701
|
The Gut Microbiota and Inflammation: An Overview. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17207618. [PMID: 33086688 PMCID: PMC7589951 DOI: 10.3390/ijerph17207618] [Citation(s) in RCA: 395] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022]
Abstract
The gut microbiota encompasses a diverse community of bacteria that carry out various functions influencing the overall health of the host. These comprise nutrient metabolism, immune system regulation and natural defence against infection. The presence of certain bacteria is associated with inflammatory molecules that may bring about inflammation in various body tissues. Inflammation underlies many chronic multisystem conditions including obesity, atherosclerosis, type 2 diabetes mellitus and inflammatory bowel disease. Inflammation may be triggered by structural components of the bacteria which can result in a cascade of inflammatory pathways involving interleukins and other cytokines. Similarly, by-products of metabolic processes in bacteria, including some short-chain fatty acids, can play a role in inhibiting inflammatory processes. In this review, we aimed to provide an overview of the relationship between the gut microbiota and inflammatory molecules and to highlight relevant knowledge gaps in this field. Based on the current literature, it appears that as the gut microbiota composition differs between individuals and is contingent on a variety of factors like diet and genetics, some individuals may possess bacteria associated with pro-inflammatory effects whilst others may harbour those with anti-inflammatory effects. Recent technological advancements have allowed for better methods of characterising the gut microbiota. Further research to continually improve our understanding of the inflammatory pathways that interact with bacteria may elucidate reasons behind varying presentations of the same disease and varied responses to the same treatment in different individuals. Furthermore, it can inform clinical practice as anti-inflammatory microbes can be employed in probiotic therapies or used to identify suitable prebiotic therapies.
Collapse
|
702
|
Marra A, Trapani D, Viale G, Criscitiello C, Curigliano G. Practical classification of triple-negative breast cancer: intratumoral heterogeneity, mechanisms of drug resistance, and novel therapies. NPJ Breast Cancer 2020; 6:54. [PMID: 33088912 PMCID: PMC7568552 DOI: 10.1038/s41523-020-00197-2] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is not a unique disease, encompassing multiple entities with marked histopathological, transcriptomic and genomic heterogeneity. Despite several efforts, transcriptomic and genomic classifications have remained merely theoretic and most of the patients are being treated with chemotherapy. Driver alterations in potentially targetable genes, including PIK3CA and AKT, have been identified across TNBC subtypes, prompting the implementation of biomarker-driven therapeutic approaches. However, biomarker-based treatments as well as immune checkpoint inhibitor-based immunotherapy have provided contrasting and limited results so far. Accordingly, a better characterization of the genomic and immune contexture underpinning TNBC, as well as the translation of the lessons learnt in the metastatic disease to the early setting would improve patients' outcomes. The application of multi-omics technologies, biocomputational algorithms, assays for minimal residual disease monitoring and novel clinical trial designs are strongly warranted to pave the way toward personalized anticancer treatment for patients with TNBC.
Collapse
Affiliation(s)
- Antonio Marra
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Dario Trapani
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Giulia Viale
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Carmen Criscitiello
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| |
Collapse
|
703
|
Zhang Q, Chen Y, Bai X, Liang T. Immune Checkpoint Blockade Therapy for Hepatocellular Carcinoma: Clinical Challenges and Considerations. Front Oncol 2020; 10:590058. [PMID: 33178615 PMCID: PMC7593704 DOI: 10.3389/fonc.2020.590058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
Although many approaches have been developed for the treatment of hepatocellular carcinoma (HCC) that has both high incidence and high mortality especially in Asian countries, the prognosis of HCC patients is still dismal. Immunotherapy, particularly immune checkpoint inhibitors show encouraging efficacy and have already been widely applied in clinic. However, in contrast to traditional therapies, immunotherapy brings many challenges when using in a real world, including biomarker discovery, response evaluation, adverse event treatment, etc. In this review, we proposed some important and intractable issues in current clinical practice regarding the strategy of immune checkpoint blockade, collected current evidence, and discuss the critical challenges and possible approaches to a bright future.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China
| | - Yiwen Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China
| |
Collapse
|
704
|
Chronopoulos A, Kalluri R. Emerging role of bacterial extracellular vesicles in cancer. Oncogene 2020; 39:6951-6960. [PMID: 33060855 PMCID: PMC7557313 DOI: 10.1038/s41388-020-01509-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/20/2020] [Accepted: 10/02/2020] [Indexed: 02/08/2023]
Abstract
Shedding of microbial extracellular vesicles constitutes a universal mechanism for inter-kingdom and intra-kingdom communication that is conserved among prokaryotic and eukaryotic microbes. In this review we delineate fundamental aspects of bacterial extracellular vesicles (BEVs) including their biogenesis, cargo composition, and interactions with host cells. We critically examine the evidence that BEVs from the host gut microbiome can enter the circulatory system to disseminate to distant organs and tissues. The potential involvement of BEVs in carcinogenesis is evaluated and future research ideas explored. We further discuss the potential of BEVs in microbiome-based liquid biopsies for cancer diagnostics and bioengineering strategies for cancer therapy.
Collapse
Affiliation(s)
- Antonios Chronopoulos
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Bioengineering, Rice University, Houston, TX, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
705
|
Abstract
Immunotherapy has transformed the treatment of many tumors. Robust data demonstrating improved overall survival and progression-free survival in patients treated with monoclonal antibodies have established immune checkpoint inhibitors as standard of care in stages III and IV non-small cell lung cancer. Nivolumab is effective in previously treated patients with metastatic non-small cell lung cancer. Pembrolizumab and atezolizumab are approved as monotherapy and in combination with other therapies. Ongoing trials investigate the potential role of immunotherapy in earlier disease settings. Identifying predictive biomarkers of response will further amplify the impact of immune checkpoint inhibitors in the treatment of non-small cell lung cancer.
Collapse
Affiliation(s)
- Melinda L Hsu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, 1650 Orleans Street CRB1 186, Baltimore, MD 21287, USA.
| | - Jarushka Naidoo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, 401 North Broadway, Baltimore, MD 21287, USA. https://twitter.com/DrJNaidoo
| |
Collapse
|
706
|
Wang J, Yang HR, Wang DJ, Wang XX. Association between the gut microbiota and patient responses to cancer immune checkpoint inhibitors. Oncol Lett 2020; 20:342. [PMID: 33123253 PMCID: PMC7583737 DOI: 10.3892/ol.2020.12205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Studies are increasingly investigating the association between the gut microbiota and the outcomes of immunotherapy in patients with cancer. Notably, certain studies have demonstrated that the gut microbiota serves a key role in regulating a patient's response to immunotherapy. In the present review, the potential associations between the gut microbiota, and cancer, host immunity and cancer immunotherapy are reviewed. Furthermore, the effects of fecal microbiota transplantation, antibiotics, probiotics, prebiotics, synbiotics, components of traditional Chinese medicine and various lifestyle factors on the gut microbiota and cancer immunotherapy outcomes are discussed. Certain dominant bacterial groups in the context of cancer immunotherapy and certain effective methods for optimizing immunotherapy by regulating the gut microbiota have been identified. Further investigation may enable the rapid conversion of these discoveries into practical products and clinically applicable methods.
Collapse
Affiliation(s)
- Jian Wang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Hong-Ru Yang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Dai-Jie Wang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, P.R. China
| | - Xing-Xia Wang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
707
|
Xiong Y, Song W, Shen L, Wang Y, Zhang J, Hu M, Liu Y, Li J, Musetti S, Liu R, Huang L. Oral Metformin and Polymetformin Reprogram Immunosuppressive Microenvironment and Boost Immune Checkpoint Inhibitor Therapy in Colorectal Cancer. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Yang Xiong
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
- College of Pharmaceutical Science Zhejiang Chinese Medical University Hangzhou Zhejiang 310053 China
| | - Wantong Song
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Limei Shen
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Ying Wang
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Jing Zhang
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Mengying Hu
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Yun Liu
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Jingjing Li
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Sara Musetti
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Rihe Liu
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Leaf Huang
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| |
Collapse
|
708
|
Antibiotics and Immunotherapy: Too Much of Anything is Bad! Eur Urol 2020; 78:544-545. [DOI: 10.1016/j.eururo.2020.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 11/17/2022]
|
709
|
Cammarota G, Ianiro G, Ahern A, Carbone C, Temko A, Claesson MJ, Gasbarrini A, Tortora G. Gut microbiome, big data and machine learning to promote precision medicine for cancer. Nat Rev Gastroenterol Hepatol 2020; 17:635-648. [PMID: 32647386 DOI: 10.1038/s41575-020-0327-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/02/2020] [Indexed: 12/13/2022]
Abstract
The gut microbiome has been implicated in cancer in several ways, as specific microbial signatures are known to promote cancer development and influence safety, tolerability and efficacy of therapies. The 'omics' technologies used for microbiome analysis continuously evolve and, although much of the research is still at an early stage, large-scale datasets of ever increasing size and complexity are being produced. However, there are varying levels of difficulty in realizing the full potential of these new tools, which limit our ability to critically analyse much of the available data. In this Perspective, we provide a brief overview on the role of gut microbiome in cancer and focus on the need, role and limitations of a machine learning-driven approach to analyse large amounts of complex health-care information in the era of big data. We also discuss the potential application of microbiome-based big data aimed at promoting precision medicine in cancer.
Collapse
Affiliation(s)
- Giovanni Cammarota
- Gastroenterology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Gianluca Ianiro
- Gastroenterology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Ahern
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Carmine Carbone
- Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andriy Temko
- School of Engineering, University College Cork, Cork, Ireland.,Qualcomm ML R&D, Cork, Ireland
| | - Marcus J Claesson
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Antonio Gasbarrini
- Gastroenterology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
710
|
Dieterich LC, Bikfalvi A. The tumor organismal environment: Role in tumor development and cancer immunotherapy. Semin Cancer Biol 2020; 65:197-206. [DOI: 10.1016/j.semcancer.2019.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/03/2019] [Accepted: 12/22/2019] [Indexed: 02/07/2023]
|
711
|
Combination of Detoxified Pneumolysin Derivative ΔA146Ply and Berbamine as a Treatment Approach for Breast Cancer. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:247-261. [PMID: 32728613 PMCID: PMC7369532 DOI: 10.1016/j.omto.2020.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022]
Abstract
Increasing evidence demonstrates that microorganisms and their products can modulate host responses to cancer therapies and contribute to tumor shrinkage via various mechanisms, including intracellular signaling pathways modulation and immunomodulation. Detoxified pneumolysin derivative ΔA146Ply is a pneumolysin mutant lacking hemolytic activity. To determine the antitumor activity of ΔA146Ply, the combination of ΔA146Ply and berbamine, a well-established antitumor agent, was used for breast cancer therapy, especially for triple-negative breast cancer. The efficacy of the combination therapy was evaluated in vitro using four breast cancer cell lines and in vivo using a synergistic mouse tumor model. We demonstrated that in vitro, the combination therapy significantly inhibited cancer cell proliferation, promoted cancer cell apoptosis, caused cancer cell-cycle arrest, and suppressed cancer cell migration and invasion. In vivo, the combination therapy significantly suppressed tumor growth and prolonged the median survival time of tumor-bearing mice partially through inhibiting tumor cell proliferation, promoting tumor cell apoptosis, and activating systemic antitumor immune responses. The safety analysis demonstrated that the combination therapy showed no obvious liver and kidney toxicity to tumor-bearing mice. Our study provides a new treatment option for breast cancer and lays the experimental basis for the development of ΔA146Ply as an antitumor agent.
Collapse
|
712
|
Kingkaw A, Nakphaichit M, Suratannon N, Nitisinprasert S, Wongoutong C, Chatchatee P, Krobthong S, Charoenlappanit S, Roytrakul S, Vongsangnak W. Analysis of the infant gut microbiome reveals metabolic functional roles associated with healthy infants and infants with atopic dermatitis using metaproteomics. PeerJ 2020; 8:e9988. [PMID: 33033661 PMCID: PMC7521340 DOI: 10.7717/peerj.9988] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/27/2020] [Indexed: 12/25/2022] Open
Abstract
The infant gut microbiome consists of a complex and diverse microbial community. Comprehensive taxonomic and metabolic functional knowledge about microbial communities supports medical and biological applications, such as fecal diagnostics. Among the omics approaches available for the investigation of microbial communities, metaproteomics-based analysis is a very powerful approach; under this method, the activity of microbial communities is explored by investigating protein expression within a sample. Through use of metaproteomics, this study aimed to investigate the microbial community composition of the infant gut to identify different key proteins playing metabolic functional roles in the microbiome of healthy infants and infants with atopic dermatitis in a Thai population-based birth cohort. Here, 18 fecal samples were analyzed by liquid chromatography-tandem mass spectrometry to conduct taxonomic, functional, and pathway-based protein annotation. Accordingly, 49,973 annotated proteins out of 68,232 total proteins were investigated in gut microbiome samples and compared between the healthy and atopic dermatitis groups. Through differentially expressed proteins (DEPs) analysis, 130 significant DEPs were identified between the healthy and atopic dermatitis groups. Among these DEPs, eight significant proteins were uniquely expressed in the atopic dermatitis group. For instance, triosephosphate isomerase (TPI) in Bifidobacteriaceae in the genus Alloscardovia and demethylmenaquinone methyltransferase (DMM) in Bacteroides were shown to potentially play metabolic functional roles related to disease. PPI network analysis revealed seven reporter proteins showing metabolic alterations between the healthy and disease groups associated with the biosynthesis of ubiquinone and other quinones as well as the energy supply. This study serves as a scaffold for microbial community-wide metabolic functional studies of the infant gut microbiome in relation to allergic disease.
Collapse
Affiliation(s)
- Amornthep Kingkaw
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Massalin Nakphaichit
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Narissara Suratannon
- Pediatric Allergy & Clinical Immunology Research Unit, Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Sunee Nitisinprasert
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Chantha Wongoutong
- Department of Statistics, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Pantipa Chatchatee
- Pediatric Allergy & Clinical Immunology Research Unit, Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Sucheewin Krobthong
- Proteomics Research Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Sawanya Charoenlappanit
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Wanwipa Vongsangnak
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, Thailand.,Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, Thailand
| |
Collapse
|
713
|
The association between proton pump inhibitors use and clinical outcome of patients receiving immune checkpoint inhibitors therapy. Int Immunopharmacol 2020; 88:106972. [PMID: 33182025 DOI: 10.1016/j.intimp.2020.106972] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/11/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The gut microbiome can mediate the efficacy of immune checkpoint inhibitors (ICI). Meanwhile, proton pump inhibitors (PPI) can modulate the gut microbiome significantly. However, the impact of PPI use on the clinical outcome of ICI therapy remains unclear. METHODS Searches of PubMed, EMBASE, and the Cochrane Library were conducted to retrieve studies exploring the relationship between PPI use and the clinical benefit of patients undergoing ICI therapy through June 2020. The pooled hazard ratio (HR) and 95% confidence intervals (CIs) were calculated to evaluate the influence of PPI use on overall survival (OS) and progression-free survival (PFS). RESULTS A total of seven studies were eligible for our final analysis. There was no significant association between PPI use and OS or PFS (PPI users versus non-users: HR for OS: 1.05, 95% CI: 0.79-1.40, P = 0.73; HR for PFS: 0.90, 95% CI: 0.66-1.23, P = 0.51). However, subgroup analyses demonstrated that PPI use was related to a superior PFS of melanoma patients (HR: 0.50, 95% CI: 0.28-0.91, P = 0.02) and an inferior PFS of non-small cell lung cancer (NSCLC) patients (HR: 1.17, 95% CI: 1.05-1.31, P = 0.006). CONCLUSIONS Our present study indicates that PPI use is not significantly associated with OS and PFS of patients undergoing ICI treatment. However, concomitant PPI therapy may have a positive effect on melanoma patients and a negative effect on NSCLC patients. It is advisable for clinical oncologists to evaluate the necessity for PPI use when they treat patients with ICI therapy.
Collapse
|
714
|
Drokhlyansky E, Smillie CS, Van Wittenberghe N, Ericsson M, Griffin GK, Eraslan G, Dionne D, Cuoco MS, Goder-Reiser MN, Sharova T, Kuksenko O, Aguirre AJ, Boland GM, Graham D, Rozenblatt-Rosen O, Xavier RJ, Regev A. The Human and Mouse Enteric Nervous System at Single-Cell Resolution. Cell 2020; 182:1606-1622.e23. [PMID: 32888429 PMCID: PMC8358727 DOI: 10.1016/j.cell.2020.08.003] [Citation(s) in RCA: 338] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/15/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
The enteric nervous system (ENS) coordinates diverse functions in the intestine but has eluded comprehensive molecular characterization because of the rarity and diversity of cells. Here we develop two methods to profile the ENS of adult mice and humans at single-cell resolution: RAISIN RNA-seq for profiling intact nuclei with ribosome-bound mRNA and MIRACL-seq for label-free enrichment of rare cell types by droplet-based profiling. The 1,187,535 nuclei in our mouse atlas include 5,068 neurons from the ileum and colon, revealing extraordinary neuron diversity. We highlight circadian expression changes in enteric neurons, show that disease-related genes are dysregulated with aging, and identify differences between the ileum and proximal/distal colon. In humans, we profile 436,202 nuclei, recovering 1,445 neurons, and identify conserved and species-specific transcriptional programs and putative neuro-epithelial, neuro-stromal, and neuro-immune interactions. The human ENS expresses risk genes for neuropathic, inflammatory, and extra-intestinal diseases, suggesting neuronal contributions to disease.
Collapse
MESH Headings
- Aging/genetics
- Aging/metabolism
- Animals
- Circadian Clocks/genetics
- Colon/cytology
- Colon/metabolism
- Endoplasmic Reticulum, Rough/genetics
- Endoplasmic Reticulum, Rough/metabolism
- Endoplasmic Reticulum, Rough/ultrastructure
- Enteric Nervous System/cytology
- Enteric Nervous System/metabolism
- Epithelial Cells/metabolism
- Female
- Gene Expression Regulation, Developmental/genetics
- Genetic Predisposition to Disease/genetics
- Humans
- Ileum/cytology
- Ileum/metabolism
- Inflammation/genetics
- Inflammation/metabolism
- Intestinal Diseases/genetics
- Intestinal Diseases/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Electron, Transmission
- Nervous System Diseases/genetics
- Nervous System Diseases/metabolism
- Neuroglia/cytology
- Neuroglia/metabolism
- Neurons/cytology
- Neurons/metabolism
- Nissl Bodies/genetics
- Nissl Bodies/metabolism
- Nissl Bodies/ultrastructure
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Seq
- Ribosomes/metabolism
- Ribosomes/ultrastructure
- Single-Cell Analysis/methods
- Stromal Cells/metabolism
Collapse
Affiliation(s)
- Eugene Drokhlyansky
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Maria Ericsson
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Gabriel K Griffin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Gokcen Eraslan
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael S Cuoco
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Olena Kuksenko
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrew J Aguirre
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Genevieve M Boland
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel Graham
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA, USA
| | | | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA.
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute and Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
715
|
Dharmawardana N, Goddard T, Woods C, Watson DI, Butler R, Ooi EH, Yazbeck R. Breath methane to hydrogen ratio as a surrogate marker of intestinal dysbiosis in head and neck cancer. Sci Rep 2020; 10:15010. [PMID: 32929151 PMCID: PMC7490703 DOI: 10.1038/s41598-020-72115-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 08/13/2020] [Indexed: 01/30/2023] Open
Abstract
Exhaled breath compounds can non-invasively detect head and neck squamous cell carcinoma (HNSCC). Here we investigated exhaled compounds related to intestinal bacterial carbohydrate fermentation. Fasting breath samples were collected into 3 litre FlexFoil PLUS bags from patients awaiting a biopsy procedure for suspected HNSCC. Samples were analysed using a Syft selected ion flow-tube mass spectrometer and a Quintron BreathTracker. Two tailed non-parametric significance testing was conducted with corrections for multiple imputations. 74 patients were diagnosed (histological) with HNSCC and 61 patients were benign (controls). The methane to hydrogen ratio was significantly different between cancer and non-cancer controls (p = 0.0440). This ratio increased with tumour stage with a significant difference between T1 and T4 tumours (p = 0.0259). Hydrogen levels were significantly higher in controls who were smokers (p = 0.0129), with no smoking dependent methane changes. There were no differences in short chain fatty acids between groups. Exhaled compounds of intestinal carbohydrate fermentation can detect HNSCC patients. These findings suggest a modified carbohydrate fermentation profile in HNSCC patients that is tumour stage and smoking status dependent.
Collapse
Affiliation(s)
- Nuwan Dharmawardana
- Department of Otorhinolaryngology-Head and Neck Surgery, Flinders Medical Centre, Bedford Park, Australia. .,Discipline of Surgery, College of Medicine and Public Health, Flinders University, Bedford Park, Australia.
| | - Thomas Goddard
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, Adelaide, Australia
| | - Charmaine Woods
- Department of Otorhinolaryngology-Head and Neck Surgery, Flinders Medical Centre, Bedford Park, Australia.,Discipline of Surgery, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - David I Watson
- Discipline of Surgery, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Ross Butler
- Discipline of Surgery, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Eng H Ooi
- Department of Otorhinolaryngology-Head and Neck Surgery, Flinders Medical Centre, Bedford Park, Australia.,Discipline of Surgery, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Roger Yazbeck
- Discipline of Surgery, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| |
Collapse
|
716
|
Deeg HJ. Individuals, Boundaries, and Graft-versus-Host Disease. Biol Blood Marrow Transplant 2020; 26:e309-e312. [PMID: 32927076 DOI: 10.1016/j.bbmt.2020.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/28/2020] [Accepted: 09/07/2020] [Indexed: 01/11/2023]
Abstract
Hematopoietic cell transplantation generates new individuals, transplant chimeras, composed of 2 genetic partners-the patient and donor-derived cells-no longer restricted by their original genomes. Interactions of donor-derived and recipient cells occur prominently at the boundary of the recipient with a third partner, the microbiome, in particular skin and intestinal tract, leading to disruption of microbiome homeostasis. These interactions of donor and patient cells at the boundary set the stage for the development of graft-versus-host disease, an expression of the defense of individuality by recipient and donor. Establishment of tolerance and return of homeostasis at the boundary will allow for the survival of the new integrated, physiologic individual.
Collapse
Affiliation(s)
- H Joachim Deeg
- Fred Hutchinson Cancer Research Center and the University of Washington, Seattle, Washington.
| |
Collapse
|
717
|
Abstract
The emergence of immune checkpoint inhibitors in recent years has transformed cancer treatment in many tumor types. Durable clinical responses in refractory cancer types have been observed. However, despite these promising long-term responses, the majority of patients fail to respond to immune checkpoint blockade, demonstrating primary resistance. In addition, some of those who initially respond to treatment eventually experience relapse secondary to acquired resistance. Both primary and acquired resistance is a result of complex and constantly evolving interactions between cancer cells and the immune system. Some mechanisms of resistance have been characterized to date, and more continue to be uncovered. The tumor microenvironment, tumor immunogenicity, and oncologic pathways play roles in response and resistance to immune checkpoint blockade. By elucidating and targeting mechanisms of resistance, treatments can be guided to improve clinical outcomes. Combination treatment strategies with immune checkpoint inhibitors are being tested in clinical trials, with several already in clinical use. This review will discuss the different resistance mechanisms and potential therapeutic strategies to overcome resistance.
Collapse
Affiliation(s)
- Xiao-Jun Liu
- Second Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Quan-Lin Guan
- Department of Surgical Oncology, the First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
718
|
Longhi G, van Sinderen D, Ventura M, Turroni F. Microbiota and Cancer: The Emerging Beneficial Role of Bifidobacteria in Cancer Immunotherapy. Front Microbiol 2020; 11:575072. [PMID: 33013813 PMCID: PMC7507897 DOI: 10.3389/fmicb.2020.575072] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022] Open
Abstract
Many intestinal bacteria are believed to be involved in various inflammatory and immune processes that influence tumor etiology because of their metabolic properties and their ability to alter the microbiota homeostasis. Although many functions of the microbiota are still unclear, there is compelling experimental evidence showing that the intestinal microbiota is able to modulate carcinogenesis and the response to anticancer therapies, both in the intestinal tract and other body sites. Among the wide variety of gut-colonizing microorganisms, various species belonging to the Bifidobacterium genus are believed to elicit beneficial effects on human physiology and on the host-immune system. Recent findings, based on preclinical mouse models and on human clinical trials, have demonstrated the impact of gut commensals including bifidobacteria on the efficacy of tumor-targeting immunotherapy. Although the underlying molecular mechanisms remain obscure, bifidobacteria and other microorganisms have become a promising aid to immunotherapeutic procedures that are currently applied to treat cancer. The present review focuses on strategies to recruit the microbiome in order to enhance anticancer responses and develop therapies aimed at fighting the onset and progression of malignancies.
Collapse
Affiliation(s)
- Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- Alimentary Pharmabotic Centre (APC) Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
719
|
Lee K, Zhang I, Kyman S, Kask O, Cope EK. Co-infection of Malassezia sympodialis With Bacterial Pathobionts Pseudomonas aeruginosa or Staphylococcus aureus Leads to Distinct Sinonasal Inflammatory Responses in a Murine Acute Sinusitis Model. Front Cell Infect Microbiol 2020; 10:472. [PMID: 33014894 PMCID: PMC7498577 DOI: 10.3389/fcimb.2020.00472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022] Open
Abstract
Host-associated bacteria and fungi, comprising the microbiota, are critical to host health. In the airways, the composition and diversity of the mucosal microbiota of patients are associated with airway health status. However, the relationship between airway microbiota and respiratory inflammation is not well-understood. Chronic rhinosinusitis (CRS) is a complex disease that affects up to 14% of the US population. Previous studies have shown decreased microbial diversity in CRS patients and enrichment of either Staphylococcus aureus or Pseudomonas aeruginosa. Although bacterial community composition is variable across CRS patients, Malassezia is a dominant fungal genus in the upper airways of the majority of healthy and CRS subjects. We hypothesize that distinct bacterial-fungal interactions differentially influence host mucosal immune response. Thus, we investigated in vitro and in vivo interactions between Malassezia sympodialis, P. aeruginosa, and S. aureus. The in vitro interactions were evaluated using the modified Kirby-Bauer Assay, Crystal Violet assay for biofilm, and FISH. A pilot murine model of acute sinusitis was used to investigate relationships with the host immune response. S. aureus and P. aeruginosa were intranasally instilled in the presence or absence of M. sympodialis (n = 66 total mice; 3–5/group). Changes in the microbiota were determined using 16S rRNA gene sequencing and host immune response was measured using quantitative real-time PCR (qRT-PCR). In vitro, only late stage planktonic P. aeruginosa and its biofilms inhibited M. sympodialis. Co-infection of mice with M. sympodialis and P. aeruginosa or S. aureus differently influenced the immune response. In co-infected mice, we demonstrate different expression of fungal sensing (Dectin-1), allergic responses (IL-5, and IL-13) and inflammation (IL-10, and IL-17) in murine sinus depending on the bacterial species that co-infected with M. sympodialis (p < 0.05). The pilot results suggest that species-specific interactions in airway-associated microbiota may be implicated driving immune responses. The understanding of the role of bacterial-fungal interactions in CRS will contribute to development of novel therapies toward manipulation of the airway microbiota.
Collapse
Affiliation(s)
- Keehoon Lee
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| | - Irene Zhang
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| | - Shari Kyman
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| | - Oliver Kask
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| | - Emily Kathryn Cope
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| |
Collapse
|
720
|
Lee K, Zhang I, Kyman S, Kask O, Cope EK. Co-infection of Malassezia sympodialis With Bacterial Pathobionts Pseudomonas aeruginosa or Staphylococcus aureus Leads to Distinct Sinonasal Inflammatory Responses in a Murine Acute Sinusitis Model. Front Cell Infect Microbiol 2020. [DOI: 10.10.3389/fcimb.2020.00472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
721
|
Tan HY, Toh YC. What can microfluidics do for human microbiome research? BIOMICROFLUIDICS 2020; 14:051303. [PMID: 33062112 PMCID: PMC7538166 DOI: 10.1063/5.0012185] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/17/2020] [Indexed: 05/13/2023]
Abstract
Dysregulation of the human microbiome has been linked to various disease states, which has galvanized the efforts to modulate human health through microbiomes. Currently, human microbiome research is going through several phases to identify the constituent components of the microbiome, associate microbiome changes with physiological and pathological states, understand causative relationships, and finally translate this knowledge into therapeutics and diagnostics. The convergence of microfluidic technologies with molecular and cell profiling, microbiology, and tissue engineering can potentially be applied to these different phases of microbiome research to overcome the existing challenges faced by conventional approaches. The goal of this paper is to discuss and highlight the opportunities of applying different microfluidic technologies to specific areas of microbiome research as well as unique challenges that microfluidics must overcome when working with microbiome-relevant biological materials, e.g., micro-organisms, host tissues, and fluids. We will discuss the applicability of integrated microfluidic systems for processing biological samples for genomic sequencing analyses. For functional analysis of the microbiota, we will cover state-of-the-art microfluidic devices for microbiota cultivation and functional measurements. Finally, we highlight the use of organs-on-chips to model various microbiome-host tissue interactions. We envision that microfluidic technologies may hold great promise in advancing the knowledge on the interplay between microbiome and human health, as well as its eventual translation into microbiome-based diagnostics and therapeutics.
Collapse
Affiliation(s)
- Hsih-Yin Tan
- Institute for Health Innovation and Technology, National University of Singapore, Singapore 117599
| | - Yi-Chin Toh
- Author to whom correspondence should be addressed:
| |
Collapse
|
722
|
Mangan BL, McAlister RK, Balko JM, Johnson DB, Moslehi JJ, Gibson A, Phillips EJ. Evolving insights into the mechanisms of toxicity associated with immune checkpoint inhibitor therapy. Br J Clin Pharmacol 2020; 86:1778-1789. [PMID: 32543711 PMCID: PMC7444794 DOI: 10.1111/bcp.14433] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Immune checkpoint inhibitors have emerged as a revolutionary treatment option for patients with various types of malignancy. Although these agents afford a significant improvement in outcomes for melanoma and other previously untreatable malignancies, their novel mechanism of action may predispose patients to immune-related adverse effects (irAEs). In the tumour neoantigen environment, these irAEs are due to the activation of the immune system by the blockade of suppressive checkpoints, leading to increases in T-cell activation and proliferation. IrAEs have been reported in almost any organ and at any point in time, even months to years after discontinuation of therapy. Certain populations with distinct physiological changes, genetic risk factors, and specific antigen exposures may be more highly predisposed to develop irAEs. This review discusses the incidence and mechanisms of irAEs and the relationship between host factors and irAE occurrence.
Collapse
Affiliation(s)
- Brendan L. Mangan
- Department of PharmacyVanderbilt University Medical CenterNashvilleTNUSA
| | - Renee K. McAlister
- Department of PharmacyVanderbilt University Medical CenterNashvilleTNUSA
| | - Justin M. Balko
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
- Breast Cancer Research ProgramVanderbilt University Medical CenterNashvilleTNUSA
- Cancer Biology ProgramVanderbilt University Medical CenterNashvilleTNUSA
| | - Douglas B. Johnson
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Javid J. Moslehi
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Andrew Gibson
- Institute for Immunology and Infectious DiseasesMurdoch UniversityMurdochWAAustralia
| | - Elizabeth J. Phillips
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
- Department of PharmacologyVanderbilt University Medical CenterNashvilleTNUSA
- Institute for Immunology and Infectious DiseasesMurdoch UniversityMurdochWAAustralia
| |
Collapse
|
723
|
Katongole P, Sande OJ, Joloba M, Reynolds SJ, Niyonzima N. The human microbiome and its link in prostate cancer risk and pathogenesis. Infect Agent Cancer 2020; 15:53. [PMID: 32884579 PMCID: PMC7460756 DOI: 10.1186/s13027-020-00319-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
There is growing evidence of the microbiome's role in human health and disease since the human microbiome project. The microbiome plays a vital role in influencing cancer risk and pathogenesis. Several studies indicate microbial pathogens to account for over 15-20% of all cancers. Furthermore, the interaction of the microbiota, especially the gut microbiota in influencing response to chemotherapy, immunotherapy, and radiotherapy remains an area of active research. Certain microbial species have been linked to the improved clinical outcome when on different cancer therapies. The recent discovery of the urinary microbiome has enabled the study to understand its connection to genitourinary malignancies, especially prostate cancer. Prostate cancer is the second most common cancer in males worldwide. Therefore research into understanding the factors and mechanisms associated with prostate cancer etiology, pathogenesis, and disease progression is of utmost importance. In this review, we explore the current literature concerning the link between the gut and urinary microbiome and prostate cancer risk and pathogenesis.
Collapse
Affiliation(s)
- Paul Katongole
- Department of Medical Microbiology, College of Health Sciences Makerere University, Kampala, Uganda
- Department of Medical Biochemistry, College of Health Sciences Makerere University, Kampala, Uganda
| | - Obondo J. Sande
- Department of Immunology and Molecular biology, College of Health Sciences Makerere University, Kampala, Uganda
| | - Moses Joloba
- Department of Immunology and Molecular biology, College of Health Sciences Makerere University, Kampala, Uganda
| | - Steven J. Reynolds
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | | |
Collapse
|
724
|
Langheinrich M, Wirtz S, Kneis B, Gittler MM, Tyc O, Schierwagen R, Brunner M, Krautz C, Weber GF, Pilarsky C, Trebicka J, Agaimy A, Grützmann R, Kersting S. Microbiome Patterns in Matched Bile, Duodenal, Pancreatic Tumor Tissue, Drainage, and Stool Samples: Association with Preoperative Stenting and Postoperative Pancreatic Fistula Development. J Clin Med 2020; 9:jcm9092785. [PMID: 32872220 PMCID: PMC7563524 DOI: 10.3390/jcm9092785] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/10/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Postoperative complications after pancreatic surgery are still a significant problem in clinical practice. The aim of this study was to characterize and compare the microbiomes of different body compartments (bile duct, duodenal mucosa, pancreatic tumor lesion, postoperative drainage fluid, and stool samples; preoperative and postoperative) in patients undergoing pancreatic surgery for suspected pancreatic cancer, and their association with relevant clinical factors (stent placement, pancreatic fistula, and gland texture). For this, solid (duodenal mucosa, pancreatic tumor tissue, stool) and liquid (bile, drainage fluid) biopsy samples of 10 patients were analyzed using 16s rRNA gene next-generation sequencing. Our analysis revealed: (i) a distinct microbiome in the different compartments, (ii) markedly higher abundance of Enterococcus in patients undergoing preoperative stent placement in the common bile duct, (iii) significant differences in the beta diversity between patients who developed a postoperative pancreatic fistula (POPF B/C), (iv) patients with POPF B/C were more likely to have bacteria belonging to the genus Enterococcus, and (v) differences in microbiome composition with regard to the pancreatic gland texture. The structure of the microbiome is distinctive in different compartments, and can be associated with the development of a postoperative pancreatic fistula.
Collapse
Affiliation(s)
- Melanie Langheinrich
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
- Correspondence:
| | - Stefan Wirtz
- Department of Internal Medicine 1, University Hospital of Erlangen, 91054 Erlangen, Germany;
| | - Barbara Kneis
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
| | - Matthias M. Gittler
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
| | - Olaf Tyc
- Translational Hepatology, Department of Internal Medicine I, University Clinic Frankfurt, 60590 Frankfurt, Germany; (O.T.); (R.S.); (J.T.)
| | - Robert Schierwagen
- Translational Hepatology, Department of Internal Medicine I, University Clinic Frankfurt, 60590 Frankfurt, Germany; (O.T.); (R.S.); (J.T.)
| | - Maximilian Brunner
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
| | - Christian Krautz
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
| | - Georg F. Weber
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
| | - Christian Pilarsky
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
| | - Jonel Trebicka
- Translational Hepatology, Department of Internal Medicine I, University Clinic Frankfurt, 60590 Frankfurt, Germany; (O.T.); (R.S.); (J.T.)
| | - Abbas Agaimy
- Department of Pathology, University Hospital of Erlangen, 91054 Erlangen, Germany;
| | - Robert Grützmann
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
| | - Stephan Kersting
- Department of Surgery, University Hospital of Erlangen, 91054 Erlangen, Germany; (B.K.); (M.M.G.); (M.B.); (C.K.); (G.F.W.); (C.P.); (R.G.); (S.K.)
| |
Collapse
|
725
|
Sun JY, Zhang D, Wu S, Xu M, Zhou X, Lu XJ, Ji J. Resistance to PD-1/PD-L1 blockade cancer immunotherapy: mechanisms, predictive factors, and future perspectives. Biomark Res 2020; 8:35. [PMID: 32864132 PMCID: PMC7450549 DOI: 10.1186/s40364-020-00212-5] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022] Open
Abstract
PD-1/PD-L1 blockade therapy is a promising cancer treatment strategy, which has revolutionized the treatment landscape of malignancies. Over the last decade, PD-1/PD-L1 blockade therapy has been trialed in a broad range of malignancies and achieved clinical success. Despite the potentially cure-like survival benefit, only a minority of patients are estimated to experience a positive response to PD-1/PD-L1 blockade therapy, and the primary or acquired resistance might eventually lead to cancer progression in patients with clinical responses. Accordingly, the resistance to PD-1/PD-L1 blockade remains a significant challenge hindering its further application. To overcome the limitation in therapy resistance, substantial effort has been made to improve or develop novel anti-PD-1/PD-L1 based immunotherapy strategies with better clinical response and reduced immune-mediated toxicity. In this review, we provide an overview on the resistance to PD-1/PD-L1 blockade and briefly introduce the mechanisms underlying therapy resistance. Moreover, we summarize potential predictive factors for the resistance to PD-1/PD-L1 blockade. Furthermore, we give an insight into the possible solutions to improve efficacy and clinical response. In the following research, combined efforts of basic researchers and clinicians are required to address the limitation of therapy resistance.
Collapse
Affiliation(s)
- Jin-Yu Sun
- The First College of Clinical Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Dengke Zhang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University/ The Fifth Affiliated Hospital of Wenzhou Medical University/ Clinical Medicine of Center Hospital of Lishui College, Lishui, 323000 China
- College of Medicine, Lishui College, Lishui, 323000 China
| | - Songquan Wu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University/ The Fifth Affiliated Hospital of Wenzhou Medical University/ Clinical Medicine of Center Hospital of Lishui College, Lishui, 323000 China
- College of Medicine, Lishui College, Lishui, 323000 China
| | - Min Xu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University/ The Fifth Affiliated Hospital of Wenzhou Medical University/ Clinical Medicine of Center Hospital of Lishui College, Lishui, 323000 China
- College of Medicine, Lishui College, Lishui, 323000 China
| | - Xiao Zhou
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Jie Lu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University/ The Fifth Affiliated Hospital of Wenzhou Medical University/ Clinical Medicine of Center Hospital of Lishui College, Lishui, 323000 China
- College of Medicine, Lishui College, Lishui, 323000 China
- Department of radiology, Affiliated Lishui Hospital of Zhejiang University, Lishui, 323000 China
| |
Collapse
|
726
|
Scazzocchio B, Minghetti L, D’Archivio M. Interaction between Gut Microbiota and Curcumin: A New Key of Understanding for the Health Effects of Curcumin. Nutrients 2020; 12:E2499. [PMID: 32824993 PMCID: PMC7551052 DOI: 10.3390/nu12092499] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Curcumin, a lipophilic polyphenol contained in the rhizome of Curcuma longa (turmeric), has been used for centuries in traditional Asian medicine, and nowadays it is widely used in food as dietary spice worldwide. It has received considerable attention for its pharmacological activities, which appear to act primarily through anti-inflammatory and antioxidant mechanisms. For this reason, it has been proposed as a tool for the management of many diseases, among which are gastrointestinal and neurological diseases, diabetes, and several types of cancer. However, the pharmacology of curcumin remains to be elucidated; indeed, a discrepancy exists between the well-documented in vitro and in vivo activities of curcumin and its poor bioavailability and chemical instability that should limit any therapeutic effect. Recently, it has been hypothesized that curcumin could exert direct regulative effects primarily in the gastrointestinal tract, where high concentrations of this polyphenol have been detected after oral administration. Consequently, it might be hypothesized that curcumin directly exerts its regulatory effects on the gut microbiota, thus explaining the paradox between its low systemic bioavailability and its wide pharmacological activities. It is well known that the microbiota has several important roles in human physiology, and its composition can be influenced by a multitude of environmental and lifestyle factors. Accordingly, any perturbations in gut microbiome profile or dysbiosis can have a key role in human disease progression. Interestingly, curcumin and its metabolites have been shown to influence the microbiota. It is worth noting that from the interaction between curcumin and microbiota two different phenomena arise: the regulation of intestinal microflora by curcumin and the biotransformation of curcumin by gut microbiota, both of them potentially crucial for curcumin activity. This review summarizes the most recent studies on this topic, highlighting the strong connection between curcumin and gut microbiota, with the final aim of adding new insight into the potential mechanisms by which curcumin exerts its effects.
Collapse
Affiliation(s)
- Beatrice Scazzocchio
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Luisa Minghetti
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Massimo D’Archivio
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| |
Collapse
|
727
|
Aydin AM, Chahoud J, Adashek JJ, Azizi M, Magliocco A, Ross JS, Necchi A, Spiess PE. Understanding genomics and the immune environment of penile cancer to improve therapy. Nat Rev Urol 2020; 17:555-570. [DOI: 10.1038/s41585-020-0359-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2020] [Indexed: 02/07/2023]
|
728
|
Phase II Clinical Trial of Pembrolizumab in Patients with Progressive Metastatic Pheochromocytomas and Paragangliomas. Cancers (Basel) 2020; 12:cancers12082307. [PMID: 32824391 PMCID: PMC7465458 DOI: 10.3390/cancers12082307] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023] Open
Abstract
Metastatic pheochromocytomas and paragangliomas (MPPGs) are rare endocrine malignancies that are associated with high rates of morbidity and mortality because of their large tumor burden and location, progression, and release of catecholamines. Systemic therapies for MPPGs are limited. MPPGs are characterized by pseudohypoxia that may prevent immune system recognition. We conducted a phase II clinical trial of pembrolizumab in patients with progressive MPPGs. The primary endpoint was the non-progression rate at 27 weeks. The secondary endpoints included the objective response and clinical benefit rates, progression free and overall survival duration, and safety. We also determined whether PDL-1 expression and the presence of infiltrating mononuclear inflammatory cells in the primary tumor were associated with clinical response and hereditary background. Eleven patients were included in this trial, four (36%) with germline mutations and seven (64%) with hormonally active tumors. Four patients (40%, 95% confidence interval (CI) 12–74%) achieved the primary endpoint. The objective response rate was 9% (95% CI: 0–41%). The clinical benefit rate was 73% (95% CI: 39–94%). Four patients had grade 3 adverse events related to pembrolizumab. No patients experienced grade 4 or 5 adverse events or a catecholamine crisis. Progression free survival time was 5.7 months (95% CI: 4.37—not reached). The median survival duration was 19 months (95% CI: 9.9—not reached). PDL-1 expression and the presence of infiltrating mononuclear inflammatory cells in the primary tumor did not seem to be associated with disease response. Single-agent pembrolizumab has modest treatment efficacy in patients with progressive MPPGs. Positive responses seemed to be independent of patients’ hereditary backgrounds, tumor hormonal status, and the presence of infiltrating mononuclear inflammatory cells or PDL-1 expression in the primary tumor.
Collapse
|
729
|
Abstract
The host immune system shapes the fate of tumor progression. Hence, manipulating patients' immune system to activate host immune responses against cancer pathogenesis is a promising strategy to develop effective therapeutic interventions for metastatic and drug-resistant cancers. Understanding the dynamic mechanisms within the tumor microenvironment (TME) that contribute to heterogeneity and metabolic plasticity is essential to enhance the patients' responsiveness to immune targeted therapies. Riera-Domingo et al. (Riera-Domingo C, Audige A, Granja S, Cheng WC, Ho PC, Baltazar F, Stockmann C, Mazzone, M. Physiol Rev 100: 1-102, 2020) describe the immune landscape within the TME and highlight the significance of metabolic and hypoxic signatures that impact immune function and response to immunotherapy strategies. Current literature in this field confirms that targeting tumor metabolism and the acidic microenvironment commonly associated with tumors may present viable strategies to modulate the host immune system in favor of response to immune targeted therapies. However, development of better tools to understand tumor-immune interactions and identify mechanisms driving nonresponders, more innovative clinical trial design, and new therapies will need to be identified to move the field forward. Personalized immune therapies incorporating metabolic and microbiome-based gene signatures to influence the therapeutic response and novel methods to generate immunologically "hot" tumors are at the forefront of immunotherapy currently. The combination of these approaches with clinically approved immunotherapies will be valuable moving forward.
Collapse
Affiliation(s)
- Kayla F Goliwas
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jessy S Deshane
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Craig A Elmets
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mohammad Athar
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
730
|
Yang M, Wang Y, Yuan M, Tao M, Kong C, Li H, Tong J, Zhu H, Yan X. Antibiotic administration shortly before or after immunotherapy initiation is correlated with poor prognosis in solid cancer patients: An up-to-date systematic review and meta-analysis. Int Immunopharmacol 2020; 88:106876. [PMID: 32799113 DOI: 10.1016/j.intimp.2020.106876] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/16/2020] [Accepted: 08/03/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Immune checkpoint inhibitors (ICIs) have recently achieved inspiring performance in improving the prognosis of various solid tumors. Gut microbiome plays a crucial modulatory role in the efficacy of ICIs, which can be influenced by antibiotic (ATB) administration. In this meta-analysis, we aimed to clarify the correlations of ATB administration with the prognosis of solid cancer patients receiving ICI treatment. METHOD The eligible literatures were searched using PubMed, Cochrane Library, Web of Science, and Clinical trials.gov databases before 29 February 2020. The correlations of ATB administration with overall survival (OS) and progression-free survival (PFS) were determined using Hazard ratios (HRs) coupled with 95% confidence intervals (CIs). RESULTS A total of 33 studies enrolling 5565 solid cancer patients receiving ICI treatment were included in this meta-analysis. As a whole, ATB administration was significantly correlated worse OS (HR = 1.76, 95%CI = 1.41-2.19, P < 0.00001) and PFS (HR = 1.76, 95%CI = 1.47-2.12, P < 0.00001). This significant association was then observed in the subgroup analysis based on region (except for OS in Europe), sample size, age, therapeutic strategy and ICI type. The similar results were also found in subgroup analysis for lung, renal cell (except for OS) and other cancers (such as melanoma) but not for mixed cancers. In addition, the ICI efficacy was more likely to be diminished by ATB administration within a time frame from 60 days before to 60 days after ICI initiation. CONCLUSION ATBs should be used cautiously in solid cancer patients receiving ICIs. However, further validations are still essential due to existing publication bias.
Collapse
Affiliation(s)
- Mengxue Yang
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Ying Wang
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Man Yuan
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Mingyang Tao
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Cheng Kong
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Hao Li
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA; Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Jiandong Tong
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
| | - Huiyuan Zhu
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.
| | - Xuebing Yan
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
| |
Collapse
|
731
|
Qu G, Sun C, Sharma M, Uy JP, Song EJ, Bhan C, Shu L. Is antibiotics use really associated with increased risk of colorectal cancer? An updated systematic review and meta-analysis of observational studies. Int J Colorectal Dis 2020; 35:1397-1412. [PMID: 32504337 DOI: 10.1007/s00384-020-03658-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2020] [Indexed: 02/04/2023]
Abstract
PURPOSE The association between antibiotics and colorectal cancer (CRC) risk has drawn increasing attention but remains controversial. This study was performed to clarify the association. METHODS A systematic review and meta-analysis was performed on seven electronic databases. The pooled odds ratios (OR) with a 95% confidence interval (CI) were calculated to estimate the association using the fixed-effects model or the random-effects model. RESULTS Ten studies that contained 4,853,289 participants were included in our study. We found that antibiotics use was associated with a higher risk of CRC (OR 1.09, 95%CI 1.02-1.17, I2 = 92.8%). More than 60 days of antibiotics use and 5 prescriptions of antibiotics were significantly associated with a higher risk of CRC. Sub-analysis on different types of antibiotics found that anti-anaerobic antibiotics, penicillins, and quinolones use led to increased risk of CRC (OR 1.22, 95% CI 1.04-1.44, I2 = 89.1%; OR 1.09, 95% CI 1.04-1.13, I2 = 69.2%; OR 1.15, 95% CI 1.03-1.35, I2 = 88.2%; respectively) and colon cancer (OR 1.28, 95% CI 1.04-1.58, I2 = 98.5%; OR 1.09, 95% CI 1.05-1.12, I2 = 0; OR 1.09, 95% CI 1.04-1.15, I2 = 0; respectively). However, antibiotics use was not significantly associated with rectal cancer (OR 1.03, 95% CI 0.92-1.16, I2 = 77.6%). CONCLUSION It needs attention that antibiotics use is associated with a higher risk of CRC, especially for colon cancer. Clinicians should be aware of the potential risk of CRC when prescribing anti-anaerobic antibiotics, penicillins, and quinolones in the future. Further studies are needed to assess any potential differences by tumor sites and class of antibiotics.
Collapse
Affiliation(s)
- Guangbo Qu
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Chenyu Sun
- Internal Medicine, AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL, 60657, USA.
| | - Monica Sharma
- Infectious Disease, AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL, 60657, USA
| | - John Patrick Uy
- Internal Medicine, AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL, 60657, USA
| | - Evelyn J Song
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chandur Bhan
- Internal Medicine, AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL, 60657, USA
| | - Liqin Shu
- Department of Child Health Care, Maternal and Child Health Care Hospital of Anhui Province (Affiliated Maternal and Child Health Care Hospital of Anhui Medical University), Hefei, 230001, Anhui, China
| |
Collapse
|
732
|
Wang LS, Mo YY, Huang YW, Echeveste CE, Wang HT, Chen J, Oshima K, Yearsley M, Simal-Gandaraf J, Battino M, Xiao J, Chen J, Sun C, Yu J, Bai W. Effects of Dietary Interventions on Gut Microbiota in Humans and the Possible Impacts of Foods on Patients' Responses to Cancer Immunotherapy. EFOOD 2020; 1:279-287. [PMID: 34308386 PMCID: PMC8301224 DOI: 10.2991/efood.k.200824.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota-the community of microorganisms in the gut-has been implicated in many physical and mental disorders in addition to intestinal diseases. Diets are the most studied and promising factors for altering it. Indeed, certain dietary interventions that increase fiber intake rapidly change levels of certain nutrients that can modify the composition of the microbiota, promoting richness and diversity. Recent intriguing evidence from several human clinical trials suggested that the composition and diversity of patients' gut microbiotas at baseline can influence their responses to cancer immunotherapy. If the factors that influence the gut microbiota were fully understood, it is conceivable that manipulating them could boost therapeutic responses in cancer patients. In this review, we investigate the possibility of using fruits, vegetables, or whole grains to enhance response to cancer therapies in humans, as current evidence suggests that these dietary components can manipulate and enhance diversity of the gut microbiota. Accordingly, dietary interventions with locally available fruits, vegetables, and whole grains might be an affordable and safe approach to enhancing the diversity of the gut microbiota before immunotherapy, in turn improving patients' responses to their treatments.
Collapse
Affiliation(s)
- Li-Shu Wang
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yue Yang Mo
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yi-Wen Huang
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carla Elena Echeveste
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hsin-Tzu Wang
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jiali Chen
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou, China
| | - Kiyoko Oshima
- Department of Pathology, Johns Hopkins University, MD, USA
| | | | - Jesus Simal-Gandaraf
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo, Spain
| | - Maurizio Battino
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo, Spain
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China
- Department of Odontostomatologic and Specialized Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, Ancona, Italy
| | - Jianbo Xiao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau, China
| | - Jiebiao Chen
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Zijingang Campus, Hangzhou, China
| | - Chongde Sun
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Zijingang Campus, Hangzhou, China
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, CA, USA
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou, China
| |
Collapse
|
733
|
Muthukrishnan PT, Vishwanathan S. Leading From Darkness to Light: Understanding Chronic Disease and Coronavirus: An Integrative Perspective. Integr Med (Encinitas) 2020; 19:8-12. [PMID: 33132779 PMCID: PMC7572137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Disease is defined as complex interaction of 3 things: disease-causing agent, host and environment. The government, CDC and hospital administrations have had reasonable success with the effects of social distancing and coronavirus testing; kudos to them. That addresses 1 of the 3 main factors of a disease; i.e. disease -causing agent. However, there are 2 other ingredients in the complex of a disease; i.e. the host of the disease and finally the environment of the disease. We are the hosts and it is time we take ownership of this ourselves and join the effort with the government towards understanding the dysregulated immune response seen in severe covid-19 and enable the public in strengthening the inner environment and the host immune response. Social distancing can be complete only when the host is prepared to fight the war in the coming fall and winter seasons. The outer environment seems to spontaneously recharging itself as a consequence of social distancing.
Collapse
Affiliation(s)
| | - Swati Vishwanathan
- Fellow in Medical Oncology, Department of Medicine, SUNY Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
734
|
Hewes SA, Wilson RL, Estes MK, Shroyer NF, Blutt SE, Grande-Allen KJ. In Vitro Models of the Small Intestine: Engineering Challenges and Engineering Solutions. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:313-326. [PMID: 32046599 PMCID: PMC7462033 DOI: 10.1089/ten.teb.2019.0334] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
Pathologies affecting the small intestine contribute significantly to the disease burden of both the developing and the developed world, which has motivated investigation into the disease mechanisms through in vitro models. Although existing in vitro models recapitulate selected features of the intestine, various important aspects have often been isolated or omitted due to the anatomical and physiological complexity. The small intestine's intricate microanatomy, heterogeneous cell populations, steep oxygen gradients, microbiota, and intestinal wall contractions are often not included in in vitro experimental models of the small intestine, despite their importance in both intestinal biology and pathology. Known and unknown interdependencies between various physiological aspects necessitate more complex in vitro models. Microfluidic technology has made it possible to mimic the dynamic mechanical environment, signaling gradients, and other important aspects of small intestinal biology. This review presents an overview of the complexity of small intestinal anatomy and bioengineered models that recapitulate some of these physiological aspects.
Collapse
Affiliation(s)
- Sarah A. Hewes
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Reid L. Wilson
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
735
|
Li Q, Yu H. The role of non- H. pylori bacteria in the development of gastric cancer. Am J Cancer Res 2020; 10:2271-2281. [PMID: 32905382 PMCID: PMC7471357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 07/05/2020] [Indexed: 06/11/2023] Open
Abstract
There is a complex ecosystem of bacteria and other microorganisms inside and outside the human body, which play an intricate role in maintaining health. In recent years, many researches focused on the relationship between microorganisms and cancer. Studies have identified that numerous microbes are presented in human stomach, which are closely linked to the development of gastric cancer (GC). Helicobacter pylori (H. pylori) is the mostly well-studied bacterial pathogen in the stomach, which account for the vast majority of GC. However, recent studies have found that microflora dysbiosis was occurred in mucosa of GC patients, and evidences have potentially proved that microbes other than H. pylori are also contribute to the development of GC, while the overall knowledge is still limited. In this review, we summarized the role of gastric flora in GC, especially the possible role of non-H. pylori bacteria in the development of GC. These knowledges and awareness may open doors for new therapeutic strategies of GC.
Collapse
Affiliation(s)
- Qing Li
- Department of Gastroenterology, Renmin Hospital of Wuhan UniversityWuhan 430060, Hubei, P. R. China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan UniversityWuhan 430060, Hubei, P. R. China
| | - Honggang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan UniversityWuhan 430060, Hubei, P. R. China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan UniversityWuhan 430060, Hubei, P. R. China
| |
Collapse
|
736
|
Zhao K, Hu Y. Microbiome harbored within tumors: a new chance to revisit our understanding of cancer pathogenesis and treatment. Signal Transduct Target Ther 2020; 5:136. [PMID: 32728023 PMCID: PMC7391753 DOI: 10.1038/s41392-020-00244-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/23/2020] [Indexed: 01/20/2023] Open
Affiliation(s)
- Kunming Zhao
- Shenzhen Graduate School of Harbin Institute of Technology, Shenzhen, 518055, China.,School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, HL, China
| | - Ying Hu
- Shenzhen Graduate School of Harbin Institute of Technology, Shenzhen, 518055, China. .,School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, HL, China.
| |
Collapse
|
737
|
Metaheuristics and Pontryagin's minimum principle for optimal therapeutic protocols in cancer immunotherapy: a case study and methods comparison. J Math Biol 2020; 81:691-723. [PMID: 32712711 DOI: 10.1007/s00285-020-01525-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 06/04/2020] [Indexed: 12/24/2022]
Abstract
In this paper, the performance appropriateness of population-based metaheuristics for immunotherapy protocols is investigated on a comparative basis while the goal is to stimulate the immune system to defend against cancer. For this purpose, genetic algorithm and particle swarm optimization are employed and compared with modern method of Pontryagin's minimum principle (PMP). To this end, a well-known mathematical model of cell-based cancer immunotherapy is described and examined to formulate the optimal control problem in which the objective is the annihilation of tumour cells by using the minimum amount of cultured immune cells. In this regard, the main aims are: (i) to introduce a single-objective optimization problem and to design the considered metaheuristics in order to appropriately deal with it; (ii) to use the PMP in order to obtain the necessary conditions for optimality, i.e. the governing boundary value problem; (iii) to measure the results obtained by using the proposed metaheuristics against those results obtained by using an indirect approach called forward-backward sweep method; and finally (iv) to produce a set of optimal treatment strategies by formulating the problem in a bi-objective form and demonstrating its advantages over single-objective optimization problem. A set of obtained results conforms the performance capabilities of the considered metaheuristics.
Collapse
|
738
|
Passaro A, Attili I, Morganti S, Del Signore E, Gianoncelli L, Spitaleri G, Stati V, Catania C, Curigliano G, de Marinis F. Clinical features affecting survival in metastatic NSCLC treated with immunotherapy: A critical review of published data. Cancer Treat Rev 2020; 89:102085. [PMID: 32771858 DOI: 10.1016/j.ctrv.2020.102085] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/26/2020] [Accepted: 07/16/2020] [Indexed: 12/18/2022]
Abstract
Immune checkpoint inhibitors (ICIs) represent one of the main steps forward for the treatment of advanced or metastatic non-small-cell lung cancer (NSCLC), without oncogenic driver alterations. Despite this recent progress, only a minority of patients achieve a broad and durable benefit and another proportion report poor survival and sometimes fast disease progression, confirming the need to optimise the patient's selection. To date, several issues are unsolved about how to personalise the immunotherapy treatment for individual patients. In this review, analysing data from pivotal randomised clinical trials (RCTs), we discuss patient baseline clinical and demographic features, including sex, age, ECOG performance status, smoking habit and specific site of metastases (liver, bone and brain) that may influence the efficacy outcomes in patients treated with ICIs. The high performance of the ICIs blurred the vision on different efficacy-limiting factors, which require extensive evaluation to improve the understanding ofthe tumour-specificimmune response, in which clinical drivers could be useful for better patient stratification.
Collapse
Affiliation(s)
- Antonio Passaro
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Ilaria Attili
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Stefania Morganti
- Division of Early Drug Development for Innovative Therapies, IEO European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Ester Del Signore
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Letizia Gianoncelli
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Gianluca Spitaleri
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Valeria Stati
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Catania
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Filippo de Marinis
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
739
|
Zhang L, Shi Y, Jenq RR, Do KA, Peterson CB. Bayesian compositional regression with structured priors for microbiome feature selection. Biometrics 2020; 77:824-838. [PMID: 32686846 DOI: 10.1111/biom.13335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 07/13/2020] [Indexed: 01/10/2023]
Abstract
The microbiome plays a critical role in human health and disease, and there is a strong scientific interest in linking specific features of the microbiome to clinical outcomes. There are key aspects of microbiome data, however, that limit the applicability of standard variable selection methods. In particular, the observed data are compositional, as the counts within each sample have a fixed-sum constraint. In addition, microbiome features, typically quantified as operational taxonomic units, often reflect microorganisms that are similar in function, and may therefore have a similar influence on the response variable. To address the challenges posed by these aspects of the data structure, we propose a variable selection technique with the following novel features: a generalized transformation and z-prior to handle the compositional constraint, and an Ising prior that encourages the joint selection of microbiome features that are closely related in terms of their genetic sequence similarity. We demonstrate that our proposed method outperforms existing penalized approaches for microbiome variable selection in both simulation and the analysis of real data exploring the relationship of the gut microbiome to body mass index.
Collapse
Affiliation(s)
- Liangliang Zhang
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yushu Shi
- Department of Statistics, University of Missouri, Columbia, Missouri
| | - Robert R Jenq
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kim-Anh Do
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christine B Peterson
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
740
|
Lei Q, Wang D, Sun K, Wang L, Zhang Y. Resistance Mechanisms of Anti-PD1/PDL1 Therapy in Solid Tumors. Front Cell Dev Biol 2020; 8:672. [PMID: 32793604 PMCID: PMC7385189 DOI: 10.3389/fcell.2020.00672] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022] Open
Abstract
In cancer-immunity cycle, the immune checkpoint PD1 and its ligand PDL1 act as accomplices to help tumors resist to immunity-induced apoptosis and promote tumor progression. Immunotherapy targeting PD1/PDL1 axis can effectively block its pro-tumor activity. Anti-PD1/PDL1 therapy has achieved great success in the past decade. However, only a subset of patients showed clinical responses. Most of the patients can not benefit from anti-PD1/PDL1 therapy. Furthermore, a large group of responders would develop acquired resistance after initial responses. Therefore, understanding the mechanisms of resistance is necessary for improving anti-PD1/PDL1 efficacy. Currently, researchers have identified primary resistance mechanisms which include insufficient tumor immunogenicity, disfunction of MHCs, irreversible T cell exhaustion, primary resistance to IFN-γ signaling, and immunosuppressive microenvironment. Some oncogenic signaling pathways also contribute to the primary resistance. Under the pressure applied by anti-PD1/PDL1 therapy, tumors experience immunoediting and preserve beneficial mutations, upregulate the compensatory inhibitory signaling and induce re-exhaustion of T cells, all of which may attenuate the durability of the therapy. Here we explore the underlying mechanisms in detail, review biomarkers that help identifying responders among patients and discuss the strategies that may relieve the anti-PD1/PDL1 resistance.
Collapse
Affiliation(s)
- Qingyang Lei
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Dan Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Kai Sun
- College of Medicine, Zhengzhou University, Zhengzhou, China
| | - Liping Wang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
741
|
Sharma VR, Singh M, Kumar V, Yadav M, Sehrawat N, Sharma DK, Sharma AK. Microbiome dysbiosis in cancer: Exploring therapeutic strategies to counter the disease. Semin Cancer Biol 2020; 70:61-70. [PMID: 32693015 DOI: 10.1016/j.semcancer.2020.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/11/2020] [Accepted: 07/11/2020] [Indexed: 02/09/2023]
Abstract
Cancer being a multiplex disease which involves many genomic and physiological alterations that occur consistently in the cancerous tissue, making the treatment and management of the disease even more complicated. The human gut microbiota (GM) harbors collective genomes of microbes comprising of trillions of bacteria along with fungi, archaea, and viruses that have the tendency to affect the development and progression of cancer. Moreover, inter-microbial interactions, diversity and distinct differences among the GM populations could influence the course of disease, making the microbiome an ideal target or to be modulated in such a way so as to improve cancer therapeutics with better efficacy and reduced toxicity. Current review focuses upon exploring the association of gut microbiota with the progression of cancer for which a structured search of bibliographic databases for peer-reviewed research literature has been carried out using focused review questions and inclusion/exclusion criteria. Through this review one could envisage a wide-spectrum role of microbiota in maintaining host metabolism, immune homeostasis paving the way for an anticancer diagnostic and therapeutic solution that has the potential to counter the menace of anti-cancer drug resistance as well.
Collapse
Affiliation(s)
- Var Ruchi Sharma
- Department of Biotechnology, Sri Guru Gobind Singh College Sector-26, Chandigarh UT, 160019, 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
| | - Mukesh Yadav
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala Haryana, 133207, India
| | - Nirmala Sehrawat
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala Haryana, 133207, India
| | | | - Anil K Sharma
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala Haryana, 133207, India.
| |
Collapse
|
742
|
Sims TT, Biegert GWG, Ramogola-Masire D, Ngoni K, Solley T, Ning MS, El Alam MB, Mezzari M, Petrosino J, Zetola NM, Schmeler KM, Colbert LE, Klopp AH, Grover S. Tumor microbial diversity and compositional differences among women in Botswana with high-grade cervical dysplasia and cervical cancer. Int J Gynecol Cancer 2020; 30:1151-1156. [PMID: 32675252 DOI: 10.1136/ijgc-2020-001547] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/17/2020] [Accepted: 06/22/2020] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION We characterized the cervical 16S rDNA microbiome of patients in Botswana with high-grade cervical dysplasia and locally advanced cervical cancer. METHODS This prospective study included 31 patients: 21 with dysplasia and 10 with cancer. The Shannon diversity index was used to evaluate alpha (intra-sample) diversity, while the UniFrac (weighted and unweighted) and Bray-Curtis distances were employed to evaluate beta (inter-sample) diversity. The relative abundance of microbial taxa was compared among samples using linear discriminant analysis effect size. RESULTS Alpha diversity was significantly higher in patients with cervical cancer than in patients with cervical dysplasia (P<0.05). Beta diversity also differed significantly (weighted UniFrac Bray-Curtis, P<0.01). Neither alpha diversity (P=0.8) nor beta diversity (P=0.19) varied by HIV status. The results of linear discriminant analysis effect size demonstrated that multiple taxa differed significantly between patients with cervical dysplasia vs cancer. Lachnospira bacteria (in the Clostridia class) were particularly enriched among cervical dysplasia patients, while Proteobacteria (members of the Firmicutes phyla and the Comamonadaceae family) were enriched in patients with cervical cancer. DISCUSSION The results of our study suggest that differences exist in the diversity and composition of the cervical microbiota between patients with cervical dysplasia and patients with cervical cancer in Botswana. Additional studies are warranted to validate these findings and elucidate their clinical significance among women living in sub-Saharan Africa, as well as other regions of the world.
Collapse
Affiliation(s)
- Travis T Sims
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Greyson W G Biegert
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Doreen Ramogola-Masire
- Obstetrics and Gynecology, Faculty of Medicine, University of Botswana, Gaborone, Botswana
| | - Kebatshabile Ngoni
- Department of Biological Sciences, University of Botswana, Gaborone, Botswana
| | - Travis Solley
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Matthew S Ning
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Molly B El Alam
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Melissa Mezzari
- Molecular Virology and Microbiology, Baylor College of Medicine Alkek Center for Molecular Discovery, Houston, Texas, USA
| | - Joseph Petrosino
- Molecular Virology and Microbiology, Baylor College of Medicine Alkek Center for Molecular Discovery, Houston, Texas, USA
| | - Nicola M Zetola
- Radiation Oncology, Botswana-University of Pennsylvania Partnership, Philadelphia, Pennsylvania, USA
| | - Kathleen M Schmeler
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren E Colbert
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ann H Klopp
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Surbhi Grover
- Radiation Oncology, Botswana-University of Pennsylvania Partnership, Philadelphia, Pennsylvania, USA
| |
Collapse
|
743
|
Zeng C, Tan H. Gut Microbiota and Heart, Vascular Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1238:107-141. [PMID: 32323183 DOI: 10.1007/978-981-15-2385-4_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gut microbiota plays an important role in maintaining human health. Accumulating evidence has indicated an intimate relationship between gut microbiota and cardiovascular diseases (CVD) which has become the leading cause of death worldwide. The alteration of gut microbial composition (gut dysbiosis) has been proven to contribute to atherosclerosis, the basic pathological process of CVD. In addition, the metabolites of gut microbiota have been found to be closely related to the development of CVD. For example, short-chain fatty acids are widely acclaimed beneficial effect against CVD, whereas trimethylamine-N-oxide is considered as a contributing factor in the development of CVD. In this chapter, we mainly discuss the gut microbial metabolite-involved mechanisms of CVD focusing on atherosclerosis, hypertension, diabetes, obesity, and heart failure. Targeting gut microbiota and related metabolites are novel and promising strategies for the treatment of CVD.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hongmei Tan
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
744
|
Li Y, Xiao H, Dong J, Luo D, Wang H, Zhang S, Zhu T, Zhu C, Cui M, Fan S. Gut Microbiota Metabolite Fights Against Dietary Polysorbate 80-Aggravated Radiation Enteritis. Front Microbiol 2020; 11:1450. [PMID: 32670255 PMCID: PMC7332576 DOI: 10.3389/fmicb.2020.01450] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy is a cornerstone of modern management methods for malignancies but is accompanied by diverse side effects. In the present study, we showed that food additives such as polysorbate 80 (P80) exacerbate irradiation-induced gastrointestinal (GI) tract toxicity. A 16S ribosomal RNA high-throughput sequencing analysis indicated that P80 consumption altered the abundance and composition of the gut microbiota, leading to severe radiation-induced GI tract injury. Mice harboring fecal microbes from P80-treated mice were highly susceptible to irradiation, and antibiotics-challenged mice also represented more sensitive to radiation following P80 treatment. Importantly, butyrate, a major metabolite of enteric microbial fermentation of dietary fibers, exhibited beneficial effects against P80 consumption-aggravated intestinal toxicity via the activation of G-protein-coupled receptors (GPCRs) and maintenance of the intestinal bacterial composition in irradiated animals. Moreover, butyrate had broad therapeutic effects on common radiation-induced injury. Collectively, our findings demonstrate that P80 are potential risk factors for cancer patients during radiotherapy and indicate that butyrate might be employed as a therapeutic option to mitigate the complications associated with radiotherapy.
Collapse
Affiliation(s)
- Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huiwen Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Dan Luo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Haichao Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.,Department of Emergency Medicine, North Shore University Hospital, Manhasset, NY, United States.,Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Shuqin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tong Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Changchun Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
745
|
Abstract
The “Right-to-Try” experimental drugs act passed by Donald Trump in 2018 provides an opportunity of early access to experimental drugs for the treatment of life-threatening diseases and a potential boon to many young and under-capitalized biotechnology or pharmaceutical companies. The pros and cons of experimental drugs, including a number of “cutting edge” scientific, clinical, and a number of synergistic approaches such as artificial intelligence, machine learning, big data, data refineries, electronic health records, data driven clinical decisions and risk mitigation are reviewed.
Collapse
|
746
|
Brassart-Pasco S, Dalstein V, Brassart B, Dewolf M, Clavel C, Oudart JB. Immunotherapy in non-small-cell lung cancer: from targeted molecules to resistance patterns. Pharmacogenomics 2020; 21:705-720. [PMID: 32567537 DOI: 10.2217/pgs-2020-0021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Immunotherapies are now considered as a pillar of non-small-cell lung cancer treatment. The main targets of immune-checkpoint inhibitors (ICI) are programmed cell death 1/programmed cell death ligand 1 and cytotoxic T-lymphocyte antigen 4, aiming at restoring antitumor immunity. Despite durable responses observed in some patients, all patients do not benefit from the treatment and almost all responders ultimately relapse after some time. In this review, we discuss the biomarkers that could be used to predict response to ICI, the current indications of ICI in non-small-cell lung cancer, the mechanisms inducing tumor-cell intrinsic or extrinsic resistance to ICI and finally, the potential treatment response monitoring.
Collapse
Affiliation(s)
- Sylvie Brassart-Pasco
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France
| | - Véronique Dalstein
- Université de Reims Champagne Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, 51100 Reims, France.,CHU Reims, Service de Pathologie, 51100 Reims, France
| | - Bertrand Brassart
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France
| | - Maxime Dewolf
- CHU Reims, Service des maladies respiratoires et allergiques, 51100 Reims, France
| | - Christine Clavel
- Université de Reims Champagne Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, 51100 Reims, France.,CHU Reims, Service de Pathologie, 51100 Reims, France
| | - Jean-Baptiste Oudart
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, 51100 Reims, France
| |
Collapse
|
747
|
Emerging role of microbiota in immunomodulation and cancer immunotherapy. Semin Cancer Biol 2020; 70:37-52. [PMID: 32580024 DOI: 10.1016/j.semcancer.2020.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 02/08/2023]
Abstract
Gut microbiota is emerging as a key modulator of the immune system. Alteration of gut microbiota impacts functioning of the immune system and pathophysiology of several diseases, including cancer. Growing evidence indicates that gut microbiota is not only involved in carcinogenesis but also has an impact on the efficacy and toxicity of cancer therapy. Recently, several pre-clinical and clinical studies across diverse cancer types reported the influence of gut microbiota on the host immune response to immunotherapy. Advancement in our understanding of the mechanism behind microbiota-mediated modulation of immune response is paramount for their utilization as cancer therapeutics. These microbial therapies in combination with conventional immunotherapeutic methods have the potential to transform the pre-existing treatment strategies to personalized cancer therapy. In this review, we have summarized the current status of research in the field and discussed the role of microbiota as an immune system modulator in context of cancer and their impact on immunotherapy.
Collapse
|
748
|
Inamura K. Gut microbiota contributes towards immunomodulation against cancer: New frontiers in precision cancer therapeutics. Semin Cancer Biol 2020; 70:11-23. [PMID: 32580023 DOI: 10.1016/j.semcancer.2020.06.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 02/08/2023]
Abstract
The microbiota influences human health and the development of diverse diseases, including cancer. Microbes can influence tumor initiation and development in either a positive or negative manner. In addition, the composition of the gut microbiota affects the efficacy and toxicity of cancer therapeutics as well as therapeutic resistance. The striking impact of microbiota on oncogenesis and cancer therapy provides compelling evidence to support the notion that manipulating microbial networks represents a promising strategy for treating and preventing cancer. Specific microbes or the microbial ecosystem can be modified via a multiplicity of processes, and therapeutic methods and approaches have been evolving. Microbial manipulation can be applied as an adjunct to traditional cancer therapies such as chemotherapy and immunotherapy. Furthermore, this approach displays great promise as a stand-alone therapy following the failure of standard therapy. Moreover, such strategies may also benefit patients by avoiding the emergence of toxic side effects that result in treatment discontinuation. A better understanding of the host-microbial ecosystem in patients with cancer, together with the development of methodologies for manipulating the microbiome, will help expand the frontiers of precision cancer therapeutics, thereby improving patient care. This review discusses the roles of the microbiota in oncogenesis and cancer therapy, with a focus on efforts to harness the microbiota to fight cancer.
Collapse
Affiliation(s)
- Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| |
Collapse
|
749
|
Thalanayar Muthukrishnan P, Faillace R. Compassionate use of others' immunity - understanding gut microbiome in Covid-19. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:358. [PMID: 32552848 PMCID: PMC7301059 DOI: 10.1186/s13054-020-03043-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/27/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Prashanth Thalanayar Muthukrishnan
- Pulmonary and Critical Care Medicine, Department of Medicine, Albert Einstein College of Medicine, NYC HHC, Jacobi Medical Center and North Central Bronx Hospital, 1400 Pelham Parkway S, Bronx, NY, 10461, USA.
| | - Robert Faillace
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, NYC HHC, 1400 Pelham Parkway S, Bronx, NY, 10461, USA
| |
Collapse
|
750
|
Gabuzda D, Jamieson BD, Collman RG, Lederman MM, Burdo TH, Deeks SG, Dittmer DP, Fox HS, Funderburg NT, Pahwa SG, Pandrea I, Wilson CC, Hunt PW. Pathogenesis of Aging and Age-related Comorbidities in People with HIV: Highlights from the HIV ACTION Workshop. Pathog Immun 2020; 5:143-174. [PMID: 32856008 PMCID: PMC7449259 DOI: 10.20411/pai.v5i1.365] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
People with HIV (PWH) experience accentuated biological aging, as defined by markers of inflammation, immune dysfunction, and the epigenetic clock. They also have an elevated risk of multiple age-associated comorbidities. To discuss current knowledge, research gaps, and priorities in aging and age-related comorbidities in treated HIV infection, the NIH program staff organized a workshop held in Bethesda, Maryland in September 2019. This review article describes highlights of discussions led by the Pathogenesis/Basic Science Research working group that focused on three high priority topics: immunopathogenesis; the microbiome/virome; and aging and senescence. We summarize knowledge in these fields and describe key questions for research on the pathogenesis of aging and age-related comorbidities in PWH. Understanding the drivers and mechanisms underlying accentuated biological aging is a high priority that will help identify potential therapeutic targets to improve healthspan in older PWH.
Collapse
Affiliation(s)
- Dana Gabuzda
- Department of Cancer Immunology and Virology; Dana-Farber Cancer Institute; Boston, Massachusetts; Department of Neurology; Harvard Medical School; Boston, Massachusetts
| | - Beth D Jamieson
- Department of Medicine; David Geffen School of Medicine; University of California; Los Angeles, California
| | - Ronald G Collman
- Department of Medicine; University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania
| | - Michael M Lederman
- Department of Medicine; Case Western Reserve University School of Medicine; Cleveland, Ohio
| | - Tricia H Burdo
- Department of Neuroscience; Lewis Katz School of Medicine; Temple University; Philadelphia, Pennsylvania
| | - Steven G Deeks
- Department of Medicine; University of California; San Francisco, California
| | - Dirk P Dittmer
- Department of Microbiology and Immunology; University of North Carolina School of Medicine; Chapel Hill, North Carolina
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience; University of Nebraska Medical Center; Omaha, Nebraska
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science; School of Health and Rehabilitation Sciences; Ohio State University College of Medicine; Columbus, Ohio
| | - Savita G Pahwa
- Department of Microbiology and Immunology; University of Miami Miller School of Medicine; Miami, Florida
| | - Ivona Pandrea
- Department of Microbiology and Molecular Genetics; School of Medicine; University of Pittsburgh; Pittsburgh, Pennsylvania
| | - Cara C Wilson
- Department of Medicine; Division of Infectious Diseases; University of Colorado Anschutz Medical Campus; Aurora, Colorado
| | - Peter W Hunt
- Department of Medicine; University of California; San Francisco, California
| |
Collapse
|