51
|
Khan I. Microbiome. Indian J Med Paediatr Oncol 2021. [DOI: 10.1055/s-0041-1735599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Imran Khan
- Department of Medical Oncology, Artemis Hospitals, Gurugram, Haryana, India
| |
Collapse
|
52
|
de Souza JB, Brelaz-de-Castro MCA, Cavalcanti IMF. Strategies for the treatment of colorectal cancer caused by gut microbiota. Life Sci 2021; 290:120202. [PMID: 34896161 DOI: 10.1016/j.lfs.2021.120202] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC), also named as colon and rectal or bowel cancer, is one of the leading neoplasia diagnosed in the world. Genetic sequencing studies of microorganisms from the intestinal microbiota of patients with CRC revealed that changes in its composition occur with the development of the disease, which can play a fundamental role in its development, being mediated by the production of metabolites and toxins that damage enterocytes. Some microorganisms are frequently reported in the literature as the main agents of this process, such as the bacteria Fusobacterium nucleatum, Escherichia coli and Bacteroides fragilis. Thus, understanding the mechanisms and function of each microorganism in CRC is essential for the development of treatment tools that focus on the gut microbiota. This review verifies current research aimed at evaluating the microorganisms present in the microbiota that can influence the development of CRC, as well as possible forms of treatment that can prevent the initiation and/or spread of this disease. Due to the incidence of CRC, alternatives have been launched considering factors beyond those already known in the disease development, such as diet, fecal microbiota transplantation, use of probiotics and antibiotics, which have been widely studied for this purpose. However, despite being promising, the studies that focus on the development of new therapeutic approaches targeting the microorganisms that cause CRC still need to be improved and better developed, involving new techniques to elucidate the effectiveness and safety of these new methods.
Collapse
Affiliation(s)
- Jaqueline Barbosa de Souza
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | | | - Isabella Macário Ferro Cavalcanti
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Recife, PE, Brazil; Laboratory of Microbiology and Immunology, Academic Center of Vitória (CAV), Federal University of Pernambuco (UFPE), Vitória de Santo Antão, PE, Brazil.
| |
Collapse
|
53
|
Huang X, Li M, Hou S, Tian B. Role of the microbiome in systemic therapy for pancreatic ductal adenocarcinoma (Review). Int J Oncol 2021; 59:101. [PMID: 34738624 PMCID: PMC8577795 DOI: 10.3892/ijo.2021.5281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/18/2021] [Indexed: 02/05/2023] Open
Abstract
A large body of evidence has revealed that the microbiome serves a role in all aspects of cancer, particularly cancer treatment. To date, studies investigating the relationship between the microbiome and systemic therapy for pancreatic ductal adenocarcinoma (PDAC) are lacking. PDAC is a high‑mortality malignancy (5‑year survival rate; <9% for all stages). Systemic therapy is one of the most important treatment choices for all patients; however, resistance or toxicity can affect its efficacy. Studies have supported the hypothesis that the microbiome is closely associated with the response to systemic therapy in PDAC, including the induction of drug resistance, or toxicity and therapy‑related changes in microbiota composition. The present review comprehensively summarized the role of the microbiome in systemic therapy for PDAC and the associated molecular mechanisms in an attempt to provide a novel direction for the improvement of treatment response and proposed potential directions for in‑depth research.
Collapse
Affiliation(s)
| | | | - Shengzhong Hou
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Bole Tian
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
54
|
Tonini V, Zanni M. Pancreatic cancer in 2021: What you need to know to win. World J Gastroenterol 2021; 27:5851-5889. [PMID: 34629806 PMCID: PMC8475010 DOI: 10.3748/wjg.v27.i35.5851] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/14/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the solid tumors with the worst prognosis. Five-year survival rate is less than 10%. Surgical resection is the only potentially curative treatment, but the tumor is often diagnosed at an advanced stage of the disease and surgery could be performed in a very limited number of patients. Moreover, surgery is still associated with high post-operative morbidity, while other therapies still offer very disappointing results. This article reviews every aspect of pancreatic cancer, focusing on the elements that can improve prognosis. It was written with the aim of describing everything you need to know in 2021 in order to face this difficult challenge.
Collapse
Affiliation(s)
- Valeria Tonini
- Department of Medical Sciences and Surgery, University of Bologna- Emergency Surgery Unit, IRCCS Sant’Orsola Hospital, Bologna 40121, Italy
| | - Manuel Zanni
- University of Bologna, Emergency Surgery Unit, IRCCS Sant'Orsola Hospital, Bologna 40121, Italy
| |
Collapse
|
55
|
Dalal P, Sharma D. Microbe defines the efficacy of chemotherapeutic drug: a complete paradigm. FEMS Microbiol Lett 2021; 368:6358522. [PMID: 34448860 DOI: 10.1093/femsle/fnab116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
The human body harbors a diverse microbiome that regulates host physiology and disease development. Several studies have also been reported where the human microbiome interferes with the efficacy of chemotherapeutics. Reports have also suggested the use of microbes in specific targeting and drug delivery. This review mainly focuses on the alteration in the efficacy of the drug by human microbiota. We have also discussed how the diversity in microbes can determine the therapeutic outcomes of a particular drug. The pathways involved in the alteration are also focused, with some highlights on microbes being used in cancer therapy.
Collapse
Affiliation(s)
- P Dalal
- Institute of Nanoscience and Technology, Knowledge City, Sector - 81, Mohali 140306, Punjab, India
| | - D Sharma
- Institute of Nanoscience and Technology, Knowledge City, Sector - 81, Mohali 140306, Punjab, India
| |
Collapse
|
56
|
Nishiumi F, Kawai Y, Nakura Y, Yoshimura M, Wu HN, Hamaguchi M, Kakizawa S, Suzuki Y, Glass JI, Yanagihara I. Blockade of endoplasmic reticulum stress-induced cell death by Ureaplasma parvum vacuolating factor. Cell Microbiol 2021; 23:e13392. [PMID: 34490709 DOI: 10.1111/cmi.13392] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/03/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022]
Abstract
Previously, we found that Ureaplasma parvum internalised into HeLa cells and cytosolic accumulation of galectin-3. U. parvum induced the host cellular membrane damage and survived there. Here, we conducted vesicular trafficking inhibitory screening in yeast to identify U. parvum vacuolating factor (UpVF). U. parvum triggered endoplasmic reticulum (ER) stress and upregulated the unfolded protein response-related factors, including BiP, P-eIF2 and IRE1 in the host cells, but it blocked the induction of the downstream apoptotic factors. MicroRNA library screening of U. parvum-infected cells and UpVF-transfected cells identified miR-211 and miR-214 as the negative regulators of the apoptotic cascade under ER stress. Transient expression of UpVF induced HeLa cell death with intracellular vacuolization; however, some stable UpVF transformant survived. U. parvum-infected cervical cell lines showed resistance to actinomycin D, and UpVF stable transformant cell lines exhibited resistance to X-ray irradiation, as well as cisplatin and paclitaxel. UpVF expressing cervical cancer xenografts in nude mice also acquired resistance to cisplatin and paclitaxel. A mycoplasma expression vector based on Mycoplasma mycoides, Syn-MBA (multiple banded antigen)-UpVF, reduced HeLa cell survival compared with that of Syn-MBA after 72 hr of infection. These findings together suggest novel mechanisms for Ureaplasma infection and the possible implications for cervical cancer malignancy. TAKE AWAYS: • Ureaplasmal novel virulence factor, UpVF, was identified. • UpVF triggered ER stress but suppressed apoptotic cascade via miR-211 and -214. • UpVF conferred resistance to anticancer treatments both in vivo and in vitro. • Dual expression of MBA and UpVF in JCVI-syn3B showed host cell damage.
Collapse
Affiliation(s)
- Fumiko Nishiumi
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan
| | - Yasuhiro Kawai
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan.,Health Evaluation Center, Kanazawa Medical University Himi Municipal Hospital, Toyama, Japan
| | - Yukiko Nakura
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan
| | - Michinobu Yoshimura
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan.,Department of Microbiology and Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Heng Ning Wu
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan
| | - Mitsuhide Hamaguchi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Emergency and Critical Care Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Shigeyuki Kakizawa
- Bioproduction Research Institute,, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Yo Suzuki
- Synthetic Biology Group, J. Craig Venter Institute, La Jolla, California, USA
| | - John I Glass
- Synthetic Biology Group, J. Craig Venter Institute, La Jolla, California, USA
| | - Itaru Yanagihara
- Department of Developmental Medicine, Research Institute, Women's and Children's Hospital, Osaka, Japan
| |
Collapse
|
57
|
Patki M, Saraswat A, Bhutkar S, Dukhande V, Patel K. In vitro assessment of a synergistic combination of gemcitabine and zebularine in pancreatic cancer cells. Exp Cell Res 2021; 405:112660. [PMID: 34048785 DOI: 10.1016/j.yexcr.2021.112660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/13/2021] [Accepted: 05/12/2021] [Indexed: 01/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers with an extremely poor prognosis. Gemcitabine (Gem) is still the mainstay drug for the treatment of PDAC. However, rapid inactivation by cytidine deaminase (CDA) present in pancreatic cancer cells severely limits anticancer efficacy of Gem. In this study, we investigated the effect of a CDA inhibitor - Zebularine (Zeb) on anticancer activity of Gem in pancreatic cancer cell lines MiaPaCa-2, BxPC-3, and Panc-1. Zeb treatment synergistically increased Gem-induced cytotoxicity in all three pancreatic cancer cell lines. The strongest synergistic activity was found at 1:10 M ratio of Gem/Zeb (combination index 0.04-0.4). Additionally, Gem + Zeb treated cells showed marked decreased in the expressions of anti-apoptotic protein including Bcl-2 and survivin while significantly increased the cleaved caspase-3, and loss of mitochondrial membrane potential was observed. Multicellular 3D spheroids of MiaPaCa-2 cells treated with combination showed significant reduction (25-60%) in spheroid size, weight compared to single drug and control group. Live/dead cell imaging showed that Gem + Zeb treated spheroids exhibited a highly distorted surface with significantly higher number of dead cells (red). The results of the present study confirm that this synergistic combination is worthy of future investigations as a potential approach for the treatment of PDAC.
Collapse
Affiliation(s)
- Manali Patki
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | | | - Shraddha Bhutkar
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Vikas Dukhande
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, NY, USA.
| |
Collapse
|
58
|
Jujube Powder Enhances Cyclophosphamide Efficiency against Murine Colon Cancer by Enriching CD8 + T Cells While Inhibiting Eosinophilia. Nutrients 2021; 13:nu13082700. [PMID: 34444860 PMCID: PMC8401958 DOI: 10.3390/nu13082700] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclophosphamide (CTX) is widely applied in cancer treatment. However, the outcome is often compromised by lymphopenia, myelosuppression, and gut dysbiosis. Here, we used jujube powder to enhance CTX efficiency through nurturing gut microbiota in order to facilitate favorable metabolisms. It was observed that the oral administration of jujube powder enriched CD8+ T cells in mouse MC38 colon tumor microenvironment and increased the diversity of gut microbiota and the abundance of Bifidobacteriales, which is helpful to the production of butyrate in the cecum content. The application of jujube powder also stimulated the production of white blood cells, especially CD8+ T cells in peripheral and bone marrow, while inhibiting the growth of eosinophils in peripheral blood and the production of IL-7 and GM-CSF in serum. All these are conductive to the significant inhibition of the tumor growth, suggesting the high potential of nurturing gut microbiota with natural products for improving the efficiency of chemotherapy.
Collapse
|
59
|
Lu SY, Hua J, Xu J, Wei MY, Liang C, Meng QC, Liu J, Zhang B, Wang W, Yu XJ, Shi S. Microorganisms in chemotherapy for pancreatic cancer: An overview of current research and future directions. Int J Biol Sci 2021; 17:2666-2682. [PMID: 34326701 PMCID: PMC8315022 DOI: 10.7150/ijbs.59117] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/08/2021] [Indexed: 01/18/2023] Open
Abstract
Pancreatic cancer is a malignant tumor of the digestive system with a very high mortality rate. While gemcitabine-based chemotherapy is the predominant treatment for terminal pancreatic cancer, its therapeutic effect is not satisfactory. Recently, many studies have found that microorganisms not only play a consequential role in the occurrence and progression of pancreatic cancer but also modulate the effect of chemotherapy to some extent. Moreover, microorganisms may become an important biomarker for predicting pancreatic carcinogenesis and detecting the prognosis of pancreatic cancer. However, the existing experimental literature is not sufficient or convincing. Therefore, further exploration and experiments are imperative to understanding the mechanism underlying the interaction between microorganisms and pancreatic cancer. In this review, we primarily summarize and discuss the influences of oncolytic viruses and bacteria on pancreatic cancer chemotherapy because these are the two types of microorganisms that are most often studied. We focus on some potential methods specific to these two types of microorganisms that can be used to improve the efficacy of chemotherapy in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Si-Yuan Lu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Miao-Yan Wei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Qing-Cai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| |
Collapse
|
60
|
Prasanthan P, Kishore N. Self-assemblies of pluronic micelles in partitioning of anticancer drugs and effectiveness of this system towards target protein. RSC Adv 2021; 11:22057-22069. [PMID: 35480822 PMCID: PMC9034178 DOI: 10.1039/d1ra03770f] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/17/2021] [Indexed: 12/24/2022] Open
Abstract
Micelles formed by pluronic triblock copolymers are known to be a promising class of drug delivery vehicles. Quantitative mechanistic insights into the ability of pluronic micelles to improve the solubility of poorly water soluble drugs, encapsulation and delivery of hydrophilic drugs are not available. The current study evaluated the energetics of encapsulation of chemotherapeutic drugs gemcitabine, cytarabine, and hydroxyurea in pluronic F127 and F68 micelles. In addition, the interactions of the drugs released from pluronic micellar media with serum albumin, which is a major circulatory transport protein, and subsequent conformational changes have also been analyzed with the help of calorimetry and spectroscopy. All the drugs showed improved partitioning in F127 micelles, the extent of which slightly increased with temperature rise. Interestingly, drug–protein binding is enhanced upon delivery from pluronic micelles without affecting the conformational integrity of the protein. This study highlights the role of drug functionalities, hydrophobicity, and steric factors towards their partitioning in pluronic micelles. Such studies are important in understanding physicochemical aspects of drug encapsulation and release, and lead to establishing structure–property–energetics correlations for developing suitable nano-drug delivery vehicles. Micelles formed by pluronic triblock copolymers are known to be a promising class of drug delivery vehicles.![]()
Collapse
Affiliation(s)
- Pooja Prasanthan
- Department of Chemistry, Indian Institute of Technology Bombay Powai Mumbai 400 076 India
| | - Nand Kishore
- Department of Chemistry, Indian Institute of Technology Bombay Powai Mumbai 400 076 India
| |
Collapse
|
61
|
Approaching precision medicine by tailoring the microbiota. Mamm Genome 2021; 32:206-222. [PMID: 33646347 DOI: 10.1007/s00335-021-09859-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/10/2021] [Indexed: 10/22/2022]
Abstract
Accumulating evidence has revealed the link between the microbiota and various human diseases. Advances in high-throughput sequencing technologies have identified some consistent disease-associated microbial features, leading to the emerging concept of microbiome-based therapeutics. However, it is also becoming clear that there are considerable variations in the microbiota among patients with the same disease. Variations in the microbial composition and function contribute to substantial differences in metabolic status of the host via production of a myriad of biochemically and functionally different microbial metabolites. Indeed, compelling evidence indicates that individuality of the microbiome may result in individualized responses to microbiome-based therapeutics and other interventions. Mechanistic understanding of the role of the microbiota in diseases and drug metabolism would help us to identify causal relationships and thus guide the development of microbiome-based precision or personalized medicine. In this review, we provide an overview of current efforts to use microbiome-based interventions for the treatment of diseases such as cancer, neurological disorders, and diabetes to approach precision medicine.
Collapse
|
62
|
Lopes BRP, Ribeiro AG, Silva TF, Barbosa LV, Jesus TI, Matsuda BK, Costa MF, Toledo KA. Diagnosis and treatment of HEp-2 cells contaminated with mycoplasma. BRAZ J BIOL 2021; 81:37-43. [PMID: 32321065 DOI: 10.1590/1519-6984.215721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 08/20/2019] [Indexed: 11/22/2022] Open
Abstract
Contamination of primary and cell cultures by mycoplasmas is one of the main economic and biological pitfalls in basic research, diagnosis and manufacture of biotechnological products. It is a common issue which may be difficult to conduct surveillance on. Mycoplasma presence may affect several physiological parameters of the cell, besides being considered an important source of inaccurate and/or non-reproducible scientific results. Each cell type presents characteristical symptoms, mainly morphological, that indicate a contamination by mycoplasma. HEp-2 cells originate from carcinoma of the larynx and are, therefore, part of the respiratory tract, which is one of mycoplasma habitats. Despite the importance these cells in several biological research (evaluation of cell proliferation and migration, apoptosis, antiviral and antitumor compounds), the alterations induced by mycoplasma contamination in HEp-2 cells have not yet been described. Here, we describe the progressive morphological alterations in culture of HEp-2 cells infected with mycoplasma, as well as the-diagnosis of the infection and its treatment. Mycoplasma contamination described within this work led to cytoplasm elongation, cell-to-cell spacing, thin plasma membrane projections, cytoplasmic vacuoles, fusion with neighboring cells, and, finally, cell death. Contamination was detected by fluorescence imaging (DAPI) and PCR reactions. The cultures were treated with BM-Cyclin antibiotic to eliminate contamination. The data presented here will be of relevance to researchers whose investigations involve cell culture, especially respiratory and HEp-2 cells.
Collapse
Affiliation(s)
- B R P Lopes
- Departamento de Ciências Biológicas, Faculdade de Ciências e Letras, Universidade Estadual Paulista - UNESP, Av. Dom Antônio, 2100, Parque Universitário, CEP 19806-900, Assis, SP, Brasil.,Instituto de Biociências, Letras e Ciências Exatas - IBILCE, Universidade Estadual Paulista - UNESP, R. Cristovão Colombo, 2265, Jardim Nazareth, CEP 15054-000, São José do Rio Preto, SP, Brasil
| | - A G Ribeiro
- Departamento de Ciências Biológicas, Faculdade de Ciências e Letras, Universidade Estadual Paulista - UNESP, Av. Dom Antônio, 2100, Parque Universitário, CEP 19806-900, Assis, SP, Brasil.,Instituto de Biociências, Letras e Ciências Exatas - IBILCE, Universidade Estadual Paulista - UNESP, R. Cristovão Colombo, 2265, Jardim Nazareth, CEP 15054-000, São José do Rio Preto, SP, Brasil
| | - T F Silva
- Departamento de Ciências Biológicas, Faculdade de Ciências e Letras, Universidade Estadual Paulista - UNESP, Av. Dom Antônio, 2100, Parque Universitário, CEP 19806-900, Assis, SP, Brasil
| | - L V Barbosa
- Departamento de Ciências Biológicas, Faculdade de Ciências e Letras, Universidade Estadual Paulista - UNESP, Av. Dom Antônio, 2100, Parque Universitário, CEP 19806-900, Assis, SP, Brasil
| | - T I Jesus
- Departamento de Ciências Biológicas, Faculdade de Ciências e Letras, Universidade Estadual Paulista - UNESP, Av. Dom Antônio, 2100, Parque Universitário, CEP 19806-900, Assis, SP, Brasil
| | - B K Matsuda
- Departamento de Ciências Biológicas, Faculdade de Ciências e Letras, Universidade Estadual Paulista - UNESP, Av. Dom Antônio, 2100, Parque Universitário, CEP 19806-900, Assis, SP, Brasil
| | - M F Costa
- Departamento de Ciências Biológicas, Faculdade de Ciências e Letras, Universidade Estadual Paulista - UNESP, Av. Dom Antônio, 2100, Parque Universitário, CEP 19806-900, Assis, SP, Brasil
| | - K A Toledo
- Departamento de Ciências Biológicas, Faculdade de Ciências e Letras, Universidade Estadual Paulista - UNESP, Av. Dom Antônio, 2100, Parque Universitário, CEP 19806-900, Assis, SP, Brasil.,Instituto de Biociências, Letras e Ciências Exatas - IBILCE, Universidade Estadual Paulista - UNESP, R. Cristovão Colombo, 2265, Jardim Nazareth, CEP 15054-000, São José do Rio Preto, SP, Brasil
| |
Collapse
|
63
|
Dai Y, Zhong F, Liu W, Song Q, Hu W. Mycoplasma hyorhinis infection promotes tyrosine kinase inhibitor (TKI) resistance in lung adenocarcinoma patients. J Cancer Res Clin Oncol 2021; 147:1379-1388. [PMID: 33550434 DOI: 10.1007/s00432-021-03547-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/27/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE To explore the relationship between Mycoplasma hyorhinis infection and tyrosine kinase inhibitor (TKI) resistance in lung adenocarcinoma patients. METHODS Mycoplasma hyorhinis infection can be verified with the monoclonal antibody PD4, which specifically recognizes a distinct protein of M. hyorhinis. Immunohistochemistry (IHC), using PD4 to detect M. hyorhinis, was performed on paraffin-embedded lung adenocarcinoma tissues of patients who had epidermal growth factor (EGFR) mutations and had received oral TKI. The number of patients enrolled in our study was 101. Assessments following TKI treatment were performed until objective disease progression or stable disease at the cutoff date was reached. In all of the patients, the primary endpoint was investigator-assessed progression-free survival (PFS). RESULTS Immunohistochemistry revealed that 61 of 101 cases (60.4%) of lung adenocarcinoma were positive for M. hyorhinis, which comprised of 31 low-positive cases and 30 high-positive cases; the remaining 40 cases (39.6%) were negative. The median PFS was significantly longer in the negative group [18 months (95% CI 14.15-21.85)] than in the low-positive group [10 months (95% CI 7.70-12.30); hazard ratio (HR) 4.095, 95% CI 2.254-7.438; p < 0.001] and in the high-positive group [4 months (95% CI 2.85-5.15); HR 31.703, 95% CI 14.425-69.678; p < 0.001]. The results of the subgroup analysis were satisfactory. The PFS benefit with negative M. hyorhinis infection was consistent across subgroups. CONCLUSIONS In this retrospective, exploratory analysis, M. hyorhinis infection significantly reduced PFS. With increased levels of M. hyorhinis infection, the progression of the disease was more advanced, likely due to the hydrolysis of TKI by M. hyorhinis. A strong correlation was found between M. hyorhinis infection and TKI resistance in lung adenocarcinoma. This study provides potent evidence that M. hyorhinis hydrolyses TKI and will assist in the research of related mechanisms in the future. IMPLICATIONS FOR CANCER SURVIVORS It provides an option to improve the efficacy of TKI, including strategies to decrease M. hyorhinis infection, thereby reducing long-term distress in TKI resistance patients with EGFR mutations.
Collapse
Affiliation(s)
- Yueyu Dai
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Fangyuan Zhong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Wenbin Liu
- College of Health Sciences and Nursing, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China.
| | - Weiguo Hu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
64
|
Gastrointestinal cancers: the role of microbiota in carcinogenesis and the role of probiotics and microbiota in anti-cancer therapy efficacy. Cent Eur J Immunol 2021; 45:476-487. [PMID: 33658894 PMCID: PMC7882408 DOI: 10.5114/ceji.2020.103353] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023] Open
Abstract
The gut epithelium is a habitat of a variety of microorganisms, including bacteria, fungi, viruses and Archaea. With the advent of sophisticated molecular techniques and bioinformatics tools, more information on the composition and thus function of gut microbiota was revealed. The gut microbiota as an integral part of the intestinal barrier has been shown to be involved in shaping the mucosal innate and adaptive immune response and to provide protection against pathogens. Consequently, a set of biochemical signals exchanged within microbes and communication between the microbiota and the host have opened a new way of thinking about cancer biology. Probiotics are living organisms which administered in adequate amounts may bring health benefits and have the potential to be an integral part of the prevention/treatment strategies in clinical approaches. Here we provide a comprehensive review of data linking gut microbiota to cancer pathogenesis and its clinical course. We focus on gastrointestinal cancers, such as gastric, colorectal, pancreatic and liver cancer.
Collapse
|
65
|
Bossù G, Di Sario R, Argentiero A, Esposito S. Antimicrobial Prophylaxis and Modifications of the Gut Microbiota in Children with Cancer. Antibiotics (Basel) 2021; 10:antibiotics10020152. [PMID: 33546312 PMCID: PMC7913491 DOI: 10.3390/antibiotics10020152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
In children with cancer, chemotherapy can produce cytotoxic effects, resulting in immunosuppression and an augmented risk of febrile neutropenia and bloodstream infections. This has led to widespread use of antibiotic prophylaxis which, combined with intensive chemotherapy treatment, could have a long-term effect on the gastrointestinal microbiome. In this review, we aimed to analyze the current literature about the widespread use of antibiotic prophylaxis in children experiencing infectious complications induced by chemotherapy and its effects on the gut microbiome. Our review of the literature shows that antimicrobial prophylaxis in children with cancer is still a trending topic and, at the moment, there are not enough data to define universal guidelines. Children with cancer experience long and painful medical treatments and side effects, which are associated with great economic and social burdens, important psychological consequences, and dysbiosis induced by antibiotics and also by chemotherapy. Considering the importance of a healthy gut microbiota, studies are needed to understand the impact of dysbiosis in response to therapy in these children and to define how to modulate the microbiome to favor a positive therapeutic outcome.
Collapse
|
66
|
Joukar F, Mavaddati S, Mansour-Ghanaei F, Samadani AA. Gut Microbiota as a Positive Potential Therapeutic Factor in Carcinogenesis: an Overview of Microbiota-Targeted Therapy. J Gastrointest Cancer 2021; 51:363-378. [PMID: 31025167 DOI: 10.1007/s12029-019-00237-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer therapeutic methods comprising chemotherapy, radiotherapy, and surgery are so routine in cancer treatment. Remarkably, there are several personal features which affect the effectiveness of such treatments including nutrition, microbiome diversity, and physical activity which has distinct significant roles during and after therapies along with their bilateral connections. In this way, the ability of gut microbiota36 in modulating the efficacy of chemotherapeutic medications in cancer and other types of disorders is of great importance. In addition, the role of dietary, probiotic, and synthetically engineered bacteria in manipulating and optimizing the gut microbiota is of interest. Conspicuously, the correlation between the commensal microbiota and also host can regulate the physiological activities comprising the immunity system and inflammatory agents and it is scanned in the category of cancers. Bacterial species have been employed in cancer therapy; commensal microbes posse a key beneficial role in this field. Practically, the microbiota has this potential to accelerate and modulates a certain response by priming in order to release the pro-inflammatory agents. We would like to discuss these vital factors in this review as gut microbiota has the potential to be the main option for personalized cancer treatment strategies in the future. Meaning, this novel data present clinical promising feasibilities of modulating cancer therapy with using microbiota.
Collapse
Affiliation(s)
- Farahnaz Joukar
- GI Cancer Screening and Prevention Research Center, Guilan University of Medical Sciences, Rasht, Iran.,Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Sara Mavaddati
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.,Caspian Digestive Disease Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Fariborz Mansour-Ghanaei
- GI Cancer Screening and Prevention Research Center, Guilan University of Medical Sciences, Rasht, Iran.,Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Akbar Samadani
- GI Cancer Screening and Prevention Research Center, Guilan University of Medical Sciences, Rasht, Iran. .,Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
67
|
Long Y, Luo J. Association Mining to Identify Microbe Drug Interactions Based on Heterogeneous Network Embedding Representation. IEEE J Biomed Health Inform 2021; 25:266-275. [PMID: 32750918 DOI: 10.1109/jbhi.2020.2998906] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Accurately identifying microbe-drug associations plays a critical role in drug development and precision medicine. Considering that the conventional wet-lab method is time-consuming, labor-intensive and expensive, computational approach is an alternative choice. The increasing availability of numerous biological data provides a great opportunity to systematically understand complex interaction mechanisms between microbes and drugs. However, few computational methods have been developed for microbe drug prediction. In this work, we leverage multiple sources of biomedical data to construct a heterogeneous network for microbes and drugs, including drug-drug interactions, microbe-microbe interactions and microbe-drug associations. And then we propose a novel Heterogeneous Network Embedding Representation framework for Microbe-Drug Association prediction, named (HNERMDA), by combining metapath2vec with bipartite network recommendation. In this framework, we introduce metapath2vec, a heterogeneous network representation learning method, to learn low-dimensional embedding representations for microbes and drugs. Following that, we further design a bias bipartite network projection recommendation algorithm to improve prediction accuracy. Comprehensive experiments on two datasets, named MDAD and aBiofilm, demonstrated that our model consistently outperformed five baseline methods in three types of cross-validations. Case study on two popular drugs (i.e., Ciprofloxacin and Pefloxacin) further validated the effectiveness of our HNERMDA model in inferring potential target microbes for drugs.
Collapse
|
68
|
Liu Y, Baba Y, Ishimoto T, Tsutsuki H, Zhang T, Nomoto D, Okadome K, Yamamura K, Harada K, Eto K, Hiyoshi Y, Iwatsuki M, Nagai Y, Iwagami S, Miyamoto Y, Yoshida N, Komohara Y, Ohmuraya M, Wang X, Ajani JA, Sawa T, Baba H. Fusobacterium nucleatum confers chemoresistance by modulating autophagy in oesophageal squamous cell carcinoma. Br J Cancer 2020; 124:963-974. [PMID: 33299132 PMCID: PMC7921654 DOI: 10.1038/s41416-020-01198-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 09/23/2020] [Accepted: 11/11/2020] [Indexed: 12/21/2022] Open
Abstract
Background Fusobacterium nucleatum (F. nucleatum) is a gut microbe implicated in gastrointestinal tumorigenesis. Predicting the chemotherapeutic response is critical to developing personalised therapeutic strategies for oesophageal cancer patients. The present study investigated the relationship between F. nucleatum and chemotherapeutic resistance in oesophageal squamous cell carcinoma (ESCC). Methods We examined the relationship between F. nucleatum and chemotherapy response in 120 ESCC resected specimens and 30 pre-treatment biopsy specimens. In vitro studies using ESCC cell lines and co-culture assays further uncovered the mechanism underlying chemotherapeutic resistance. Results ESCC patients with F. nucleatum infection displayed lesser chemotherapeutic response. The infiltration and subsistence of F. nucleatum in the ESCC cells were observed by transmission electron microscopy and laser scanning confocal microscopy. We also observed that F. nucleatum modulates the endogenous LC3 and ATG7 expression, as well as autophagosome formation to induce chemoresistance against 5-FU, CDDP, and Docetaxel. ATG7 knockdown resulted in reversal of F. nucleatum-induced chemoresistance. In addition, immunohistochemical studies confirmed the correlation between F. nucleatum infection and ATG7 expression in 284 ESCC specimens. Conclusions F. nucleatum confers chemoresistance to ESCC cells by modulating autophagy. These findings suggest that targeting F. nucleatum, during chemotherapy, could result in variable therapeutic outcomes for ESCC patients.
Collapse
Affiliation(s)
- Yang Liu
- Second Oncology Department, Shengjing Hospital affiliated of China Medical University, Shenyang, Liaoning, China.,Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Next-Generation Surgical Therapy Development, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyasu Tsutsuki
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tianli Zhang
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daichi Nomoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuo Okadome
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kensuke Yamamura
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuto Harada
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kojiro Eto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukiharu Hiyoshi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yohei Nagai
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shiro Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaki Ohmuraya
- Department of Genetics, Hyogo College of Medicine, Hyogo, Japan
| | - Xiaoming Wang
- Radiology Department, Shengjing Hospital affiliated of China Medical University, Shenyang, Liaoning, China
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan. .,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
69
|
Garajová I, Balsano R, Wang H, Leonardi F, Giovannetti E, Deng D, Peters GJ. The role of the microbiome in drug resistance in gastrointestinal cancers. Expert Rev Anticancer Ther 2020; 21:165-176. [PMID: 33115280 DOI: 10.1080/14737140.2021.1844007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The microbiota is recognized for its impact on both human health and disease. The human microbiota is made up of trillions of cells, including bacteria, viruses, and fungi. The largest population of microbes reside in the gut, prompting research for better understanding of the impact of gastrointestinal microbiota in different diseases. Evidence from numerous studies has pointed out the role of commensal microbes as key determinants of cancer pathogenesis. Moreover, gut microbiota may play an important role in chemoresistance; consequently, this knowledge might be important for novel strategies to improve anticancer treatment efficacy.Areas covered: We describe the role of microbiota in different gastrointestinal cancer types (esophageal, gastric, colorectal, hepatocellular and pancreatic-biliary tract cancers). Moreover, we analyzed the impact of the microbiota on resistance to anticancer therapies, and, lastly, we focused on possibilities of microbiota modulation to enhance anticancer therapy efficacy.Expert opinion: Increasing evidence shows that gut microbiota might influence resistance to anticancer treatment, including conventional chemotherapy, immunotherapy, radiotherapy, and surgery. Therefore, a better knowledge of gut microbiota and its interactions with anticancer drugs will enable us to develop novel anticancer treatment strategies and subsequently improve the cancer patients' outcome.
Collapse
Affiliation(s)
- Ingrid Garajová
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Rita Balsano
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Heling Wang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | | | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienz, Pisa, Italy
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
70
|
Innao V, Allegra AG, Musolino C, Allegra A. New Frontiers about the Role of Human Microbiota in Immunotherapy: The Immune Checkpoint Inhibitors and CAR T-Cell Therapy Era. Int J Mol Sci 2020; 21:ijms21238902. [PMID: 33255336 PMCID: PMC7727716 DOI: 10.3390/ijms21238902] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Microbiota is considered an independent organ with the capability to modulate tumor growth and response to therapies. In the chemo-free era, the use of new immunotherapies, more selective and effective and less toxic, led to the extension of overall survival of patients, subject to their ability to not stop treatment. This has focused scientists’ attention to optimize responses by understanding and changing microbiota composition. While we have obtained abundant data from studies in oncologic and hematologic patients receiving conventional chemotherapy, we have less data about alterations in intestinal flora in those undergoing immunotherapy, especially based on Chimeric Antigen Receptor (CAR) T-cells. Actually, we know that the efficacy of Programmed Cell Death 1 (PD-1), PD-1 ligand, and Cytotoxic T lymphocyte-associated protein 4 (CTLA-4) is improved by probiotics rich in Bifidobacterium spp., while compounds of Bacteroidales and Burkholderiales protect from the development of the anti-CTLA-4-induced colitis in mouse models. CAR T-cell therapy seems to not be interfering with microbiota; however, the numerous previous therapies may have caused permanent damage, thus obscuring the data we might have obtained. Therefore, this review opens a new chapter to transfer known acquisitions to a typology of patients destined to grow.
Collapse
Affiliation(s)
- Vanessa Innao
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood, University of Messina, 98122 Messina, Italy;
- Correspondence: (V.I.); (A.A.)
| | - Andrea Gaetano Allegra
- Radiation Oncology Unit, Department of Biomedical, Experimental, and Clinical Sciences “Mario Serio”, Azienda Ospedaliero-Universitaria Careggi, University of Florence, 50100 Florence, Italy;
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood, University of Messina, 98122 Messina, Italy;
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood, University of Messina, 98122 Messina, Italy;
- Correspondence: (V.I.); (A.A.)
| |
Collapse
|
71
|
Yao Y, Cai X, Fei W, Ren F, Wang F, Luan X, Chen F, Zheng C. Regulating Gut Microbiome: Therapeutic Strategy for Rheumatoid Arthritis During Pregnancy and Lactation. Front Pharmacol 2020; 11:594042. [PMID: 33343364 PMCID: PMC7748111 DOI: 10.3389/fphar.2020.594042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation and bone destruction. Microbial infection is considered to be the most important inducement of RA. The pregnancy planning of women in childbearing age is seriously affected by the disease activity of RA. Gut microbiome, related to immunity and inflammatory response of the host. At present, emerging evidence suggested there are significant differences in the diversity and abundance of gut microbiome during pregnancy and lactation, which may be associated with the fluctuation of RA disease activity. Based on these research foundations, we pioneer the idea of regulating gut microbiome for the treatment of RA during pregnancy and lactation. In this review, we mainly introduce the potential treatment strategies for controlling the disease activity of RA based on gut microbiome during pregnancy and lactation. Besides, we also briefly generalize the effects of conventional anti-rheumatic drugs on gut microbiome, the effects of metabolic changes during pregnancy on gut microbiome, alteration of gut microbiome during pregnancy and lactation, and the effects of anti-rheumatic drugs commonly used during pregnancy and lactation on gut microbiome. These will provide a clear knowledge framework for researchers in immune-related diseases during pregnancy. Regulating gut microbiome may be a potential and effective treatment to control the disease activity of RA during pregnancy and lactation.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Cai
- Department of Pharmacy, Hangzhou First People's Hospital, Hangzhou, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fujia Ren
- Department of Pharmacy, Hangzhou Women's Hospital, Hangzhou, China
| | - Fengmei Wang
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaofei Luan
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fengying Chen
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
72
|
Li Q, Jin M, Liu Y, Jin L. Gut Microbiota: Its Potential Roles in Pancreatic Cancer. Front Cell Infect Microbiol 2020; 10:572492. [PMID: 33117731 PMCID: PMC7575684 DOI: 10.3389/fcimb.2020.572492] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is considered a lethal disease with a low survival rate due to its late-stage diagnosis, few opportunities for resection and lack of effective therapeutic strategies. Multiple, highly complex effects of gut microbiota on pancreatic cancer have been recognized as potential strategies for targeting tumorigenesis, development and treatment in recent decades; some of the treatments include antibiotics, probiotics, and fecal microbiota transplantation. Several bacterial species are associated with carcinogenesis of the pancreas, while some bacterial metabolites contribute to tumor-associated low-grade inflammation and immune responses via several proinflammatory factors and signaling pathways. Given the limited evidence on the interplay between gut microbiota and pancreatic cancer, risk factors associated with pancreatic cancer, such as diabetes, chronic pancreatitis and obesity, should also be taken into consideration. In terms of treatment of pancreatic cancer, gut microbiota has exhibited multiple effects on both traditional chemotherapy and the recently successful immunotherapy. Therefore, in this review, we summarize the latest developments and advancements in gut microbiota in relation to pancreatic cancer to elucidate its potential value.
Collapse
Affiliation(s)
- Quanxiao Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Meng Jin
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Limin Jin
- Department of Anesthesia, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
73
|
Park R, Umar S, Kasi A. Immunotherapy in Colorectal Cancer: Potential of Fecal Transplant and Microbiota-augmented Clinical Trials. CURRENT COLORECTAL CANCER REPORTS 2020; 16:81-88. [PMID: 32607098 PMCID: PMC7325521 DOI: 10.1007/s11888-020-00456-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW This review summarizes the role of the microbiome in colorectal cancer (CRC) in the setting of immunotherapy and emphasizes the potential of microbiota-influencing strategies with a focus on the use of fecal microbiota transplant (FMT). RECENT FINDINGS Observations from preclinical and clinical studies suggest that the human gut microbiome is implicated in the CRC carcinogenesis and is integral in determining the clinical response and toxicity to immunotherapy. Among the therapeutic methods devised to exploit the microbiome, FMT is the most direct method and is backed by the highest level of evidence of efficacy in nonneoplastic disease settings. Furthermore, a favorable microbiome has the potential to overcome immunotherapy resistance and ameliorate immune-related adverse events (irAEs). To this end, clinical trials are underway to evaluate the potential of FMT and microbiota-augmented methods in the setting of immunotherapy in CRC. SUMMARY Evidence from animal studies, retrospective studies, and smaller-scale prospective human studies have led to initiation of a number of microbiota-augmented clinical trials in CRC. Given the intimate relationship between the gut microbiota and the immune system as well as antitumor immune responses, potentiating immunotherapy and managing its toxicity are major areas of research in microbiota-augmented therapies in cancer. Therefore, evaluation of the patient microbiome as a routine part of clinical outcome analysis is warranted in future clinical trials.
Collapse
Affiliation(s)
- Robin Park
- Department of Medicine, MetroWest Medical Center/Tufts University School of Medicine, Framingham, MA, U.S.A
| | - Shahid Umar
- Department of Medicine, Division of Surgery, Kansas University Medical Center, Kansas City, KS, U.S.A
| | - Anup Kasi
- Department of Medicine, Division of Medical Oncology, Kansas University Medical Center, Kansas City, KS, U.S.A
| |
Collapse
|
74
|
Chattopadhyay I, Nandi D, Nag A. The pint- sized powerhouse: Illuminating the mighty role of the gut microbiome in improving the outcome of anti- cancer therapy. Semin Cancer Biol 2020; 70:98-111. [PMID: 32739479 DOI: 10.1016/j.semcancer.2020.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/20/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023]
Abstract
Cancer persists as a major health catastrophe and a leading cause of widespread mortality across every nation. Research of several decades has increased our understanding of the pivotal pathways and key players of the host during tumor development and progression, which has enabled generation of precision therapeutics with improved efficacy. Despite such tremendous advancements in our combat against this fatal disease, a majority of the cancer patients suffer from poor tumor- free survival owing to the increased incidence of recurrent tumor. This is primarily due to the development of resistance against contemporary anti- cancer strategies. Recent studies have pointed towards the involvement of the human symbiotic gut microbiota in regulating the outcome of chemotherapy and immunotherapy. It does so primarily by modulating the metabolism of the drugs and host immune response, thereby enhancing the efficacy and ameliorating the toxicity. The interactions between the therapeutic agents, microbial community and host immunity may provide a new avenue for the clinical management of cancer. In addition, consumption of dietary pro-, pre- and synbiotics has been recognized to confer protection against tumor genesis and also promote improved response to traditional tumor suppressive strategies. Naturally, the use of various combinatorial regimes containing dietary supplements that improve the gut microbiome in amalgamation with conventional cancer treatment methods may significantly augment the therapeutic outcome of cancer patients and circumnavigate the resistance mechanisms that confound traditional therapies. In this review, we have summarized the role of the gut microbiome, which is the largest assembly of commensals within the human body, in regulating the efficacy and toxicity of various existing anti- cancer therapies including chemotherapy, immunotherapy and surgery. Furthermore, we have discussed how novel strategies integrating the application of probiotics, prebiotics, synbiotics and antibiotics in combination with the aforementioned anti- cancer modules manipulate the gut microbiota and, therefore, augment their therapeutic outcome. Together, such innovative anti- tumorigenic approaches may prove highly effective in improving the prognosis of cancer patients.
Collapse
Affiliation(s)
- Indranil Chattopadhyay
- Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, 610001, India.
| | - Deeptashree Nandi
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|
75
|
Ohadian Moghadam S, Momeni SA. Human microbiome and prostate cancer development: current insights into the prevention and treatment. Front Med 2020; 15:11-32. [PMID: 32607819 DOI: 10.1007/s11684-019-0731-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022]
Abstract
The huge communities of microorganisms that symbiotically colonize humans are recognized as significant players in health and disease. The human microbiome may influence prostate cancer development. To date, several studies have focused on the effect of prostate infections as well as the composition of the human microbiome in relation to prostate cancer risk. Current studies suggest that the microbiota of men with prostate cancer significantly differs from that of healthy men, demonstrating that certain bacteria could be associated with cancer development as well as altered responses to treatment. In healthy individuals, the microbiome plays a crucial role in the maintenance of homeostasis of body metabolism. Dysbiosis may contribute to the emergence of health problems, including malignancy through affecting systemic immune responses and creating systemic inflammation, and changing serum hormone levels. In this review, we discuss recent data about how the microbes colonizing different parts of the human body including urinary tract, gastrointestinal tract, oral cavity, and skin might affect the risk of developing prostate cancer. Furthermore, we discuss strategies to target the microbiome for risk assessment, prevention, and treatment of prostate cancer.
Collapse
Affiliation(s)
| | - Seyed Ali Momeni
- Uro-Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
76
|
Singh A, Nayak N, Rathi P, Verma D, Sharma R, Chaudhary A, Agarwal A, Tripathi YB, Garg N. Microbiome and host crosstalk: A new paradigm to cancer therapy. Semin Cancer Biol 2020; 70:71-84. [PMID: 32479952 DOI: 10.1016/j.semcancer.2020.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022]
Abstract
The commensal microbiome of humans has co-evolved for thousands of years. The microbiome regulates human health and is also linked to several diseases, including cancer. The advances in next-generation sequencing have significantly contributed to our understanding of the microbiome and its association with cancer and cancer therapy. Recent studies have highlighted a close relationship of the microbiome to the pharmacological effect of chemotherapy and immunotherapy. The chemo-drugs usually interfere with the host immune system and reduces the microbiome diversity inside the body, which in turn leads to decreased efficacy of these drugs. The human microbiome, specifically the gut microbiome, increases the potency of chemo-drugs through metabolism, enzymatic degradation, ecological differences, and immunomodulation. Recent research exploits the involvement of microbiome to shape the efficacy and decrease the toxicity of these chemo-drugs. In this review, we have highlighted the recent development in understanding the relationship of the human microbiome with cancer and also emphasize on various roles of the microbiome in the modulation of cancer therapy. Additionally, we also summarize the ongoing research focussed on the improved efficacy of chemotherapy and immunotherapy using the host microbiome.
Collapse
Affiliation(s)
- Ashutosh Singh
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi 175005, Himachal Pradesh, India
| | - Namyashree Nayak
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi 175005, Himachal Pradesh, India
| | - Preeti Rathi
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi 175005, Himachal Pradesh, India
| | - Deepanshu Verma
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi 175005, Himachal Pradesh, India
| | - Rohit Sharma
- Department of Rasashastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, BHU, Varanasi 221005, Uttar Pradesh, India
| | - Ashun Chaudhary
- Central University of Himachal Pradesh, Shahpur, Dist. Kangra, Himachal Pradesh 176206, India
| | - Alka Agarwal
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, BHU, Varanasi 221005, Uttar Pradesh, India
| | - Yamini Bhushan Tripathi
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, BHU, Varanasi 221005, Uttar Pradesh, India
| | - Neha Garg
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, BHU, Varanasi 221005, Uttar Pradesh, India.
| |
Collapse
|
77
|
Bjånes TK, Jordheim LP, Schjøtt J, Kamceva T, Cros-Perrial E, Langer A, Ruiz de Garibay G, Kotopoulis S, McCormack E, Riedel B. Intracellular Cytidine Deaminase Regulates Gemcitabine Metabolism in Pancreatic Cancer Cell Lines. Drug Metab Dispos 2020; 48:153-158. [PMID: 31871136 PMCID: PMC11022907 DOI: 10.1124/dmd.119.089334] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/11/2019] [Indexed: 04/19/2024] Open
Abstract
Cytidine deaminase (CDA) is a determinant of in vivo gemcitabine elimination kinetics and cellular toxicity. The impact of CDA activity in pancreatic ductal adenocarcinoma (PDAC) cell lines has not been elucidated. We hypothesized that CDA regulates gemcitabine flux through its inactivation and activation pathways in PDAC cell lines. Three PDAC cell lines (BxPC-3, MIA PaCa-2, and PANC-1) were incubated with 10 or 100 µM gemcitabine for 60 minutes or 24 hours, with or without tetrahydrouridine, a CDA inhibitor. Extracellular inactive gemcitabine metabolite (dFdU) and intracellular active metabolite (dFdCTP) were quantified with liquid chromatography tandem mass spectrometry. Cellular expression of CDA was assessed with real-time PCR and Western blot. Gemcitabine conversion to dFdU was extensive in BxPC-3 and low in MIA PaCa-2 and PANC-1, in accordance with their respective CDA expression levels. CDA inhibition was associated with low or undetectable dFdU in all three cell lines. After 24 hours gemcitabine incubation, dFdCTP was highest in MIA PaCa-2 and lowest in BxPC-3. CDA inhibition resulted in a profound dFdCTP increase in BxPC-3 but not in MIA PaCa-2 or PANC-1. dFdCTP concentrations were not higher after exposure to 100 versus 10 µM gemcitabine when CDA activities were low (MIA PaCa-2 and PANC-1) or inhibited (BxPC-3). The results suggest a regulatory role of CDA for gemcitabine activation in PDAC cells but within limits related to the capacity in the activation pathway in the cell lines. SIGNIFICANCE STATEMENT: The importance of cytidine deaminase (CDA) for cellular gemcitabine toxicity, linking a lower activity to higher toxicity, is well described. An underlying assumption is that CDA, by inactivating gemcitabine, limits the amount available for the intracellular activation pathway. Our study is the first to illustrate this regulatory role of CDA in pancreatic ductal adenocarcinoma cell lines by quantifying intracellular and extracellular gemcitabine metabolite concentrations.
Collapse
Affiliation(s)
- Tormod K Bjånes
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Lars Petter Jordheim
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Jan Schjøtt
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Tina Kamceva
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Emeline Cros-Perrial
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Anika Langer
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Gorka Ruiz de Garibay
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Spiros Kotopoulis
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Emmet McCormack
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Bettina Riedel
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| |
Collapse
|
78
|
Xu J, Peng JJ, Yang W, Fu K, Zhang Y. Vaginal microbiomes and ovarian cancer: a review. Am J Cancer Res 2020; 10:743-756. [PMID: 32266088 PMCID: PMC7136922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/11/2020] [Indexed: 06/11/2023] Open
Abstract
The human microbiome, often termed as "the forgotten organ", is an aggregation of microorganisms and their genomes that forms a mutualistic complex with the host. Recent research has shown the symbiotic merits of a microbiome ecosystem and its crucial role in the hosts' physiological functions. Disruption of this symbiotic relationship is prone to cause a broad spectrum of ailments, including cancer. The compositional and environmental factors that tip the scales from beneficial co-existence to the development of malignancy is actively investigated. Herein we review the latest research in knowledge regarding the association between the vaginal microbiomes and oncogenesis, with a particular focus on ovarian carcinoma.
Collapse
Affiliation(s)
- Jinyun Xu
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South UniversityNo. 87 Xiangya Road, Changsha, China
- Hunan Provincial Gynecological Cancer Diagnosis and Treatment Engineering Research CenterHunan Province, China
| | - Jing-Jie Peng
- Department of Opthalmology, Xiangya Hospital, Central South UniversityNo. 87 Xiangya Road, Changsha, China
| | - Wenqing Yang
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South UniversityNo. 87 Xiangya Road, Changsha, China
- Hunan Provincial Gynecological Cancer Diagnosis and Treatment Engineering Research CenterHunan Province, China
| | - Kun Fu
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South UniversityNo. 87 Xiangya Road, Changsha, China
- Hunan Provincial Gynecological Cancer Diagnosis and Treatment Engineering Research CenterHunan Province, China
| | - Yu Zhang
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South UniversityNo. 87 Xiangya Road, Changsha, China
- Hunan Provincial Gynecological Cancer Diagnosis and Treatment Engineering Research CenterHunan Province, China
| |
Collapse
|
79
|
Guven DC, Aktas BY, Simsek C, Aksoy S. Gut microbiota and cancer immunotherapy: prognostic and therapeutic implications. Future Oncol 2020; 16:497-506. [PMID: 32100550 DOI: 10.2217/fon-2019-0783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The immune checkpoint inhibitors have opened new horizons in oncology. Although the indications for the use of Immune checkpoint inhibitors in cancer patients are expanding, there is still a need for markers that can aid in patient selection. Gastrointestinal microbiota can be among these markers. Recently, gastrointestinal microbiota stated to have a bidirectional relation with cancer immunotherapy with roles in both prognostic and therapeutic sides. Preclinical data suggest that modulation of the microbiota could become a novel strategy for improving the efficacy of immunotherapy. However, its labile structure prone to be affected by many factors. Further research can delineate the mechanisms of the relationship between microbiota and immunotherapy can have clinical implications.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara 06100, Turkey
| | - Burak Yasin Aktas
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara 06100, Turkey
| | - Cem Simsek
- Department of Gastroenterology, Hacettepe University Faculty of Medicine, Ankara 06100, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara 06100, Turkey
| |
Collapse
|
80
|
Zhang X, Liu Q, Liao Q, Zhao Y. Pancreatic Cancer, Gut Microbiota, and Therapeutic Efficacy. J Cancer 2020; 11:2749-2758. [PMID: 32226493 PMCID: PMC7086274 DOI: 10.7150/jca.37445] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 01/04/2020] [Indexed: 01/18/2023] Open
Abstract
Pancreatic cancer remains one of the leading causes of cancer-related death worldwide and has a poor prognosis. Current treatment relies on surgical resection and adjuvant therapies. The gut microbiota plays important roles in metabolism and immunomodulation. Accumulating evidence has implied that the gut microbiota is involved in the metabolism of chemotherapeutic drugs and the tumor microenvironment (TME), which could affect the efficacy of both conventional chemotherapy and immunotherapy for pancreatic cancer. Herein, we comprehensively reviewed the history and highlights of the interactions among pancreatic cancer, the gut microbiota and therapeutic efficacy and showed the promising future of manipulating the gut microbiota to improve clinical outcomes of pancreatic cancer.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
81
|
Bjånes T, Kotopoulis S, Murvold ET, Kamčeva T, Gjertsen BT, Gilja OH, Schjøtt J, Riedel B, McCormack E. Ultrasound- and Microbubble-Assisted Gemcitabine Delivery to Pancreatic Cancer Cells. Pharmaceutics 2020; 12:pharmaceutics12020141. [PMID: 32046005 PMCID: PMC7076495 DOI: 10.3390/pharmaceutics12020141] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer death worldwide. Poor drug delivery to tumours is thought to limit chemotherapeutic treatment efficacy. Sonoporation combines ultrasound (US) and microbubbles to increase the permeability of cell membranes. We assessed gemcitabine uptake combined with sonoporation in vitro in three PDAC cell lines (BxPC-3, MIA PaCa-2 and PANC-1). Cells were cultured in hypoxic bioreactors, while gemcitabine incubation ± sonoporation was conducted in cells with operational or inhibited nucleoside membrane transporters. Intracellular active metabolite (dFdCTP), extracellular gemcitabine, and inactive metabolite (dFdU) concentrations were measured with liquid chromatography tandem mass spectrometry. Sonoporation with increasing US intensities resulted in decreasing extracellular gemcitabine concentrations in all three cell lines with inhibited membrane transporters. In cells with inhibited membrane transporters, without sonoporation, dFdCTP concentrations were reduced down to 10% of baseline. Sonoporation partially restored gemcitabine uptake in these cells, as indicated by a moderate increase in dFdCTP concentrations (up to 37% of baseline) in MIA PaCa-2 and PANC-1. In BxPC-3, gemcitabine was effectively inactivated to dFdU, which might represent a protective mechanism against dFdCTP accumulation in these cells. Intracellular dFdCTP concentrations did not change significantly following sonoporation in any of the cell lines with operational membrane transporters, indicating that the gemcitabine activation pathway may have been saturated with the drug. Sonoporation allowed a moderate increase in gemcitabine transmembrane uptake in all three cell lines, but pre-existing nucleoside transporters were the major determinants of gemcitabine uptake and retention.
Collapse
Affiliation(s)
- Tormod Bjånes
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen 5021, Norway; (T.K.); (J.S.); (B.R.)
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen 5021, Norway;
- Correspondence: (T.B.); (E.M.)
| | - Spiros Kotopoulis
- Phoenix Solutions AS, Ullernchausseen 64, 0379 Oslo, Norway;
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen 5021, Norway;
- Department of Clinical Medicine, University of Bergen, Bergen 5021, Norway
| | | | - Tina Kamčeva
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen 5021, Norway; (T.K.); (J.S.); (B.R.)
| | - Bjørn Tore Gjertsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen 5021, Norway;
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen 5021, Norway
| | - Odd Helge Gilja
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen 5021, Norway;
- Department of Clinical Medicine, University of Bergen, Bergen 5021, Norway
| | - Jan Schjøtt
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen 5021, Norway; (T.K.); (J.S.); (B.R.)
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen 5021, Norway;
| | - Bettina Riedel
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen 5021, Norway; (T.K.); (J.S.); (B.R.)
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen 5021, Norway;
| | - Emmet McCormack
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen 5021, Norway;
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen 5021, Norway
- Correspondence: (T.B.); (E.M.)
| |
Collapse
|
82
|
Wang Y, Yang G, You L, Yang J, Feng M, Qiu J, Zhao F, Liu Y, Cao Z, Zheng L, Zhang T, Zhao Y. Role of the microbiome in occurrence, development and treatment of pancreatic cancer. Mol Cancer 2019; 18:173. [PMID: 31785619 PMCID: PMC6885316 DOI: 10.1186/s12943-019-1103-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies. Recent studies indicated that development of pancreatic cancer may be intimately connected with the microbiome. In this review, we discuss the mechanisms through which microbiomes affect the development of pancreatic cancer, including inflammation and immunomodulation. Potential therapeutic and diagnostic applications of microbiomes are also discussed. For example, microbiomes may serve as diagnostic markers for pancreatic cancer, and may also play an important role in determining the efficacies of treatments such as chemo- and immunotherapies. Future studies will provide additional insights into the various roles of microbiomes in pancreatic cancer.
Collapse
Affiliation(s)
- Yicheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| |
Collapse
|
83
|
Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol 2019; 16:690-704. [PMID: 31554963 DOI: 10.1038/s41575-019-0209-8] [Citation(s) in RCA: 689] [Impact Index Per Article: 137.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2019] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) accounts for about 10% of all new cancer cases globally. Located at close proximity to the colorectal epithelium, the gut microbiota comprises a large population of microorganisms that interact with host cells to regulate many physiological processes, such as energy harvest, metabolism and immune response. Sequencing studies have revealed microbial compositional and ecological changes in patients with CRC, whereas functional studies in animal models have pinpointed the roles of several bacteria in colorectal carcinogenesis, including Fusobacterium nucleatum and certain strains of Escherichia coli and Bacteroides fragilis. These findings give new opportunities to take advantage of our knowledge on the gut microbiota for clinical applications, such as gut microbiota analysis as screening, prognostic or predictive biomarkers, or modulating microorganisms to prevent cancer, augment therapies and reduce adverse effects of treatment. This Review aims to provide an overview and discussion of the gut microbiota in colorectal neoplasia, including relevant mechanisms in microbiota-related carcinogenesis, the potential of utilizing the microbiota as CRC biomarkers, and the prospect for modulating the microbiota for CRC prevention or treatment. These scientific findings will pave the way to clinically translate the use of gut microbiota for CRC in the near future.
Collapse
|
84
|
Untapped "-omics": the microbial metagenome, estrobolome, and their influence on the development of breast cancer and response to treatment. Breast Cancer Res Treat 2019; 179:287-300. [PMID: 31646389 DOI: 10.1007/s10549-019-05472-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023]
Abstract
With the advent of next generation sequencing technologies, there is an increasingly complex understanding of the role of gastrointestinal and local breast microbial dysbiosis in breast cancer. In this review, we summarize the current understanding of the microbiome's role in breast carcinogenesis, discussing modifiable risk factors that may affect breast cancer risk by inducing dysbiosis as well as recent sequencing data illustrating breast cancer subtype-specific differences in local breast tissue microbiota. We outline how the 'estrobolome,' the aggregate of estrogen-metabolizing enteric bacterial genes, may affect the risk of developing postmenopausal estrogen receptor-positive breast cancer. We also discuss the microbiome's potent capacity for anticancer therapy activation and deactivation, an important attribute of the gastrointestinal microbiome that has yet to be harnessed clinically.
Collapse
|
85
|
Villéger R, Lopès A, Carrier G, Veziant J, Billard E, Barnich N, Gagnière J, Vazeille E, Bonnet M. Intestinal Microbiota: A Novel Target to Improve Anti-Tumor Treatment? Int J Mol Sci 2019; 20:ijms20184584. [PMID: 31533218 PMCID: PMC6770123 DOI: 10.3390/ijms20184584] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 12/18/2022] Open
Abstract
Recently, preclinical and clinical studies targeting several types of cancer strongly supported the key role of the gut microbiota in the modulation of host response to anti-tumoral therapies such as chemotherapy, immunotherapy, radiotherapy and even surgery. Intestinal microbiome has been shown to participate in the resistance to a wide range of anticancer treatments by direct interaction with the treatment or by indirectly stimulating host response through immunomodulation. Interestingly, these effects were described on colorectal cancer but also in other types of malignancies. In addition to their role in therapy efficacy, gut microbiota could also impact side effects induced by anticancer treatments. In the first part of this review, we summarized the role of the gut microbiome on the efficacy and side effects of various anticancer treatments and underlying mechanisms. In the second part, we described the new microbiota-targeting strategies, such as probiotics and prebiotics, antibiotics, fecal microbiota transplantation and physical activity, which could be effective adjuvant therapies developed in order to improve anticancer therapeutic efficiency.
Collapse
Affiliation(s)
- Romain Villéger
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
| | - Amélie Lopès
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
- Biologics Research, Sanofi R&D, 94400 Vitry-Sur-Seine, France.
| | - Guillaume Carrier
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
- Surgical Oncology Department, Institut du Cancer de Montpellier (ICM), Univ Montpellier, 34298 Montpellier, France.
| | - Julie Veziant
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
- Service de Chirurgie Digestive, CHU Clermont-Ferrand, Inserm, Université Clermont Auvergne, 63003 Clermont-Ferrand, France.
- 3iHP, CHU Clermont-Ferrand, Inserm, Université Clermont Auvergne, 63003 Clermont-Ferrand, France.
| | - Elisabeth Billard
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
| | - Nicolas Barnich
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
| | - Johan Gagnière
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
- Service de Chirurgie Digestive, CHU Clermont-Ferrand, Inserm, Université Clermont Auvergne, 63003 Clermont-Ferrand, France.
- 3iHP, CHU Clermont-Ferrand, Inserm, Université Clermont Auvergne, 63003 Clermont-Ferrand, France.
| | - Emilie Vazeille
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
- 3iHP, CHU Clermont-Ferrand, Inserm, Université Clermont Auvergne, 63003 Clermont-Ferrand, France.
- Service d'Hépato-gastro-entérologie, CHU Clermont-Ferrand, Inserm, Université Clermont Auvergne, 63003 Clermont-Ferrand, France.
| | - Mathilde Bonnet
- Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
86
|
Bodkhe R, Balakrishnan B, Taneja V. The role of microbiome in rheumatoid arthritis treatment. Ther Adv Musculoskelet Dis 2019; 11:1759720X19844632. [PMID: 31431810 PMCID: PMC6685117 DOI: 10.1177/1759720x19844632] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder with multifactorial etiology; both genetic and environmental factors are known to be involved in pathogenesis. Treatment with disease-modifying antirheumatic drugs (DMARDs) plays an essential role in controlling disease progression and symptoms. DMARDs have immunomodulatory properties and suppress immune response by interfering in various pro-inflammatory pathways. Recent evidence has shown that the gut microbiota directly and indirectly modulates the host immune system. RA has been associated with dysbiosis of the gut microbiota. Patients with RA treated with DMARDs show partial restoration of eubiotic gut microbiome. Hence, it is essential to understand the impact of DMARDs on the microbial composition and its consequent influences on the host immune system to identify novel therapies for RA. In this review, we discuss the importance of antirheumatic-drug-induced host microbiota modulations and possible probiotics that can generate eubiosis.
Collapse
Affiliation(s)
- Rahul Bodkhe
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | | | - Veena Taneja
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
87
|
Yatoo MI, Parray OR, Bhat RA, Nazir QU, Haq AU, Malik HU, Fazilli MUR, Gopalakrishnan A, Bashir ST, Tiwari R, Khurana SK, Chaicumpa W, Dhama K. Novel Candidates for Vaccine Development Against Mycoplasma Capricolum Subspecies Capripneumoniae (Mccp)-Current Knowledge and Future Prospects. Vaccines (Basel) 2019; 7:E71. [PMID: 31340571 PMCID: PMC6789616 DOI: 10.3390/vaccines7030071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/05/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023] Open
Abstract
Exploration of novel candidates for vaccine development against Mycoplasma capricolum subspecies capripneumoniae (Mccp), the causative agent of contagious caprine pleuropneumonia (CCPP), has recently gained immense importance due to both the increased number of outbreaks and the alarming risk of transboundary spread of disease. Treatment by antibiotics as the only therapeutic strategy is not a viable option due to pathogen persistence, economic issues, and concerns of antibiotic resistance. Therefore, prophylactics or vaccines are becoming important under the current scenario. For quite some time inactivated, killed, or attenuated vaccines proved to be beneficial and provided good immunity up to a year. However, their adverse effects and requirement for larger doses led to the need for production of large quantities of Mccp. This is challenging because the required culture medium is costly and Mycoplasma growth is fastidious and slow. Furthermore, quality control is always an issue with such vaccines. Currently, novel candidate antigens including capsular polysaccharides (CPS), proteins, enzymes, and genes are being evaluated for potential use as vaccines. These have shown potential immunogenicity with promising results in eliciting protective immune responses. Being easy to produce, specific, effective and free from side effects, these novel vaccine candidates can revolutionize vaccination against CCPP. Use of novel proteomic approaches, including sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), two-dimensional gel electrophoresis, immunoblotting, matrix-assisted laser desorption/ionization-time-of-flight (MALDI-TOF) mass spectrometry, tandem mass spectroscopy, fast protein liquid chromatography (FPLC), bioinformatics, computerized simulation and genomic approaches, including multilocus sequence analysis, next-generation sequencing, basic local alignment search tool (BLAST), gene expression, and recombinant expression, will further enable recognition of ideal antigenic proteins and virulence genes with vaccination potential.
Collapse
Affiliation(s)
- Mohd Iqbal Yatoo
- Mycoplasma Laboratory, Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, Jammu and Kashmir, Srinagar 190006, India.
| | - Oveas Raffiq Parray
- Mycoplasma Laboratory, Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, Jammu and Kashmir, Srinagar 190006, India
| | - Riyaz Ahmed Bhat
- Mycoplasma Laboratory, Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, Jammu and Kashmir, Srinagar 190006, India
| | - Qurat Un Nazir
- Mycoplasma Laboratory, Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, Jammu and Kashmir, Srinagar 190006, India
| | - Abrar Ul Haq
- Mycoplasma Laboratory, Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, Jammu and Kashmir, Srinagar 190006, India
| | - Hamid Ullah Malik
- Mycoplasma Laboratory, Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, Jammu and Kashmir, Srinagar 190006, India
| | - Mujeeb Ur Rehman Fazilli
- Mycoplasma Laboratory, Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, Jammu and Kashmir, Srinagar 190006, India
| | - Arumugam Gopalakrishnan
- Department of Veterinary Clinical Medicine, Madras Veterinary College, Tamilnadu Veterinary and Animal Sciences University, Vepery 600007, India
| | - Shah Tauseef Bashir
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India
| | - Sandip Kumar Khurana
- ICAR-Central Institute for Research on Buffaloes, Sirsa Road, Hisar 125001, India
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India.
| |
Collapse
|
88
|
Hekmatshoar Y, Rahbar Saadat Y, Hosseiniyan Khatibi SM, Ozkan T, Zununi Vahed F, Nariman-Saleh-Fam Z, Pourghassem Gargari B, Sunguroglu A, Zununi Vahed S. The impact of tumor and gut microbiotas on cancer therapy: Beneficial or detrimental? Life Sci 2019; 233:116680. [PMID: 31344431 DOI: 10.1016/j.lfs.2019.116680] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/15/2019] [Accepted: 07/21/2019] [Indexed: 12/18/2022]
Abstract
Cancer is a globally challenging health problem threatening mankind. Despite therapeutic advances in dealing with this malignancy, heterogeneous response and resistance to chemotherapeutic agents remain the hallmarks of cancer therapy. On the other hand, the involvement of the microbiota in affecting human health is well defined. An ever-growing body of evidence implicates the potential link between the microbiome and the efficacy of cancer therapies. Gut microbiota can modulate the metabolism of drugs in a number of ways. The presence of bacteria within the tumor environment can also impact the responses to cancer therapies; changing the chemical structure of chemotherapeutic drugs, affecting their activity, and local concentration. However, the underlying mechanisms by which gut and tumor microbial communities affect the response to cancer therapy are poorly understood and deciphering these mechanisms is of paramount importance. This review provides an overview of how gut and tumor microbiota might affect the efficacy of chemotherapy, radiotherapy, and immunotherapy and alleviate the adverse side effects of these therapies for the development of personalized and effective anticancer therapy.
Collapse
Affiliation(s)
- Yalda Hekmatshoar
- Department of Medical Biology, School of Medicine, Ankara University, Ankara, Turkey
| | - Yalda Rahbar Saadat
- Nutrition Research Center, Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Mahdi Hosseiniyan Khatibi
- International Rice Research Institute (IRRI), Los Banos, Philippines; Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tulin Ozkan
- Department of Medical Biology, School of Medicine, Ankara University, Ankara, Turkey
| | | | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahram Pourghassem Gargari
- Nutrition Research Center, Department of Biochemistry and Diet Therapy, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asuman Sunguroglu
- Department of Medical Biology, School of Medicine, Ankara University, Ankara, Turkey
| | | |
Collapse
|
89
|
Chamseddine AN, Ducreux M, Armand JP, Paoletti X, Satar T, Paci A, Mir O. Intestinal bacterial β-glucuronidase as a possible predictive biomarker of irinotecan-induced diarrhea severity. Pharmacol Ther 2019; 199:1-15. [DOI: 10.1016/j.pharmthera.2019.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
90
|
Emerging Antibiotic Resistance in Mycoplasma Microorganisms, Designing Effective and Novel Drugs / Therapeutic Targets: Current Knowledge and Futuristic Prospects. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2019. [DOI: 10.22207/jpam.13.1.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
91
|
Mikó E, Kovács T, Sebő É, Tóth J, Csonka T, Ujlaki G, Sipos A, Szabó J, Méhes G, Bai P. Microbiome-Microbial Metabolome-Cancer Cell Interactions in Breast Cancer-Familiar, but Unexplored. Cells 2019; 8:E293. [PMID: 30934972 PMCID: PMC6523810 DOI: 10.3390/cells8040293] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is a leading cause of death among women worldwide. Dysbiosis, an aberrant composition of the microbiome, characterizes breast cancer. In this review we discuss the changes to the metabolism of breast cancer cells, as well as the composition of the breast and gut microbiome in breast cancer. The role of the breast microbiome in breast cancer is unresolved, nevertheless it seems that the gut microbiome does have a role in the pathology of the disease. The gut microbiome secretes bioactive metabolites (reactivated estrogens, short chain fatty acids, amino acid metabolites, or secondary bile acids) that modulate breast cancer. We highlight the bacterial species or taxonomical units that generate these metabolites, we show their mode of action, and discuss how the metabolites affect mitochondrial metabolism and other molecular events in breast cancer. These metabolites resemble human hormones, as they are produced in a "gland" (in this case, the microbiome) and they are subsequently transferred to distant sites of action through the circulation. These metabolites appear to be important constituents of the tumor microenvironment. Finally, we discuss how bacterial dysbiosis interferes with breast cancer treatment through interfering with chemotherapeutic drug metabolism and availability.
Collapse
Affiliation(s)
- Edit Mikó
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
- Department of Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Tünde Kovács
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Éva Sebő
- Kenézy Breast Center, Kenézy Gyula County Hospital, 4032 Debrecen, Hungary.
| | - Judit Tóth
- Kenézy Breast Center, Kenézy Gyula County Hospital, 4032 Debrecen, Hungary.
| | - Tamás Csonka
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Gyula Ujlaki
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Adrienn Sipos
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Judit Szabó
- Department of Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Péter Bai
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032 Debrecen, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| |
Collapse
|
92
|
Choy AT, Carnevale I, Coppola S, Meijer LL, Kazemier G, Zaura E, Deng D, Giovannetti E. The microbiome of pancreatic cancer: from molecular diagnostics to new therapeutic approaches to overcome chemoresistance caused by metabolic inactivation of gemcitabine. Expert Rev Mol Diagn 2018; 18:1005-1009. [PMID: 30392417 DOI: 10.1080/14737159.2018.1544495] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Arthur T.F. Choy
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | - Ilaria Carnevale
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-UP Unit, University of Pisa, Pisa, Italy
| | - Stefano Coppola
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Leiden, The Netherlands
| | - Laura L. Meijer
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Geert Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
| | - Egija Zaura
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-UP Unit, University of Pisa, Pisa, Italy
| |
Collapse
|
93
|
von Frieling J, Fink C, Hamm J, Klischies K, Forster M, Bosch TCG, Roeder T, Rosenstiel P, Sommer F. Grow With the Challenge - Microbial Effects on Epithelial Proliferation, Carcinogenesis, and Cancer Therapy. Front Microbiol 2018; 9:2020. [PMID: 30294304 PMCID: PMC6159313 DOI: 10.3389/fmicb.2018.02020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022] Open
Abstract
The eukaryotic host is in close contact to myriads of resident and transient microbes, which influence the crucial physiological pathways. Emerging evidence points to their role of host-microbe interactions for controlling tissue homeostasis, cell fate decisions, and regenerative capacity in epithelial barrier organs including the skin, lung, and gut. In humans and mice, it has been shown that the malignant tumors of these organs harbor an altered microbiota. Mechanistic studies have shown that the altered metabolic properties and secreted factors contribute to epithelial carcinogenesis and tumor progression. Exciting recent work points toward a crucial influence of the associated microbial communities on the response to chemotherapy and immune-check point inhibitors during cancer treatment, which suggests that the modulation of the microbiota might be a powerful tool for personalized oncology. In this article, we provide an overview of how the bacterial signals and signatures may influence epithelial homeostasis across taxa from cnidarians to vertebrates and delineate mechanisms, which might be potential targets for therapy of human diseases by either harnessing barrier integrity (infection and inflammation) or restoring uncontrolled proliferation (cancer).
Collapse
Affiliation(s)
- Jakob von Frieling
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Christine Fink
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Jacob Hamm
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Kenneth Klischies
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Michael Forster
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Thomas C G Bosch
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Thomas Roeder
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Felix Sommer
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
94
|
Compositional differences in gastrointestinal microbiota in prostate cancer patients treated with androgen axis-targeted therapies. Prostate Cancer Prostatic Dis 2018; 21:539-548. [PMID: 29988102 PMCID: PMC6283851 DOI: 10.1038/s41391-018-0061-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND It is well known that the gastrointestinal (GI) microbiota can influence the metabolism, pharmacokinetics, and toxicity of cancer therapies. Conversely, the effect of cancer treatments on the composition of the GI microbiota is poorly understood. We hypothesized that oral androgen receptor axis-targeted therapies (ATT), including bicalutamide, enzalutamide, and abiraterone acetate, may be associated with compositional differences in the GI microbiota. METHODS We profiled the fecal microbiota in a cross-sectional study of 30 patients that included healthy male volunteers and men with different clinical states of prostate cancer (i.e., localized, biochemically recurrent, and metastatic disease) using 16S rDNA amplicon sequencing. Functional inference of identified taxa was performed using PICRUSt. RESULTS We report a significant difference in alpha diversity in GI microbiota among men with versus without a prostate cancer diagnosis. Further analysis identified significant compositional differences in the GI microbiota of men taking ATT, including a greater abundance of species previously linked to response to anti-PD-1 immunotherapy such as Akkermansia muciniphila and Ruminococcaceae spp. In functional analyses, we found an enriched representation of bacterial gene pathways involved in steroid biosynthesis and steroid hormone biosynthesis in the fecal microbiota of men taking oral ATT. CONCLUSIONS There are measurable differences in the GI microbiota of men receiving oral ATT. We speculate that oral hormonal therapies for prostate cancer may alter the GI microbiota, influence clinical responses to ATT, and/or potentially modulate the antitumor effects of future therapies including immunotherapy. Given our findings, larger, longitudinal studies are warranted.
Collapse
|
95
|
Villéger R, Lopès A, Veziant J, Gagnière J, Barnich N, Billard E, Boucher D, Bonnet M. Microbial markers in colorectal cancer detection and/or prognosis. World J Gastroenterol 2018; 24:2327-2347. [PMID: 29904241 PMCID: PMC6000297 DOI: 10.3748/wjg.v24.i22.2327] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/03/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer worldwide. CRC is still associated with a poor prognosis among patients with advanced disease. On the contrary, due to its slow progression from detectable precancerous lesions, the prognosis for patients with early stages of CRC is encouraging. While most robust methods are invasive and costly, actual patient-friendly screening methods for CRC suffer of lack of sensitivity and specificity. Therefore, the development of sensitive, non-invasive and cost-effective methods for CRC detection and prognosis are necessary for increasing the chances of a cure. Beyond its beneficial functions for the host, increasing evidence suggests that the intestinal microbiota is a key factor associated with carcinogenesis. Many clinical studies have reported a disruption in the gut microbiota balance and an alteration in the faecal metabolome of CRC patients, suggesting the potential use of a microbial-based test as a non-invasive diagnostic and/or prognostic tool for CRC screening. This review aims to discuss the microbial signatures associated with CRC known to date, including dysbiosis and faecal metabolome alterations, and the potential use of microbial variation markers for non-invasive early diagnosis and/or prognostic assessment of CRC and advanced adenomas. We will finally discuss the possible use of these markers as predicators for treatment response and their limitations.
Collapse
Affiliation(s)
- Romain Villéger
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
| | - Amélie Lopès
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Research Biologics, Sanofi R&D, Vitry-Sur-Seine 94400, France
| | - Julie Veziant
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Chirurgie digestive, Centre Hospitalier Universitaire, Clermont-Ferrand 63000, France
| | - Johan Gagnière
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Chirurgie digestive, Centre Hospitalier Universitaire, Clermont-Ferrand 63000, France
| | - Nicolas Barnich
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Université Clermont Auvergne, Institut Universitaire de Technologie de Clermont-Ferrand, Clermont-Ferrand 63000, France
| | - Elisabeth Billard
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Université Clermont Auvergne, Institut Universitaire de Technologie de Clermont-Ferrand, Clermont-Ferrand 63000, France
| | - Delphine Boucher
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Université Clermont Auvergne, Institut Universitaire de Technologie de Clermont-Ferrand, Clermont-Ferrand 63000, France
| | - Mathilde Bonnet
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Université Clermont Auvergne, Institut Universitaire de Technologie de Clermont-Ferrand, Clermont-Ferrand 63000, France
| |
Collapse
|
96
|
Porter CM, Shrestha E, Peiffer LB, Sfanos KS. The microbiome in prostate inflammation and prostate cancer. Prostate Cancer Prostatic Dis 2018; 21:345-354. [PMID: 29795140 DOI: 10.1038/s41391-018-0041-1] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 01/26/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND The human microbiome may influence prostate cancer initiation and/or progression through both direct and indirect interactions. To date, the majority of studies have focused on direct interactions including the influence of prostate infections on prostate cancer risk and, more recently, on the composition of the urinary microbiome in relation to prostate cancer. Less well understood are indirect interactions of the microbiome with prostate cancer, such as the influence of the gastrointestinal or oral microbiota on pro- or anti-carcinogenic xenobiotic metabolism, and treatment response. METHODS We review the literature to date on direct and indirect interactions of the microbiome with prostate inflammation and prostate cancer. RESULTS Emerging studies indicate that the microbiome can influence prostate inflammation in relation to benign prostate conditions such as prostatitis/chronic pelvic pain syndrome and benign prostatic hyperplasia, as well as in prostate cancer. We provide evidence that the human microbiome present at multiple anatomic sites (urinary tract, gastrointestinal tract, oral cavity, etc.) may play an important role in prostate health and disease. CONCLUSIONS In health, the microbiome encourages homeostasis and helps educate the immune system. In dysbiosis, a systemic inflammatory state may be induced, predisposing remote anatomical sites to disease, including cancer. The microbiome's ability to affect systemic hormone levels may also be important, particularly in a disease such as prostate cancer that is dually affected by estrogen and androgen levels. Due to the complexity of the potential interconnectedness between prostate cancer and the microbiome, it is vital to further explore and understand the relationships that are involved.
Collapse
Affiliation(s)
- Corey M Porter
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eva Shrestha
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lauren B Peiffer
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Comparative and Molecular Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA. .,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
97
|
Panebianco C, Andriulli A, Pazienza V. Pharmacomicrobiomics: exploiting the drug-microbiota interactions in anticancer therapies. MICROBIOME 2018; 6:92. [PMID: 29789015 PMCID: PMC5964925 DOI: 10.1186/s40168-018-0483-7] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/14/2018] [Indexed: 05/18/2023]
Abstract
Cancer is a major health burden worldwide, and despite continuous advances in medical therapies, resistance to standard drugs and adverse effects still represent an important cause of therapeutic failure. There is a growing evidence that gut bacteria can affect the response to chemo- and immunotherapeutic drugs by modulating either efficacy or toxicity. Moreover, intratumor bacteria have been shown to modulate chemotherapy response. At the same time, anticancer treatments themselves significantly affect the microbiota composition, thus disrupting homeostasis and exacerbating discomfort to the patient. Here, we review the existing knowledge concerning the role of the microbiota in mediating chemo- and immunotherapy efficacy and toxicity and the ability of these therapeutic options to trigger dysbiotic condition contributing to the severity of side effects. In addition, we discuss the use of probiotics, prebiotics, synbiotics, postbiotics, and antibiotics as emerging strategies for manipulating the microbiota in order to improve therapeutic outcome or at least ensure patients a better quality of life all along of anticancer treatments.
Collapse
Affiliation(s)
- Concetta Panebianco
- Division of Gastroenterology, IRCCS "Casa Sollievo della Sofferenza" Hospital, Viale dei Cappuccini, 1, 71013, San Giovanni Rotondo, FG, Italy
| | - Angelo Andriulli
- Division of Gastroenterology, IRCCS "Casa Sollievo della Sofferenza" Hospital, Viale dei Cappuccini, 1, 71013, San Giovanni Rotondo, FG, Italy
| | - Valerio Pazienza
- Division of Gastroenterology, IRCCS "Casa Sollievo della Sofferenza" Hospital, Viale dei Cappuccini, 1, 71013, San Giovanni Rotondo, FG, Italy.
| |
Collapse
|
98
|
Disruptions in gut microbial-host co-metabolism and the development of metabolic disorders. Clin Sci (Lond) 2018; 132:791-811. [PMID: 29661926 DOI: 10.1042/cs20171328] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/20/2018] [Accepted: 02/23/2018] [Indexed: 12/14/2022]
Abstract
The microbial-mammalian metabolic axis has become recognized as an important component governing the overall homeostatic balance of the mammalian host. Disruption of the state of homeostasis among the gut microbiota has been shown to be causally linked to the development of host metabolic diseases including obesity, cardiovascular, diabetes, and fatty liver disease. This disruption is often referred to as gut dysbiosis. Gut dysbiosis leads to altered metabolic products derived from the microbiota and these in turn, typically shift the homeostatic metabolic balance of the host towards a low-grade chronic inflammation, a hallmark of metabolic syndrome. The primary objective of this review is to examine and discuss some very current research that has been done to study the effect of bacterial metabolites on host metabolism, sometimes referred to as microbiota-host co-metabolism. The metabolic conditions reviewed here include obesity, a known risk factor for all of the other metabolic conditions, as well as, cardiovascular disease, diabetes and nonalcoholic fatty liver disease. Only by further understanding the cause and result of gut dysbiosis will an adequate solution be found for metabolic disease, a viewpoint shared by many.
Collapse
|
99
|
Geller LT, Straussman R. Intratumoral bacteria may elicit chemoresistance by metabolizing anticancer agents. Mol Cell Oncol 2017; 5:e1405139. [PMID: 29404397 DOI: 10.1080/23723556.2017.1405139] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
We recently reported that bacteria can be found within pancreatic ductal adenocarcinoma (PDAC) tumors. Some of these bacteria can metabolize and thereby inactivate the nucleoside analog gemcitabine. We demonstrated that the long isoform of the bacterial enzyme cytidine deaminase (CDD) mediates the metabolism of gemcitabine. The clinical effect of overcoming this potential mechanism of drug resistance has yet to be studied.
Collapse
Affiliation(s)
- Leore T Geller
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
100
|
Talas L, Banfalvi G, Fidrus E, Szigeti ZM, Nagy G. Mycoplasma infection followed by time-lapse microscopy. Med Hypotheses 2017; 108:154-158. [PMID: 29055390 DOI: 10.1016/j.mehy.2017.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 09/06/2017] [Indexed: 11/29/2022]
Abstract
Early detection of mycoplasma infection is crucial for saving precious often irreplaceable data from the tissues of patients. Mycoplasma infections cause diseases in the upper and lower respiratory tracts, urethritis in men resulting in painful dysuria, urgency and urethral discharge. Cough, fever, headache, urethritis may persist for several weeks and convalescence is slow. The symptoms of these diseases are aggravated by the detection of mycoplasma infections, that takes either a long time, besides being expensive or is specific and restricted to only a limited number of contaminant strains. Mycoplasmas are hard to detect visually but could be seen and followed by time-lapse microscopy. Our hypothesis is that one can detect mycoplasma infection irrespective of its origin and type of mycoplasma. Main lines of supporting evidence are provided by the time-lapse microscopy showing dynamic morphological alterations caused by mycoplasmas before changes in human cell cultures become visible. Morphometric measurements of mycoplasma infections revealed four subphases: i) detachment of infected cells, ii) aggregation, iii) biofilm formation and iv) shrinkage of infected cells. The applicability of time-lapse microscopy for the detection of mycoplasma infection was validated by a mycoplasma test Kit. Most important implications related to morphometric parameters include the observation of mycoplasma infected cultures for an extended period of time instead of applying static snap-shot microscopy. A reliable method is offered to estimate the time of mycoplasma exposure that elapsed during the cell growth. This microphotometric approach served a more economical detection of mycoplasma contamination at its early stage of cell growth and spread, irrespective of the origin of contaminated serum, without defining the type of mycoplasma.
Collapse
Affiliation(s)
- Laszlo Talas
- Department of Biotechnology and Microbiology, University of Debrecen, Hungary
| | - Gaspar Banfalvi
- Department of Biotechnology and Microbiology, University of Debrecen, Hungary.
| | - Eszter Fidrus
- Department of Biotechnology and Microbiology, University of Debrecen, Hungary
| | - Zsuzsa M Szigeti
- Department of Biotechnology and Microbiology, University of Debrecen, Hungary
| | - Gabor Nagy
- Department of Biotechnology and Microbiology, University of Debrecen, Hungary
| |
Collapse
|