cancer Therapy. Front Oncol 2021; 11:644454. [PMID: 33928033 PMCID: PMC8076595 DOI: 10.3389/fonc.2021.644454] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
In the recent decade, gut microbiota has received growing interest due to its role in human health and disease. On the one hand, by utilizing the signaling pathways of the host and interacting with the immune system, the gut microbiota is able to maintain the homeostasis in human body. This important role is mainly modulated by the composition of microbiota, as a normal microbiota composition is responsible for maintaining the homeostasis of human body, while an altered microbiota profile could contribute to several pathogenic conditions and may further lead to oncogenesis and tumor progression. Moreover, recent insights have especially focused on the important role of gut microbiota in current anticancer therapies, including chemotherapy, radiotherapy, immunotherapy and surgery. Research findings have indicated a bidirectional interplay between gut microbiota and these therapeutic methods, in which the implementation of different therapeutic methods could lead to different alterations in gut microbiota, and the presence of gut microbiota could in turn contribute to different therapeutic responses. As a result, manipulating the gut microbiota to reduce the therapy-induced toxicity may provide an adjuvant therapy to achieve a better therapeutic outcome. Given the complex role of gut microbiota in cancer treatment, this review summarizes the interactions between gut microbiota and anticancer therapies, and demonstrates the current strategies for reshaping gut microbiota community, aiming to provide possibilities for finding an alternative approach to lower the damage and improve the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Wenyu Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Xiaorong Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tingtao Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, The First Affiliated Hospital, Nanchang University, Nanchang, China
| |
Collapse
|
622
|
Boeri L, Perottoni S, Izzo L, Giordano C, Albani D. Microbiota-Host Immunity Communication in Neurodegenerative Disorders: Bioengineering Challenges for In Vitro Modeling. Adv Healthc Mater 2021; 10:e2002043. [PMID: 33661580 PMCID: PMC11468246 DOI: 10.1002/adhm.202002043] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Human microbiota communicates with its host by secreting signaling metabolites, enzymes, or structural components. Its homeostasis strongly influences the modulation of human tissue barriers and immune system. Dysbiosis-induced peripheral immunity response can propagate bacterial and pro-inflammatory signals to the whole body, including the brain. This immune-mediated communication may contribute to several neurodegenerative disorders, as Alzheimer's disease. In fact, neurodegeneration is associated with dysbiosis and neuroinflammation. The interplay between the microbial communities and the brain is complex and bidirectional, and a great deal of interest is emerging to define the exact mechanisms. This review focuses on microbiota-immunity-central nervous system (CNS) communication and shows how gut and oral microbiota populations trigger immune cells, propagating inflammation from the periphery to the cerebral parenchyma, thus contributing to the onset and progression of neurodegeneration. Moreover, an overview of the technological challenges with in vitro modeling of the microbiota-immunity-CNS axis, offering interesting technological hints about the most advanced solutions and current technologies is provided.
Collapse
Affiliation(s)
- Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Diego Albani
- Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSvia Mario Negri 2Milan20156Italy
| |
Collapse
|
623
|
Baruch EN, Wang J, Wargo JA. Gut Microbiota and Antitumor Immunity: Potential Mechanisms for Clinical Effect. Cancer Immunol Res 2021; 9:365-370. [PMID: 34003768 DOI: 10.1158/2326-6066.cir-20-0877] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Several landmark preclinical studies have shown an association between the gut microbiota and the effectiveness of immunotherapy for cancer. These studies have sparked clinical trials aimed at modulating the gut microbiota in order to improve clinical response rates to immunotherapy. Despite this, the mechanisms through which the gut microbiota influences the effectiveness of immunotherapy are still incompletely characterized. Preclinical and preliminary clinical findings from numerous types of gut microbiota modulation studies, including fecal transplantation, probiotics, consortia, and diet, demonstrate that favorable microbiota modulation is associated with increased intratumoral infiltration of CD8+ effector T cells. This CD8+ T-cell infiltration is often associated with enhanced intratumoral activity of T-helper type 1 cells and dendritic cells and a lower density of immunosuppressive cells. Herein, we discuss how gut microbiota may affect the activity of immune cells by at least three interlacing mechanisms: activation of pattern recognition receptors, molecular mimicry, and impact of metabolites. We also discuss the therapeutic potential and limitations of the different gut microbiota modulation techniques and their putative mechanisms of immune activation.
Collapse
Affiliation(s)
- Erez N Baruch
- Department of Internal Medicine, The University of Texas Health Science Center, Houston, Texas. .,Program for Innovative Microbiome and Translational Research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jingjing Wang
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jennifer A Wargo
- Program for Innovative Microbiome and Translational Research, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
624
|
Cortellini A, Di Maio M, Nigro O, Leonetti A, Cortinovis DL, Aerts JG, Guaitoli G, Barbieri F, Giusti R, Ferrara MG, Bria E, D'Argento E, Grossi F, Rijavec E, Guida A, Berardi R, Torniai M, Sforza V, Genova C, Mazzoni F, Garassino MC, De Toma A, Signorelli D, Gelibter A, Siringo M, Marchetti P, Macerelli M, Rastelli F, Chiari R, Rocco D, Della Gravara L, Inno A, Michele DT, Grassadonia A, Di Marino P, Mansueto G, Zoratto F, Filetti M, Santini D, Citarella F, Russano M, Cantini L, Tuzi A, Bordi P, Minuti G, Landi L, Ricciardi S, Migliorino MR, Passiglia F, Bironzo P, Metro G, Adamo V, Russo A, Spinelli GP, Banna GL, Friedlaender A, Addeo A, Cannita K, Ficorella C, Porzio G, Pinato DJ. Differential influence of antibiotic therapy and other medications on oncological outcomes of patients with non-small cell lung cancer treated with first-line pembrolizumab versus cytotoxic chemotherapy. J Immunother Cancer 2021; 9:e002421. [PMID: 33827906 PMCID: PMC8031700 DOI: 10.1136/jitc-2021-002421] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Some concomitant medications including antibiotics (ATB) have been reproducibly associated with worse survival following immune checkpoint inhibitors (ICIs) in unselected patients with non-small cell lung cancer (NSCLC) (according to programmed death-ligand 1 (PD-L1) expression and treatment line). Whether such relationship is causative or associative is matter of debate. METHODS We present the outcomes analysis according to concomitant baseline medications (prior to ICI initiation) with putative immune-modulatory effects in a large cohort of patients with metastatic NSCLC with a PD-L1 expression ≥50%, receiving first-line pembrolizumab monotherapy. We also evaluated a control cohort of patients with metastatic NSCLC treated with first-line chemotherapy. The interaction between key medications and therapeutic modality (pembrolizumab vs chemotherapy) was validated in pooled multivariable analyses. RESULTS 950 and 595 patients were included in the pembrolizumab and chemotherapy cohorts, respectively. Corticosteroid and proton pump inhibitor (PPI) therapy but not ATB therapy was associated with poorer performance status at baseline in both the cohorts. No association with clinical outcomes was found according to baseline statin, aspirin, β-blocker and metformin within the pembrolizumab cohort. On the multivariable analysis, ATB emerged as a strong predictor of worse overall survival (OS) (HR=1.42 (95% CI 1.13 to 1.79); p=0.0024), and progression free survival (PFS) (HR=1.29 (95% CI 1.04 to 1.59); p=0.0192) in the pembrolizumab but not in the chemotherapy cohort. Corticosteroids were associated with shorter PFS (HR=1.69 (95% CI 1.42 to 2.03); p<0.0001), and OS (HR=1.93 (95% CI 1.59 to 2.35); p<0.0001) following pembrolizumab, and shorter PFS (HR=1.30 (95% CI 1.08 to 1.56), p=0.0046) and OS (HR=1.58 (95% CI 1.29 to 1.94), p<0.0001), following chemotherapy. PPIs were associated with worse OS (HR=1.49 (95% CI 1.26 to 1.77); p<0.0001) with pembrolizumab and shorter OS (HR=1.12 (95% CI 1.02 to 1.24), p=0.0139), with chemotherapy. At the pooled analysis, there was a statistically significant interaction with treatment (pembrolizumab vs chemotherapy) for corticosteroids (p=0.0020) and PPIs (p=0.0460) with respect to OS, for corticosteroids (p<0.0001), ATB (p=0.0290), and PPIs (p=0.0487) with respect to PFS, and only corticosteroids (p=0.0033) with respect to objective response rate. CONCLUSION In this study, we validate the significant negative impact of ATB on pembrolizumab monotherapy but not chemotherapy outcomes in NSCLC, producing further evidence about their underlying immune-modulatory effect. Even though the magnitude of the impact of corticosteroids and PPIs is significantly different across the cohorts, their effects might be driven by adverse disease features.
Collapse
Affiliation(s)
- Alessio Cortellini
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
- Department of Biotechnology and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | - Massimo Di Maio
- Department of Oncology and Medical Oncology, University of Turin and AO Ordine Mauriziano, Turin, Italy
| | - Olga Nigro
- Medical Oncology, ASST dei Sette Laghi, Varese, Italy
| | | | | | - Joachim Gjv Aerts
- Department of Pulmonary Disease, Erasmus Medical Center, Rotterdam, Netherlands
| | - Giorgia Guaitoli
- Dipartimento di Oncologia ed Ematologia, AOU Policlinico di Modena, Modena, Italy
| | - Fausto Barbieri
- Dipartimento di Oncologia ed Ematologia, AOU Policlinico di Modena, Modena, Italy
| | - Raffaele Giusti
- Medical Oncology Unit, Sant'Andrea Hospital of Rome, Roma, Italy
| | - Miriam G Ferrara
- Comprehensive Cancer Center, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Roma, Italy
- Department of Translational Medicine and Surgery, Universitá Cattolica del Sacro Cuore, Roma, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Roma, Italy
- Department of Translational Medicine and Surgery, Universitá Cattolica del Sacro Cuore, Roma, Italy
| | - Ettore D'Argento
- Comprehensive Cancer Center, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Roma, Italy
| | - Francesco Grossi
- Division of Medical Oncology, University of Insubria, Varese, Italy
| | - Erika Rijavec
- Medical Oncology, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Annalisa Guida
- Struttura Complessa di Oncologia Medica e Traslazionale, Azienda Ospedaliera Santa Maria di Terni, Terni, Italy
| | - Rossana Berardi
- Oncology Clinic, Università Politecnica delle Marche, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Mariangela Torniai
- Oncology Clinic, Università Politecnica delle Marche, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Vincenzo Sforza
- Thoracic Medical Oncology, National Cancer Institute IRCCS Pascale Foundation, Napoli, Italy
| | - Carlo Genova
- Lung Cancer Unit, IRCCS Ospedal Policlinico San Martino, Genova, Italy
| | - Francesca Mazzoni
- Department of Medical Oncology, Careggi University Hospital, Firenze, Toscana, Italy
| | | | - Alessandro De Toma
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Diego Signorelli
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alain Gelibter
- Medical Oncology Unit B, Policlinico Umberto I, Sapienza University of Rome, Roma, Italy
| | - Marco Siringo
- Medical Oncology Unit B, Policlinico Umberto I, Sapienza University of Rome, Roma, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Roma, Italy
| | - Marianna Macerelli
- Medical Oncology, University Hospital Santa Maria della Misericordia, Udine, Italy
| | | | - Rita Chiari
- Medical Oncology, Ospedali riuniti Padova Sud "Madre Teresa Di Calcutta", Monselice, Padova, Italy
| | - Danilo Rocco
- Pneumo-Oncology Unit, Ospedali dei Colli Monaldi Cotugno CTO, Napoli, Italy
| | | | - Alessandro Inno
- Oncology Unit, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar, Italy
| | - De Tursi Michele
- Dipartimento di Terapie Innovative in Medicina ed Odontoiatria, Universitá G. D'Annunzio, Chieti-Pescara, Italy
| | - Antonino Grassadonia
- Dipartimento di Terapie Innovative in Medicina ed Odontoiatria, Universitá G. D'Annunzio, Chieti-Pescara, Italy
| | | | - Giovanni Mansueto
- Medical Oncology, Azienda Sanitaria Locale Frosinone, Frosinone, Italy
| | | | - Marco Filetti
- Medical Oncology Unit, Sant'Andrea Hospital of Rome, Roma, Italy
| | | | | | - Marco Russano
- Medical Oncology, Campus Bio-Medico University, Roma, Italy
| | - Luca Cantini
- Department of Pulmonary Disease, Erasmus Medical Center, Rotterdam, Netherlands
- Oncology Clinic, Università Politecnica delle Marche, Ospedali Riuniti di Ancona, Ancona, Italy
| | | | - Paola Bordi
- Medical Oncology, University Hospital of Parma, Parma, Italy
| | - Gabriele Minuti
- Department of Oncology and Hematology, AUSL della Romagna, Ravenna, Italy
| | - Lorenza Landi
- Department of Oncology and Hematology, AUSL della Romagna, Ravenna, Italy
| | - Serena Ricciardi
- Pneumo-Oncology Unit, San Camillo Forlanini Hospital, Roma, Italy
| | | | - Francesco Passiglia
- Department of Oncology, San Luigi Gonzaga University Hospital, Orbassano, Italy
| | - Paolo Bironzo
- Department of Oncology, San Luigi Hospital, Orbassano, Italy
| | - Giulio Metro
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Vincenzo Adamo
- Medical Oncology and Department of Human Pathology, Azienda Ospedaliera Papardo and Università degli Studi di Messina, Messina, Italy
| | - Alessandro Russo
- Medical Oncology and Department of Human Pathology, Azienda Ospedaliera Papardo and Università degli Studi di Messina, Messina, Italy
| | - Gian Paolo Spinelli
- UOC Territorial Oncology of Aprilia, AUSL Latina, Sapienza University of Rome, Aprilia, Italy
| | - Giuseppe L Banna
- Medical Oncology, Portsmouth University Hospitals NHS Trust, Portsmouth, UK
| | - Alex Friedlaender
- Oncology Department, University Hospital of Geneva, Geneve, Switzerland
| | - Alfredo Addeo
- Oncology Department, University Hospital of Geneva, Geneve, Switzerland
| | - Katia Cannita
- Medical Oncology, San Salvatore Hospital, L'Aquila, Italy
| | - Corrado Ficorella
- Department of Biotechnology and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
- Medical Oncology, San Salvatore Hospital, L'Aquila, Italy
| | | | - David J Pinato
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
- Department of Translational Medicine, Universitá del Piemonte Orientale "A. Avogadro", Novara, Italy
| |
Collapse
|
625
|
González‐Sánchez P, DeNicola GM. The microbiome(s) and cancer: know thy neighbor(s). J Pathol 2021; 254:332-343. [DOI: 10.1002/path.5661] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Paloma González‐Sánchez
- Department of Cancer Physiology H. Lee Moffitt Cancer Center and Research Institute Tampa FL USA
| | - Gina M DeNicola
- Department of Cancer Physiology H. Lee Moffitt Cancer Center and Research Institute Tampa FL USA
| |
Collapse
|
626
|
Abstract
Microbial roles in cancer formation, diagnosis, prognosis, and treatment have been disputed for centuries. Recent studies have provocatively claimed that bacteria, viruses, and/or fungi are pervasive among cancers, key actors in cancer immunotherapy, and engineerable to treat metastases. Despite these findings, the number of microbes known to directly cause carcinogenesis remains small. Critically evaluating and building frameworks for such evidence in light of modern cancer biology is an important task. In this Review, we delineate between causal and complicit roles of microbes in cancer and trace common themes of their influence through the host's immune system, herein defined as the immuno-oncology-microbiome axis. We further review evidence for intratumoral microbes and approaches that manipulate the host's gut or tumor microbiome while projecting the next phase of experimental discovery.
Collapse
Affiliation(s)
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1015, Villejuif, France
- Université Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jeff Hasty
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- BioCircuits Institute, University of California, San Diego, La Jolla, CA, USA
- Molecular Biology Section, Division of Biological Science, University of California, San Diego, La Jolla, CA, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rob Knight
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
627
|
Extracellular Vesicles and Their Role in the Spatial and Temporal Expansion of Tumor-Immune Interactions. Int J Mol Sci 2021; 22:ijms22073374. [PMID: 33806053 PMCID: PMC8036938 DOI: 10.3390/ijms22073374] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) serve as trafficking vehicles and intercellular communication tools. Their cargo molecules directly reflect characteristics of their parental cell. This includes information on cell identity and specific cellular conditions, ranging from normal to pathological states. In cancer, the content of EVs derived from tumor cells is altered and can induce oncogenic reprogramming of target cells. As a result, tumor-derived EVs compromise antitumor immunity and promote cancer progression and spreading. However, this pro-oncogenic phenotype is constantly being challenged by EVs derived from the local tumor microenvironment and from remote sources. Here, we summarize the role of EVs in the tumor–immune cross-talk that includes, but is not limited to, immune cells in the tumor microenvironment. We discuss the potential of remotely released EVs from the microbiome and during physical activity to shape the tumor–immune cross-talk, directly or indirectly, and confer antitumor activity. We further discuss the role of proinflammatory EVs in the temporal development of the tumor–immune interactions and their potential use for cancer diagnostics.
Collapse
|
628
|
Lione L, Salvatori E, Petrazzuolo A, Massacci A, Maggio R, Confroti A, Compagnone M, Aurisicchio L, Ciliberto G, Palombo F. Antitumor efficacy of a neoantigen cancer vaccine delivered by electroporation is influenced by microbiota composition. Oncoimmunology 2021; 10:1898832. [PMID: 33796408 PMCID: PMC7993125 DOI: 10.1080/2162402x.2021.1898832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/05/2021] [Accepted: 02/24/2021] [Indexed: 01/22/2023] Open
Abstract
Cancer is a heterogeneous disease and its treatment remains unsatisfactory with inconstant therapeutic responses. This variability could be related, at least in part, to different and highly personalized gut microbiota compositions. Different studies have shown an impact of microbiota on antitumor therapy. It has been demonstrated that some gut bacteria influences the development and differentiation of immune cells, suggesting that different microbiota compositions could affect the efficacy of the antitumor vaccine. Emerging data suggest that recognition of neoantigens for the generation of neoantigen cancer vaccines (NCVs) could have a key role in the activity of clinical immunotherapies. However, it is still unknown whether there is a crosstalk between microbiota and NCV. This study aimed to understand the possible mechanisms of interaction between gut microbiota and NCV delivered by DNA-electroporation (DNA-EP). We found that decreased microbiota diversity induced by prolonged antibiotic (ATB) treatment is associated with higher intratumor specific immune responses and consequently to a better antitumor effect induced by NCV delivered by DNA-EP.
Collapse
Affiliation(s)
- Lucia Lione
- Cancer Immunology, Takis, Rome, Italy
- Università Magna Grecia, Catanzaro, Italy
| | | | - Adriana Petrazzuolo
- Cancer Immunology, Takis, Rome, Italy
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université De Paris, Sorbonne Université, INSERM U1138, Centre De Recherche Des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Biogem, Ariano Irpino, Italy
| | | | | | | | - Mirco Compagnone
- Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, France
| | - Luigi Aurisicchio
- Cancer Immunology, Takis, Rome, Italy
- Evvivax, Rome, Italy
- Biogem, Ariano Irpino, Italy
- Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, France
| | - Gennaro Ciliberto
- Cancer immunology, Neomatrix, Rome, Italy
- Scientific Directorate, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabio Palombo
- Cancer Immunology, Takis, Rome, Italy
- Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, France
| |
Collapse
|
629
|
An intact gut microbiome protects genetically predisposed mice against leukemia. Blood 2021; 136:2003-2017. [PMID: 32911536 DOI: 10.1182/blood.2019004381] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 08/21/2020] [Indexed: 12/22/2022] Open
Abstract
The majority of childhood leukemias are precursor B-cell acute lymphoblastic leukemias (pB-ALLs) caused by a combination of prenatal genetic predispositions and oncogenic events occurring after birth. Although genetic predispositions are frequent in children (>1% to 5%), fewer than 1% of genetically predisposed carriers will develop pB-ALL. Although infectious stimuli are believed to play a major role in leukemogenesis, the critical determinants are not well defined. Here, by using murine models of pB-ALL, we show that microbiome disturbances incurred by antibiotic treatment early in life were sufficient to induce leukemia in genetically predisposed mice, even in the absence of infectious stimuli and independent of T cells. By using V4 and full-length 16S ribosomal RNA sequencing of a series of fecal samples, we found that genetic predisposition to pB-ALL (Pax5 heterozygosity or ETV6-RUNX1 fusion) shaped a distinct gut microbiome. Machine learning accurately (96.8%) predicted genetic predisposition using 40 of 3983 amplicon sequence variants as proxies for bacterial species. Transplantation of either wild-type (WT) or Pax5+/- hematopoietic bone marrow cells into WT recipient mice revealed that the microbiome is shaped and determined in a donor genotype-specific manner. Gas chromatography-mass spectrometry (GC-MS) analyses of sera from WT and Pax5+/- mice demonstrated the presence of a genotype-specific distinct metabolomic profile. Taken together, our data indicate that it is a lack of commensal microbiota rather than the presence of specific bacteria that promotes leukemia in genetically predisposed mice. Future large-scale longitudinal studies are required to determine whether targeted microbiome modification in children predisposed to pB-ALL could become a successful prevention strategy.
Collapse
|
630
|
Thinking Small: Small Molecules as Potential Synergistic Adjuncts to Checkpoint Inhibition in Melanoma. Int J Mol Sci 2021; 22:ijms22063228. [PMID: 33810078 PMCID: PMC8005112 DOI: 10.3390/ijms22063228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Metastatic melanoma remains the deadliest form of skin cancer. Immune checkpoint inhibition (ICI) immunotherapy has defined a new age in melanoma treatment, but responses remain inconsistent and some patients develop treatment resistance. The myriad of newly developed small molecular (SM) inhibitors of specific effector targets now affords a plethora of opportunities to increase therapeutic responses, even in resistant melanoma. In this review, we will discuss the multitude of SM classes currently under investigation, current and prospective clinical combinations of ICI and SM therapies, and their potential for synergism in melanoma eradication based on established mechanisms of immunotherapy resistance.
Collapse
|
631
|
Huang C, Li M, Liu B, Zhu H, Dai Q, Fan X, Mehta K, Huang C, Neupane P, Wang F, Sun W, Umar S, Zhong C, Zhang J. Relating Gut Microbiome and Its Modulating Factors to Immunotherapy in Solid Tumors: A Systematic Review. Front Oncol 2021; 11:642110. [PMID: 33816289 PMCID: PMC8012896 DOI: 10.3389/fonc.2021.642110] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Gut microbiome is proved to affect the activity of immunotherapy in certain tumors. However, little is known if there is universal impact on both the treatment response and adverse effects (AEs) of immune checkpoint inhibitors (ICIs) across multiple solid tumors, and whether such impact can be modulated by common gut microbiome modifiers, such as antibiotics and diet. Methods: A systematic search in PubMed followed by stringent manual review were performed to identify clinical cohort studies that evaluated the relevance of gut microbiome to ICIs (response and/or AEs, 12 studies), or association of antibiotics with ICIs (17 studies), or impact of diet on gut microbiome (16 studies). Only original studies published in English before April 1st, 2020 were used. Qualified studies identified in the reference were also included. Results: At the phylum level, patients who had enriched abundance in Firmicutes and Verrucomicrobia almost universally had better response from ICIs, whereas those who were enriched in Proteobacteria universally presented with unfavorable outcome. Mixed correlations were observed for Bacteroidetes in relating to treatment response. Regarding the AEs, Firmicutes correlated to higher incidence whereas Bacteroidetes were clearly associated with less occurrence. Interestingly, across various solid tumors, majority of the studies suggested a negative association of antibiotic use with clinical response from ICIs, especially within 1-2 month prior to the initiation of ICIs. Finally, we observed a significant correlation of plant-based diet in relating to the enrichment of “ICI-favoring” gut microbiome (P = 0.0476). Conclusions: Gut microbiome may serve as a novel modifiable biomarker for both the treatment response and AEs of ICIs across various solid tumors. Further study is needed to understand the underlying mechanism, minimize the negative impact of antibiotics on ICIs, and gain insight regarding the role of diet so that this important lifestyle factor can be harnessed to improve the therapeutic outcomes of cancer immunotherapy partly through its impact on gut microbiome.
Collapse
Affiliation(s)
- Chengliang Huang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Meizhang Li
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ben Liu
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, United States
| | - Huanbo Zhu
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States.,Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Xianming Fan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kathan Mehta
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Chao Huang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Prakash Neupane
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Fen Wang
- Department of Radiation Oncology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Weijing Sun
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States
| | - Shahid Umar
- Department of Surgery, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States.,Department of Cancer Biology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, United States
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, University of Kansas Medical Center, Westwood, KS, United States.,Department of Cancer Biology, University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
632
|
Zhang L, Shi Y, Do KA, Peterson CB, Jenq RR. ProgPerm: Progressive permutation for a dynamic representation of the robustness of microbiome discoveries. BMC Bioinformatics 2021; 22:126. [PMID: 33731016 PMCID: PMC7972227 DOI: 10.1186/s12859-021-04061-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Identification of features is a critical task in microbiome studies that is complicated by the fact that microbial data are high dimensional and heterogeneous. Masked by the complexity of the data, the problem of separating signals (differential features between groups) from noise (features that are not differential between groups) becomes challenging and troublesome. For instance, when performing differential abundance tests, multiple testing adjustments tend to be overconservative, as the probability of a type I error (false positive) increases dramatically with the large numbers of hypotheses. Moreover, the grouping effect of interest can be obscured by heterogeneity. These factors can incorrectly lead to the conclusion that there are no differences in the microbiome compositions. RESULTS We translate and represent the problem of identifying differential features, which are differential in two-group comparisons (e.g., treatment versus control), as a dynamic layout of separating the signal from its random background. More specifically, we progressively permute the grouping factor labels of the microbiome samples and perform multiple differential abundance tests in each scenario. We then compare the signal strength of the most differential features from the original data with their performance in permutations, and will observe a visually apparent decreasing trend if these features are true positives identified from the data. Simulations and applications on real data show that the proposed method creates a U-curve when plotting the number of significant features versus the proportion of mixing. The shape of the U-Curve can convey the strength of the overall association between the microbiome and the grouping factor. We also define a fragility index to measure the robustness of the discoveries. Finally, we recommend the identified features by comparing p-values in the observed data with p-values in the fully mixed data. CONCLUSIONS We have developed this into a user-friendly and efficient R-shiny tool with visualizations. By default, we use the Wilcoxon rank sum test to compute the p-values, since it is a robust nonparametric test. Our proposed method can also utilize p-values obtained from other testing methods, such as DESeq. This demonstrates the potential of the progressive permutation method to be extended to new settings.
Collapse
Affiliation(s)
- Liangliang Zhang
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Yushu Shi
- Department of Statistics, University of Missouri, Columbia, MO USA
| | - Kim-Anh Do
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Christine B. Peterson
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Robert R. Jenq
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
633
|
Targeting the Gut Microbiome to Mitigate Immunotherapy-Induced Colitis in Cancer. Trends Cancer 2021; 7:583-593. [PMID: 33741313 DOI: 10.1016/j.trecan.2021.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have been a transformational advance in cancer therapy in the past decade. However, ICIs can produce immune-related adverse effects (irAEs), which can lead to both morbidity and premature termination of therapy. Recent studies suggest that the gut microbiota and its metabolites affect ICI efficacy and toxicity. Herein, we review such evidence in the context of ICI-induced colitis. In particular, the short-chain fatty acid butyrate, a microbial metabolite, has known protective effects on the intestine. We discuss how the use of dietary prebiotics, which can be metabolized by bacteria to produce butyrate, can be an intriguing new investigational approach to prevent ICI-associated colitis and lead to improved patient outcomes.
Collapse
|
634
|
Minnie SA, Hill GR. Autologous Stem Cell Transplantation for Myeloma: Cytoreduction or an Immunotherapy? Front Immunol 2021; 12:651288. [PMID: 33777050 PMCID: PMC7994609 DOI: 10.3389/fimmu.2021.651288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 02/19/2021] [Indexed: 12/22/2022] Open
Abstract
The incidence of multiple myeloma (MM), a bone marrow (BM) resident hematological malignancy, is increasing globally. The disease has substantial morbidity and mortality and remains largely incurable. Clinical studies show that autologous stem cell transplantation (ASCT) remains efficacious in eligible patients, providing a progression free survival (PFS) benefit beyond novel therapies alone. Conventionally, improved PFS after ASCT is attributed to cytoreduction from myeloablative chemotherapy. However, ASCT results in immune effects beyond cytoreduction, including inflammation, lymphodepletion, T cell priming via immunogenic cell death, and disruption of the tumor BM microenvironment. In fact, a small subset of patients achieve very long-term control of disease post-ASCT, akin to that seen in the context of immune-mediated graft-vs.-myeloma effects after allogeneic SCT. These clinical observations coupled with recent definitive studies in mice demonstrating that progression after ASCT represents immune escape as a consequence of T cell exhaustion, highlight the potential for new immunotherapy maintenance strategies to prevent myeloma progression following consolidation with ASCT.
Collapse
Affiliation(s)
- Simone A Minnie
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Division of Medical Oncology, University of Washington, Seattle, WA, United States
| |
Collapse
|
635
|
Pancreatic Cancer Meets Human Microbiota: Close Encounters of the Third Kind. Cancers (Basel) 2021; 13:cancers13061231. [PMID: 33799784 PMCID: PMC7998494 DOI: 10.3390/cancers13061231] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/07/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The microorganisms colonizing the epithelial surfaces of the human body, called microbiota, have been shown to influence the initiation, progression and response to therapy of many solid tumors, including pancreatic ductal adenocarcinoma, the most prominent form of pancreatic cancer. Here, we summarize the current knowledge about the influence of oral, gut and intratumoral microbiota on pancreatic ductal adenocarcinoma development and chemoresistance. Abstract Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal types of cancer with a dismal prognosis. The five-year survival rate has not changed significantly in over 40 years. Current first-line treatments only offer a modest increase in overall survival in unselected populations, and there is an urgent need to personalize treatment in this aggressive disease and develop new therapeutic strategies. Evolving evidence suggests that the human microbiome impacts cancerogenesis and cancer resistance to therapy. The mechanism of action and interaction of microbiome and PDAC is still under investigation. Direct and indirect effects have been proposed, and the use of several microbiome signatures as predictive and prognostic biomarkers for pancreatic cancer are opening new therapeutic horizons. In this review, we provide an overview for the clinicians of studies describing the influence and associations of oral, gastrointestinal and intratumoral microbiota on PDAC development, progression and resistance to therapy and the potential use of microbiota as a diagnostic, prognostic and predictive biomarker for PDAC.
Collapse
|
636
|
Li Y, Elmén L, Segota I, Xian Y, Tinoco R, Feng Y, Fujita Y, Segura Muñoz RR, Schmaltz R, Bradley LM, Ramer-Tait A, Zarecki R, Long T, Peterson SN, Ronai ZA. Prebiotic-Induced Anti-tumor Immunity Attenuates Tumor Growth. Cell Rep 2021; 30:1753-1766.e6. [PMID: 32049008 PMCID: PMC7053418 DOI: 10.1016/j.celrep.2020.01.035] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 10/06/2019] [Accepted: 01/08/2020] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the importance of gut microbiota in the control of tumor growth and response to therapy. Here, we select prebiotics that can enrich bacterial taxa that promote anti-tumor immunity. Addition of the prebiotics inulin or mucin to the diet of C57BL/6 mice induces anti-tumor immune responses and inhibition of BRAF mutant melanoma growth in a subcutaneously implanted syngeneic mouse model. Mucin fails to inhibit tumor growth in germ-free mice, indicating that the gut microbiota is required for the activation of the anti-tumor immune response. Inulin and mucin drive distinct changes in the microbiota, as inulin, but not mucin, limits tumor growth in syngeneic mouse models of colon cancer and NRAS mutant melanoma and enhances the efficacy of a MEK inhibitor against melanoma while delaying the emergence of drug resistance. We highlight the importance of gut microbiota in anti-tumor immunity and the potential therapeutic role for prebiotics in this process.
Collapse
Affiliation(s)
- Yan Li
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Lisa Elmén
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Igor Segota
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yibo Xian
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Roberto Tinoco
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yongmei Feng
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yu Fujita
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Rafael R Segura Muñoz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Robert Schmaltz
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Linda M Bradley
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Amanda Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Raphy Zarecki
- Technion Integrated Cancer Center, Faculty of Medicine, Technion, Haifa 3525433, Israel
| | - Tao Long
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Scott N Peterson
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| | - Ze'ev A Ronai
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
637
|
Rao W, Lin Z, Liu S, Zhang Z, Xie Q, Chen H, Lin X, Chen Y, Yang H, Yu K, Hu Z. Association between alcohol consumption and oesophageal microbiota in oesophageal squamous cell carcinoma. BMC Microbiol 2021; 21:73. [PMID: 33673801 PMCID: PMC7936487 DOI: 10.1186/s12866-021-02137-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Microbiota has been reported to play a role in cancer patients. Nevertheless, little is known about the association between alcohol consumption and resultant changes in the diversity and composition of oesophageal microbiota in oesophageal squamous cell carcinoma (ESCC). METHODS We performed a hospital-based retrospective study of 120 patients with pathologically diagnosed primary ESCC. The relevant information for all study participants were collected through a detailed questionnaire. The differences in adjacent tissues between non-drinkers and drinkers were explored using 16S rRNA gene sequencing. Raw sequencing data were imported into QIIME 2 to analyse the diversity and abundance of microbiota. Linear discriminant analysis effect size (LEfSe) and unconditional logistic regression were performed to determine the bacterial taxa that were associated with drinking. RESULTS The Shannon diversity index and Bray-Curtis distance of oesophageal microbiota were significantly different among drinkers(P < 0.05). The alcohol-related bacteria were primarily from the orders Clostridiales, Gemellales and Pasteurellales, family Clostridiaceae, Lanchnospiraceae, Helicobacteraceae, Alcaligenaceae, Bacteroidaceae, Pasteurellaceae and Gemellaceae; genus Clostridium, Helicobacter, Catonella, Bacteroides, Bacillus, Moraxella, and Bulleidia; and species B. moorei and longum (genus Bifidobacterium). In addition, the diversity and abundance of these microbiota were observed to be affected by the age, residential districts of the patients, and sampling seasons. Moreover, the higher the frequency and years of alcohol consumption, the lower was the relative abundance of genus Catonella that was observed. CONCLUSION Alcohol consumption is associated with alterations in both the diversity and composition the of the oesophageal microbiota in ESCC patients.
Collapse
Affiliation(s)
- Wenqing Rao
- Department of Epidemiology and Health Statistics, Fujian Medical University Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Zheng Lin
- Department of Epidemiology and Health Statistics, Fujian Medical University Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Shuang Liu
- Department of Epidemiology and Health Statistics, Fujian Medical University Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Zhihui Zhang
- Department of Epidemiology and Health Statistics, Fujian Medical University Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Qianwen Xie
- Department of Epidemiology and Health Statistics, Fujian Medical University Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Huilin Chen
- Department of Radiation Oncology, Anxi County Hospital, Quanzhou, 352400, China
| | - Xi Lin
- Department of Statistics Office, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, China
| | - Yuanmei Chen
- Department of Thoracic Surgery, Fujian Provincial Cancer Hospital Affiliation to Fujian Medical University, Fuzhou, 350014, China
| | - Huimin Yang
- Department of Epidemiology and Health Statistics, Fujian Medical University Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Kaili Yu
- Department of Epidemiology and Health Statistics, Fujian Medical University Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, Fujian Medical University Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, China.
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
638
|
dos Santos LV, Abrahão CM, William WN. Overcoming Resistance to Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinomas. Front Oncol 2021; 11:596290. [PMID: 33747915 PMCID: PMC7973277 DOI: 10.3389/fonc.2021.596290] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/08/2021] [Indexed: 12/11/2022] Open
Abstract
Preclinical data suggest that head and neck squamous cell carcinomas (HNSCC) may evade immune surveillance and induce immunosuppression. One mechanism of immune evasion involves the expression of programmed death ligand-1 (PD-L1) in tumor and immune cells, which is, to date, the only biomarker routinely used in clinical practice to select patients with advanced HNSCCs more likely to benefit from anti-PD-1 therapy. Nonetheless, PD-L1 expression alone incompletely captures the degree of sensitivity of HNSCCs to PD-1 inhibitors. Most patients exposed to anti-PD-1 antibodies do not respond to therapy, suggesting the existence of mechanisms of de novo resistance to immunotherapy. Furthermore, patients that initially respond to PD-1 inhibitors will eventually develop acquired resistance to immunotherapy through mechanisms that have not yet been completely elucidated. In this article, we will provide an overview of the immune landscape of HNSCCs. We will briefly describe the clinical activity of inhibitors of the PD-1/PD-L1 axis in this disease, as well as biomarkers of benefit from these agents that have been identified so far. We will review pre-clinical and clinical work in cancers in general, and in HNSCCs specifically, that have characterized the mechanisms of de novo and acquired resistance to immunotherapy. Lastly, we will provide insights into novel strategies under investigation to overcome resistance to immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | - William N. William
- Centro de Oncologia, Hospital BP, A Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| |
Collapse
|
639
|
Diaz-Garrido N, Cordero C, Olivo-Martinez Y, Badia J, Baldomà L. Cell-to-Cell Communication by Host-Released Extracellular Vesicles in the Gut: Implications in Health and Disease. Int J Mol Sci 2021; 22:ijms22042213. [PMID: 33672304 PMCID: PMC7927122 DOI: 10.3390/ijms22042213] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Communication between cells is crucial to preserve body homeostasis and health. Tightly controlled intercellular dialog is particularly relevant in the gut, where cells of the intestinal mucosa are constantly exposed to millions of microbes that have great impact on intestinal homeostasis by controlling barrier and immune functions. Recent knowledge involves extracellular vesicles (EVs) as mediators of such communication by transferring messenger bioactive molecules including proteins, lipids, and miRNAs between cells and tissues. The specific functions of EVs principally depend on the internal cargo, which upon delivery to target cells trigger signal events that modulate cellular functions. The vesicular cargo is greatly influenced by genetic, pathological, and environmental factors. This finding provides the basis for investigating potential clinical applications of EVs as therapeutic targets or diagnostic biomarkers. Here, we review current knowledge on the biogenesis and cargo composition of EVs in general terms. We then focus the attention to EVs released by cells of the intestinal mucosa and their impact on intestinal homeostasis in health and disease. We specifically highlight their role on epithelial barrier integrity, wound healing of epithelial cells, immunity, and microbiota shaping. Microbiota-derived EVs are not reviewed here.
Collapse
Affiliation(s)
- Natalia Diaz-Garrido
- Secció de Bioquímica i Biología Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (N.D.-G.); (C.C.); (Y.O.-M.); (J.B.)
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | - Cecilia Cordero
- Secció de Bioquímica i Biología Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (N.D.-G.); (C.C.); (Y.O.-M.); (J.B.)
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | - Yenifer Olivo-Martinez
- Secció de Bioquímica i Biología Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (N.D.-G.); (C.C.); (Y.O.-M.); (J.B.)
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | - Josefa Badia
- Secció de Bioquímica i Biología Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (N.D.-G.); (C.C.); (Y.O.-M.); (J.B.)
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | - Laura Baldomà
- Secció de Bioquímica i Biología Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (N.D.-G.); (C.C.); (Y.O.-M.); (J.B.)
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-403-44-96
| |
Collapse
|
640
|
Li M, Zhang C. Are silver nanoparticles better than triclosan as a daily antimicrobial? Answers from the perspectives of gut microbiome disruption and pathogenicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 756:143983. [PMID: 33302073 DOI: 10.1016/j.scitotenv.2020.143983] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/15/2020] [Accepted: 11/15/2020] [Indexed: 05/23/2023]
Abstract
As an alternative to triclosan (TCS), the widespread use of silver nanoparticles (AgNPs) in daily products shows genuine potential. However, information regarding whether AgNPs are substantially better than TCS in their potential disruption of the gut microbiome and health effects is lacking. Using a simulator of the human intestinal microbial ecosystem (SHIME), we systemically compared the effects of TCS and AgNPs (at 1 μg/L and 30 μg/L) on the human gut microbiome in terms of changes in gut homeostasis, microbial community structure, antibiotic resistance profiles and abundances of opportunistic pathogens. Generally, TCS exerted more severe effects than AgNPs on gut disturbances (i.e., decreased production of short-chain fatty acids, increased contents of ammonium and total bile acids, and increased β-glucosidase activities) in a dose-dependent manner, whereas no clear dose effect was observed for the AgNP treatment because of potential nanoparticle transformation. The more serious effect of TCS than AgNPs on the microbiota composition was indicated by the dynamic increase in the Firmicutes/Bacteroidetes ratio determined using 16S rDNA sequencing. Metagenomic analyses revealed a more pronounced effect of TCS than AgNPs on the selection and dissemination of multiple resistance genes to antibiotics, TCS, and even Ag via the enrichment of genes encoding efflux pumps and mobile genetic elements. Consequently, the overgrowth of opportunistic pathogens was observed upon TCS exposure due to an imbalanced microbiome, in contrast to a slight increase in the abundance of some beneficial bacteria (i.e., Bifidobacterium) induced by the AgNP treatment. In conclusion, from the perspective of effects on gut health, AgNPs may prevail over TCS to some extent. However, the stress and potential selection of Ag resistance indicates the need for targeted surveillance of AgNP commercialization for daily use.
Collapse
Affiliation(s)
- Mingzhu Li
- School of Environment, Beijing Normal University, Beijing 100875, China; College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Chengdong Zhang
- School of Environment, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
641
|
Ma J, Zhu W, Liu B. Role of gut microbiome in the outcome of cancer immunotherapy. Int J Cancer 2021; 149:760-768. [PMID: 33600603 DOI: 10.1002/ijc.33524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/20/2020] [Accepted: 01/27/2021] [Indexed: 12/11/2022]
Abstract
Nearly 3 × 1013 types of bacteria colonize the human intestine. These colonized bacteria help in maintaining intestinal homeostasis by establishing a complex relationship with the intestinal epithelium and lymphoid tissue. Alteration in the composition of the intestinal microbiota is associated with susceptibility to various pathological conditions, such as autoimmune disorders, diabetes, inflammation and cancer. Of late, several researchers have focused on examining the effects of gut microbiota on the outcome of various cancer treatment protocols. Side effects and complications of traditional chemotherapy and allogeneic hematopoietic cell transplantation are associated with intestinal dysbiosis. Gut microbiota affects the efficacy of immune checkpoint inhibitor-based immunotherapy. The gut is inhabited by diverse resident bacteria, of which, few enhance, while others inhibit the host response to immunotherapy. This review focuses on the correlation between intestinal microbiota and the outcome of tumor immunotherapy. Additionally, the molecular mechanisms underlying the effects of gut microbiota on the efficacy of cancer immunotherapy have been reviewed. Further studies are needed for the identification of distinct gut microbiota and their efficacy in tumor immunotherapy as certain types of intestinal bacteria could function as novel adjuvant drugs to enhance the effectiveness of antitumor therapy in humans.
Collapse
Affiliation(s)
- Junting Ma
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, China
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Wenwen Zhu
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Beixing Liu
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
642
|
Ghosh S, Whitley CS, Haribabu B, Jala VR. Regulation of Intestinal Barrier Function by Microbial Metabolites. Cell Mol Gastroenterol Hepatol 2021; 11:1463-1482. [PMID: 33610769 PMCID: PMC8025057 DOI: 10.1016/j.jcmgh.2021.02.007] [Citation(s) in RCA: 349] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/11/2022]
Abstract
The human gastrointestinal tract (GI) harbors a diverse population of microbial life that continually shapes host pathophysiological responses. Despite readily available abundant metagenomic data, the functional dynamics of gut microbiota remain to be explored in various health and disease conditions. Microbiota generate a variety of metabolites from dietary products that influence host health and pathophysiological functions. Since gut microbial metabolites are produced in close proximity to gut epithelium, presumably they have significant impact on gut barrier function and immune responses. The goal of this review is to discuss recent advances on gut microbial metabolites in the regulation of intestinal barrier function. While the mechanisms of action of these metabolites are only beginning to emerge, they mainly point to a small group of shared pathways that control gut barrier functions. Amidst expanding technology and broadening knowledge, exploitation of beneficial microbiota and their metabolites to restore pathophysiological balance will likely prove to be an extremely useful remedial tool.
Collapse
Affiliation(s)
- Sweta Ghosh
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Caleb Samuel Whitley
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Venkatakrishna Rao Jala
- Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
643
|
Characterization of microbiota in acute leukemia patients following successful remission induction chemotherapy without antimicrobial prophylaxis. Int Microbiol 2021; 24:263-273. [DOI: 10.1007/s10123-021-00163-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/10/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
|
644
|
Chakravarty K, Antontsev V, Bundey Y, Varshney J. Driving success in personalized medicine through AI-enabled computational modeling. Drug Discov Today 2021; 26:1459-1465. [PMID: 33609781 DOI: 10.1016/j.drudis.2021.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/26/2021] [Accepted: 02/10/2021] [Indexed: 12/29/2022]
Abstract
The development of successful drugs is expensive and time-consuming because of high clinical attrition rates. This is caused partially by the rupture seen in the translatability of the drug from the bench to the clinic in the context of personalized medicine. Artificial intelligence (AI)-driven platforms integrated with mechanistic modeling have become instrumental in accelerating the drug development process by leveraging data ubiquitously across the various phases. AI can counter the deficiencies and ambiguities that arise during the classical drug development process while reducing human intervention and bridging the translational gap in discovering the connections between drugs and diseases.
Collapse
Affiliation(s)
| | - Victor Antontsev
- VeriSIM Life Inc., 1 Sansome St. Suite 3500, San Francisco, CA 94104, USA
| | - Yogesh Bundey
- VeriSIM Life Inc., 1 Sansome St. Suite 3500, San Francisco, CA 94104, USA
| | - Jyotika Varshney
- VeriSIM Life Inc., 1 Sansome St. Suite 3500, San Francisco, CA 94104, USA.
| |
Collapse
|
645
|
Hommes JW, Verheijden RJ, Suijkerbuijk KPM, Hamann D. Biomarkers of Checkpoint Inhibitor Induced Immune-Related Adverse Events-A Comprehensive Review. Front Oncol 2021; 10:585311. [PMID: 33643899 PMCID: PMC7905347 DOI: 10.3389/fonc.2020.585311] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have substantially improved the prognosis of patients with different types of cancer. Through blockade of cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1), negative feedback mechanisms of the immune system are inhibited, potentially resulting in very durable anti-tumor responses. Despite their promise, ICIs can also elicit auto-immune toxicities. These immune-related adverse events (irAEs) can be severe and sometimes even fatal. Therefore, being able to predict severe irAEs in patients would be of added value in clinical decision making. A search was performed using “adverse events”, “immune checkpoint inhibitor”, “biomarker”, and synonyms in PubMed, yielding 3580 search results. After screening title and abstract on the relevance to the review question, statistical significance of reported potential biomarkers, and evaluation of the remaining full papers, 35 articles were included. Five additional reports were obtained by means of citations and by using the similar article function on PubMed. The current knowledge is presented in comprehensive tables summarizing blood-based, immunogenetic and microbial biomarkers predicting irAEs prior to and during ICI therapy. Until now, no single biomarker has proven to be sufficiently predictive for irAE development. Recommendations for further research on this topic are presented.
Collapse
Affiliation(s)
- Josefien W Hommes
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rik J Verheijden
- Department of Medical Oncology, Cancer Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, Cancer Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Dörte Hamann
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
646
|
Velikova T, Krastev B, Lozenov S, Gencheva R, Peshevska-Sekulovska M, Nikolaev G, Peruhova M. Antibiotic-Related Changes in Microbiome: The Hidden Villain behind Colorectal Carcinoma Immunotherapy Failure. Int J Mol Sci 2021; 22:1754. [PMID: 33578709 PMCID: PMC7916407 DOI: 10.3390/ijms22041754] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/26/2021] [Accepted: 02/05/2021] [Indexed: 02/05/2023] Open
Abstract
The interplay between drugs and microbiota is critical for successful treatment. An accumulating amount of evidence has identified the significant impact of intestinal microbiota composition on cancer treatment response, particularly immunotherapy. The possible molecular pathways of the interaction between immune checkpoint inhibitors (ICIs) and the microbiome can be used to reverse immunotherapy tolerance in cancer by using various kinds of interventions on the intestinal bacteria. This paper aimed to review the data available on how the antibiotic-related changes in human microbiota during colorectal cancer (CRC) treatment can affect and determine ICI treatment outcomes. We also covered the data that support the potential intimate mechanisms of both local and systemic immune responses induced by changes in the intestinal microbiota. However, further better-powered studies are needed to thoroughly assess the clinical significance of antibiotic-induced alteration of the gut microbiota and its impact on CRC treatment by direct observations of patients receiving antibiotic treatment.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Department of Clinical Immunology, University Hospital Lozenetz, Sofia University St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria
| | - Boris Krastev
- Clinic of Medical Oncology, MHAT Hospital for Women Health Nadezhda, 1330 Sofia, Bulgaria; (B.K.); (R.G.)
| | - Stefan Lozenov
- Laboratory for Control and Monitoring of the Antibiotic Resistance, National Centre for Infectious and Parasitic Diseases, 26 Yanko Sakazov Blvd, 1504 Sofia, Bulgaria;
| | - Radostina Gencheva
- Clinic of Medical Oncology, MHAT Hospital for Women Health Nadezhda, 1330 Sofia, Bulgaria; (B.K.); (R.G.)
| | - Monika Peshevska-Sekulovska
- Department of Gastroenterology, University Hospital Lozenetz, Sofia University St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (M.P.-S.); (M.P.)
| | - Georgi Nikolaev
- Faculty of Biology, Sofia University St. Kliment Ohridski, 1407 Sofia, Bulgaria;
| | - Milena Peruhova
- Department of Gastroenterology, University Hospital Lozenetz, Sofia University St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria; (M.P.-S.); (M.P.)
| |
Collapse
|
647
|
Brandvold KR, Miller CJ, Volk RF, Killinger BJ, Whidbey C, Wright AT. Activity-Based Protein Profiling of Bile Salt Hydrolysis in the Human Gut Microbiome with Beta-Lactam or Acrylamide-Based Probes. Chembiochem 2021; 22:1448-1455. [PMID: 33314683 DOI: 10.1002/cbic.202000748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/30/2020] [Indexed: 12/19/2022]
Abstract
Microbial bile salt hydrolases (BSHs) found in the intestine catalyze the deconjugation of taurine- and glycine-linked bile salts produced in the liver. The resulting bile salts are biological detergents and are critical in aiding lipophilic nutrient digestion. Therefore, the activity of BSHs in the gut microbiome is directly linked to human metabolism and overall health. Bile salt metabolism has also been associated with disease phenotypes such as liver and colorectal cancer. In order to reshape the gut microbiome to optimize bile salt metabolism, tools to characterize and quantify these processes must exist to enable a much-improved understanding of how metabolism goes awry in the face of disease, and how it can be improved through an altered lifestyle and environment. Furthermore, it is necessary to attribute metabolic activity to specific members and BSHs within the microbiome. To this end, we have developed activity-based probes with two different reactive groups to target bile salt hydrolases. These probes bind similarly to the authentic bile salt substrates, and we demonstrate enzyme labeling of active bile salt hydrolases by using purified protein, cell lysates, and in human stool.
Collapse
Affiliation(s)
- Kristoffer R Brandvold
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Carson J Miller
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Regan F Volk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Bryan J Killinger
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99163, USA
| | - Christopher Whidbey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Aaron T Wright
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99163, USA
| |
Collapse
|
648
|
Renson A, Mullan Harris K, Dowd JB, Gaydosh L, McQueen MB, Krauter KS, Shannahan M, Aiello AE. Early Signs of Gut Microbiome Aging: Biomarkers of Inflammation, Metabolism, and Macromolecular Damage in Young Adulthood. J Gerontol A Biol Sci Med Sci 2021; 75:1258-1266. [PMID: 32421783 DOI: 10.1093/gerona/glaa122] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Emerging links between gut microbiota and diseases of aging point to possible shared immune, metabolic, and cellular damage mechanisms, operating long before diseases manifest. We conducted 16S rRNA sequencing of fecal samples collected from a subsample (n = 668) of Add Health Wave V, a nationally representative longitudinal study of adults aged 32-42. An overlapping subsample (n = 345) included whole-blood RNA-seq. We examined associations between fecal taxonomic abundances and dried blood spot-based markers of lipid and glucose homeostasis and C-reactive protein (measured in Wave IV), as well as gene expression markers of inflammation, cellular damage, immune cell composition, and transcriptomic age (measured in Wave V), using Bayesian hierarchical models adjusted for potential confounders. We additionally estimated a co-abundance network between inflammation-related genes and bacterial taxa using penalized Gaussian graphical models. Strong and consistent microbiota associations emerged for HbA1c, glucose, C-reactive protein, and principal components of genes upregulated in inflammation, DNA repair, and reactive oxygen species, with Streptococcus infantis, Pseudomonas spp., and Peptoniphilus as major players for each. This pattern was largely echoed (though attenuated) for immunological cell composition gene sets, and only Serratia varied meaningfully by transcriptomic age. Network co-abundance indicated relationships between Prevotella sp., Bacteroides sp., and Ruminococcus sp. and gut immune/metabolic regulatory activity, and Ruminococcus sp, Dialister, and Butyrivibrio crossotus with balance between Th1 and Th2 inflammation. In conclusion, many common associations between microbiota and major physiologic aging mechanisms are evident in early-mid adulthood and suggest avenues for early detection and prevention of accelerated aging.
Collapse
Affiliation(s)
- Audrey Renson
- Department of Epidemiology, The Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - Kathleen Mullan Harris
- Department of Sociology and Carolina Population Center, University of North Carolina at Chapel Hill
| | - Jennifer B Dowd
- Leverhulme Centre for Demographic Science, University of Oxford, UK
| | - Lauren Gaydosh
- Center for Medicine, Health, and Society, Vanderbilt University, Nashville, Tennessee
| | - Matthew B McQueen
- Department of Integrative Physiology, University of Colorado Boulder
| | - Kenneth S Krauter
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder
| | | | - Allison E Aiello
- Department of Epidemiology, The Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| |
Collapse
|
649
|
Liu Y, Li Z, Wu Y, Jing X, Li L, Fang X. Intestinal Bacteria Encapsulated by Biomaterials Enhance Immunotherapy. Front Immunol 2021; 11:620170. [PMID: 33643302 PMCID: PMC7902919 DOI: 10.3389/fimmu.2020.620170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/31/2020] [Indexed: 12/12/2022] Open
Abstract
The human intestine contains thousands of bacterial species essential for optimal health. Aside from their pathogenic effects, these bacteria have been associated with the efficacy of various treatments of diseases. Due to their impact on many human diseases, intestinal bacteria are receiving increasing research attention, and recent studies on intestinal bacteria and their effects on treatments has yielded valuable results. Particularly, intestinal bacteria can affect responses to numerous forms of immunotherapy, especially cancer therapy. With the development of precision medicine, understanding the factors that influence intestinal bacteria and how they can be regulated to enhance immunotherapy effects will improve the application prospects of intestinal bacteria therapy. Further, biomaterials employed for the convenient and efficient delivery of intestinal bacteria to the body have also become a research hotspot. In this review, we discuss the recent findings on the regulatory role of intestinal bacteria in immunotherapy, focusing on immune cells they regulate. We also summarize biomaterials used for their delivery.
Collapse
Affiliation(s)
- Yilun Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongmin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuanyu Wu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiabin Jing
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Lin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
650
|
Liu W, Wang Y, Luo J, Liu M, Luo Z. Pleiotropic Effects of Metformin on the Antitumor Efficiency of Immune Checkpoint Inhibitors. Front Immunol 2021; 11:586760. [PMID: 33603734 PMCID: PMC7884468 DOI: 10.3389/fimmu.2020.586760] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/16/2020] [Indexed: 01/01/2023] Open
Abstract
Cancer is an important threat to public health because of its high morbidity and mortality. In recent decades, immune checkpoint inhibitors (ICIs) have ushered a new therapeutic era in clinical oncology. The rapid development of immune checkpoint therapy is due to its inspiring clinical efficacy in a group of cancer types. Metformin, an effective agent for the management of type 2 diabetes mellitus (T2DM), has shown beneficial effects on cancer prevention and cancer treatment. Emerging studies have suggested that metformin in combination with ICI treatment could improve the anticancer effects of ICIs. Hence, we conducted a review to summarize the effects of metformin on ICI therapy. We also review the pleiotropic mechanisms of metformin combined with ICIs in cancer therapy, including its direct and indirect effects on the host immune system.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Ying Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jianquan Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Mouze Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Zhiying Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|