1
|
Wu M, Tian C, Zou Z, Jin M, Liu H. Gastrointestinal Microbiota in Gastric Cancer: Potential Mechanisms and Clinical Applications-A Literature Review. Cancers (Basel) 2024; 16:3547. [PMID: 39456641 PMCID: PMC11506470 DOI: 10.3390/cancers16203547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Emerging evidence highlights the crucial role of gastrointestinal microbiota in the pathogenesis of gastric cancer. Helicobacter pylori (H. pylori) infection stands out as a primary pathogenic factor. However, interventions such as anti-H. pylori therapy, gastric surgeries, immunotherapy, and chronic inflammation significantly remodel the gastric microbiome, implicating a broader spectrum of microorganisms in cancer development. These microbial populations can modulate gastric carcinogenesis through various mechanisms, including sustained chronic inflammation, bacterial genotoxins, alterations in short-chain fatty acids, elevated gastrointestinal bile acids, impaired mucus barrier function, and increased concentrations of N-nitrosamines and lactic acid. The dynamic changes in gut microbiota also critically influence the outcomes of anti-cancer therapies by modifying drug bioavailability and metabolism, thus affecting therapeutic efficacy and side effect profiles. Additionally, the effectiveness of radiotherapy can be significantly impacted by gut microbiota alterations. Novel therapeutic strategies targeting the microbiome, such as dietary interventions, probiotic and synbiotic supplementation, and fecal microbiota transplantation, are showing promise in cancer treatment. Understanding the intricate relationship between the gut microbiota and gastric cancer is essential for developing new, evidence-based approaches to the prevention and treatment of this malignancy.
Collapse
Affiliation(s)
- Mengjiao Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chenjun Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China;
| | - Zhenwei Zou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- The Eighth Hospital of Wuhan, Wuhan 430012, China
| | - Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
2
|
Khorashadizadeh S, Abbasifar S, Yousefi M, Fayedeh F, Moodi Ghalibaf A. The Role of Microbiome and Probiotics in Chemo-Radiotherapy-Induced Diarrhea: A Narrative Review of the Current Evidence. Cancer Rep (Hoboken) 2024; 7:e70029. [PMID: 39410854 PMCID: PMC11480522 DOI: 10.1002/cnr2.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND In this article, we review the most recent research on probiotics effects on diarrhea in both human and animal models of the condition along with the therapeutic potential of these compounds based on their findings. RECENT FINDINGS Nearly 50%-80% of cancer patients experience chemotherapy-induced diarrhea (CID), serious gastrointestinal toxicity of chemotherapeutic and radiation regimens that leads to prolonged hospitalizations, cardiovascular problems, electrolyte imbalances, disruptions in cancer treatment, poor cancer prognosis, and death. CID is typically categorized as osmotic diarrhea. The depletion of colonic crypts and villi by radiotherapy and chemotherapy agents interferes with the absorptive function of the intestine, thereby decreasing the absorption of chloride and releasing water into the intestinal lumen. Probiotic supplements have been found to be able to reverse the intestinal damage caused by chemo-radiation therapy by promoting the growth of crypt and villi and reducing inflammatory pathways. In addition, they support the modulation of immunological and angiogenesis responses in the gut as well as the metabolism of certain digestive enzymes by altering the gut microbiota. CONCLUSION Beyond the benefits of probiotics, additional clinical research is required to clarify the most effective strain combinations and dosages for preventing chemotherapy and radiotherapy diarrhea.
Collapse
Affiliation(s)
| | - Sara Abbasifar
- Student Research CommitteeBirjand University of Medical SciencesBirjandIran
| | - Mohammad Yousefi
- Student Research CommitteeBirjand University of Medical SciencesBirjandIran
| | - Farzad Fayedeh
- Student Research CommitteeBirjand University of Medical SciencesBirjandIran
| | | |
Collapse
|
3
|
Nakatsukasa H, Takahashi M, Shibano M, Ishigami Y, Kawaguchi T, Nakamura Y, Kaneda H. Clinical impact of concomitant BIO-three use in advanced or recurrent non-small cell lung cancer treated with immune-checkpoint inhibitor. Int J Clin Oncol 2024:10.1007/s10147-024-02622-z. [PMID: 39278980 DOI: 10.1007/s10147-024-02622-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/01/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been approved as first-line therapy for advanced non-small cell lung cancer (NSCLC). The probiotic MIYAIRI 588 can potentially improve the outcomes of patients with advanced NSCLC treated with ICI. However, the impact of other probiotics on ICI-treatment efficacy remains unclear. Thus, we aimed to clarify the association between BIO-three use and treatment outcomes in patients with advanced NSCLC treated with ICI. METHODS This retrospective study included patients aged ≥ 18 years with advanced or recurrent NSCLC who had received ICI monotherapy or ICI plus chemotherapy. Concomitant therapy with probiotic bacteria was defined as receiving it within 180 days before ICI therapy. RESULTS Here, 289 patients were enrolled, including 23 (8.0%) receiving BIO-three. In the multivariable analysis, the progression-free survival (PFS) and overall survival (OS) of patients receiving BIO-three tended to be longer than those of patients not receiving probiotic therapy (PFS, hazard ratio [HR] 0.75; 95% confidence interval [CI] 0.43-1.30; p = 0.33; OS, HR 0.69; 95% CI 0.37-1.28; p = 0.24). After propensity score matching with weighted adjustment, patients receiving BIO-three tended to have prolonged PFS (median PFS [range] 7.6 months [2.6-17.4] vs 3.2 months [1.6-7.0]; HR 0.53; 95% CI 0.25-1.12; p = 0.09) and OS (median OS [range] 25.6 months [10.8-not reached] vs 10.9 months [7.3-not reached]; HR 0.57; 95% CI 0.24-1.36; p = 0.20) than those not receiving probiotic therapy. CONCLUSION This study suggests the prognostic impact of concomitant BIO-three use in patients with advanced NSCLC on ICI treatment.
Collapse
Affiliation(s)
- Hitomi Nakatsukasa
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Masaya Takahashi
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
- Department of Quality and Safety Management, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Masahito Shibano
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Yusuke Ishigami
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Tomoya Kawaguchi
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yasutaka Nakamura
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Hiroyasu Kaneda
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| |
Collapse
|
4
|
Stringer AM, Hargreaves BM, Mendes RA, Blijlevens NMA, Bruno JS, Joyce P, Kamath S, Laheij AMGA, Ottaviani G, Secombe KR, Tonkaboni A, Zadik Y, Bossi P, Wardill HR. Updated perspectives on the contribution of the microbiome to the pathogenesis of mucositis using the MASCC/ISOO framework. Support Care Cancer 2024; 32:558. [PMID: 39080025 PMCID: PMC11289053 DOI: 10.1007/s00520-024-08752-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Advances in the treatment of cancer have significantly improved mortality rates; however, this has come at a cost, with many treatments still limited by their toxic side effects. Mucositis in both the mouth and gastrointestinal tract is common following many anti-cancer agents, manifesting as ulcerative lesions and associated symptoms throughout the alimentary tract. The pathogenesis of mucositis was first defined in 2004 by Sonis, and almost 20 years on, the model continues to be updated reflecting ongoing research initiatives and more sophisticated analytical techniques. The most recent update, published by the Multinational Association for Supportive Care in Cancer and the International Society for Oral Oncology (MASCC/ISOO), highlights the numerous co-occurring events that underpin mucositis development. Most notably, a role for the ecosystem of microorganisms that reside throughout the alimentary tract (the oral and gut microbiota) was explored, building on initial concepts proposed by Sonis. However, many questions remain regarding the true causal contribution of the microbiota and associated metabolome. This review aims to provide an overview of this rapidly evolving area, synthesizing current evidence on the microbiota's contribution to mucositis development and progression, highlighting (i) components of the 5-phase model where the microbiome may be involved, (ii) methodological challenges that have hindered advances in this area, and (iii) opportunities for intervention.
Collapse
Affiliation(s)
- Andrea M Stringer
- Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Benjamin M Hargreaves
- Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Rui Amaral Mendes
- Faculty of Medicine, University of Porto/CINTESIS@RISE, Porto, Portugal
- Department of Oral and Maxillofacial Medicine and Diagnostic Sciences, Case Western Reserve University, Cleveland, OH, 44106-7401, USA
| | - Nicole M A Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Julia S Bruno
- Molecular Oncology Center, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Paul Joyce
- Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Srinivas Kamath
- Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Alexa M G A Laheij
- Department of Oral Medicine, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Giulia Ottaviani
- Department of Surgical, Medical and Health Sciences, University of Trieste, Trieste, Italy
| | - Kate R Secombe
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Arghavan Tonkaboni
- Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Yehuda Zadik
- Department of Military Medicine and "Tzameret", Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral Medicine, Sedation and Imaging, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Paolo Bossi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Hannah R Wardill
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia.
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Level 5S, Adelaide, 5000, Australia.
| |
Collapse
|
5
|
Boeder AM, Spiller F, Carlstrom M, Izídio GS. Enterococcus faecalis: implications for host health. World J Microbiol Biotechnol 2024; 40:190. [PMID: 38702495 DOI: 10.1007/s11274-024-04007-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
The microbiota represents a crucial area of research in maintaining human health due to its potential for uncovering novel biomarkers, therapies, and molecular mechanisms relevant to population identification and experimental model characterization. Among these microorganisms, Enterococcus faecalis, a Gram-positive bacterium found in the gastrointestinal tract of humans and animals, holds particular significance. Strains of this bacterial species have sparked considerable debate in the literature due to their dual nature; they can either be utilized as probiotics in the food industry or demonstrate resistance to antibiotics, potentially leading to severe illness, disability, and death. Given the diverse characteristics of Enterococcus faecalis strains, this review aims to provide a comprehensive understanding of their impact on various systems within the host, including the immunological, cardiovascular, metabolic, and nervous systems. Furthermore, we summarize the bacterium-host interaction characteristics and molecular effects to highlight their targets, features, and overall impact on microbial communities and host health.
Collapse
Affiliation(s)
- Ariela Maína Boeder
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Fernando Spiller
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Mattias Carlstrom
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Geison Souza Izídio
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil.
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil.
- Department of Psychiatry and Legal Medicine, Autonomous University of Barcelona, Barcelona, Spain.
- Laboratório de Genética do Comportamento, Universidade Federal de Santa Catarina, Centro de Ciências Biológicas, Departamento de Biologia Celular, Embriologia e Genética, Florianopolis, SC, Brazil.
| |
Collapse
|
6
|
Ni Y, Li R, Shen X, Yi D, Ren Y, Wang F, Geng Y, You Q. Diaphorobacter nitroreducens synergize with oxaliplatin to reduce tumor burden in mice with lung adenocarcinoma. mSystems 2024; 9:e0132323. [PMID: 38483163 PMCID: PMC11019951 DOI: 10.1128/msystems.01323-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024] Open
Abstract
Lung adenocarcinoma (LADC) is the most common lung cancer and the leading cause of cancer-related deaths globally. Accumulating evidence suggests that the gut microbiota regulates the host response to chemotherapeutic drugs and can be targeted to reduce the toxicity of current chemotherapeutic agents. However, the effect of Diaphorobacter nitroreducens synergized with oxaliplatin on the gut microbiota and their impact on LADC have never been explored. This study aimed to evaluate the anti-cancer effects of D. nitroreducens, oxaliplatin, and their combined treatment on tumor growth in tumor-bearing mice. The composition of gut microbiota and the immune infiltration of tumors were evaluated by using 16S rRNA gene high-throughput sequencing and immunofluorescence, respectively. The inhibitory effect of the combination treatment with D. nitroreducens and oxaliplatin was significantly stronger than that of oxaliplatin alone in tumor-bearing mice. Furthermore, we observed that the combination treatment significantly increased the relative abundance of Lactobacillus and Akkermansia in the gut microbiota. Meanwhile, the combination treatment significantly increased the proportions of macrophage but decreased the proportion of regulatory T cells in the LADC tumor tissues of mice. These findings underscored the relationship between D. nitroreducens and the gut microbiota-immune cell-LADC axis, highlighting potential therapeutic avenues for LADC treatment. IMPORTANCE Oxaliplatin is widely used as an effective chemotherapeutic agent in cancer treatment, but its side effects and response rate still need to be improved. Conventional probiotics potentially benefit cancer chemotherapy by regulating gut microbiota and tumor immune infiltration. This study was novel in reporting a more significant inhibitory effect of Diaphorobacter nitroreducens on lung adenocarcinoma (LADC) cells compared with common traditional probiotics and validating its potential as an adjuvant therapy for LADC chemotherapy in mice. This study investigated the impact of D. nitroreducens combined with oxaliplatin on the gut microbiota and immune infiltration of tumors as a potential mechanism to improve anticancer effects.
Collapse
Affiliation(s)
- Yalan Ni
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Rui Li
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoyu Shen
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Deli Yi
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yilin Ren
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Fudong Wang
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Geng
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, China
| | - Qingjun You
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
7
|
Chen C, Xu JL, Gu ZC, Zhou SS, Wei GL, Gu JL, Ma HL, Feng YQ, Song ZW, Yan ZP, Deng S, Ding R, Li SL, Huo JG. Danggui Sini decoction alleviates oxaliplatin-induced peripheral neuropathy by regulating gut microbiota and potentially relieving neuroinflammation related metabolic disorder. Chin Med 2024; 19:58. [PMID: 38584284 PMCID: PMC10999090 DOI: 10.1186/s13020-024-00929-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 04/01/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Danggui Sini decoction (DSD), a traditional Chinese medicine formula, has the function of nourishing blood, warming meridians, and unblocking collaterals. Our clinical and animal studies had shown that DSD can effectively protect against oxaliplatin (OXA)-induced peripheral neuropathy (OIPN), but the detailed mechanisms remain uncertain. Multiple studies have confirmed that gut microbiota plays a crucial role in the development of OIPN. In this study, the potential mechanism of protective effect of DSD against OIPN by regulating gut microbiota was investigated. METHODS The neuroprotective effects of DSD against OIPN were examined on a rat model of OIPN by determining mechanical allodynia, biological features of dorsal root ganglia (DRG) as well as proinflammatory indicators. Gut microbiota dysbiosis was characterized using 16S rDNA gene sequencing and metabolism disorders were evaluated using untargeted and targeted metabolomics. Moreover the gut microbiota mediated mechanisms were validated by antibiotic intervention and fecal microbiota transplantation. RESULTS DSD treatment significantly alleviated OIPN symptoms by relieving mechanical allodynia, preserving DRG integrity and reducing proinflammatory indicators lipopolysaccharide (LPS), IL-6 and TNF-α. Besides, DSD restored OXA induced intestinal barrier disruption, gut microbiota dysbiosis as well as systemic metabolic disorders. Correlation analysis revealed that DSD increased bacterial genera such as Faecalibaculum, Allobaculum, Dubosiella and Rhodospirillales_unclassified were closely associated with neuroinflammation related metabolites, including positively with short-chain fatty acids (SCFAs) and sphingomyelin (d18:1/16:0), and negatively with pi-methylimidazoleacetic acid, L-glutamine and homovanillic acid. Meanwhile, antibiotic intervention apparently relieved OIPN symptoms. Furthermore, fecal microbiota transplantation further confirmed the mediated effects of gut microbiota. CONCLUSION DSD alleviates OIPN by regulating gut microbiota and potentially relieving neuroinflammation related metabolic disorder.
Collapse
Affiliation(s)
- Chen Chen
- Department of Oncology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
- Department of Oncology, Yancheng TCM Hospital, Yancheng, 224001, Jiangsu, China
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jian-Lin Xu
- Department of Oncology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
- Department of Oncology, Yancheng TCM Hospital, Yancheng, 224001, Jiangsu, China
| | - Zhan-Cheng Gu
- Department of Oncology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, 215399, China
| | - Shan-Shan Zhou
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Guo-Li Wei
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, 211299, Jiangsu, China
| | - Jia-Lin Gu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hai-Long Ma
- Department of Paediatrics, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
| | - Yan-Qi Feng
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zi-Wei Song
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zhan-Peng Yan
- Clinical Research Department of Chinese and Western Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Shan Deng
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Rong Ding
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China.
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| | - Jie-Ge Huo
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China.
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| |
Collapse
|
8
|
Dai JH, Tan XR, Qiao H, Liu N. Emerging clinical relevance of microbiome in cancer: promising biomarkers and therapeutic targets. Protein Cell 2024; 15:239-260. [PMID: 37946397 PMCID: PMC10984626 DOI: 10.1093/procel/pwad052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/22/2023] [Indexed: 11/12/2023] Open
Abstract
The profound influence of microbiota in cancer initiation and progression has been under the spotlight for years, leading to numerous researches on cancer microbiome entering clinical evaluation. As promising biomarkers and therapeutic targets, the critical involvement of microbiota in cancer clinical practice has been increasingly appreciated. Here, recent progress in this field is reviewed. We describe the potential of tumor-associated microbiota as effective diagnostic and prognostic biomarkers, respectively. In addition, we highlight the relationship between microbiota and the therapeutic efficacy, toxicity, or side effects of commonly utilized treatments for cancer, including chemotherapy, radiotherapy, and immunotherapy. Given that microbial factors influence the cancer treatment outcome, we further summarize some dominating microbial interventions and discuss the hidden risks of these strategies. This review aims to provide an overview of the applications and advancements of microbes in cancer clinical relevance.
Collapse
Affiliation(s)
- Jia-Hao Dai
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510050, China
| | - Xi-Rong Tan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510050, China
| | - Han Qiao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510050, China
| | - Na Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510050, China
| |
Collapse
|
9
|
Chen Y, Wang X, Ye Y, Ren Q. Gut microbiota in cancer: insights on microbial metabolites and therapeutic strategies. Med Oncol 2023; 41:25. [PMID: 38129370 DOI: 10.1007/s12032-023-02249-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/11/2023] [Indexed: 12/23/2023]
Abstract
In recent years, the role of gut microbiota in cancer treatment has attracted substantial attention. It is now well established that gut microbiota and its metabolites significantly contribute to the incidence, treatment, and prognosis of various cancers. This review provides a comprehensive review on the pivotal role of gut microbiota and their metabolites in cancer initiation and progression. Furthermore, it evaluates the impact of gut microbiota on the efficacy and associated side effects of anticancer therapies, including radiotherapy, chemotherapy, and immunotherapy, thus emphasizing the clinical importance of gut microbiota reconstitution in cancer treatment.
Collapse
Affiliation(s)
- Yalan Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Xibin Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Yuwei Ye
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Gansu Province Clinical Research Center for Digestive Diseases, Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Qian Ren
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China.
- Gansu Province Clinical Research Center for Digestive Diseases, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
10
|
Zhang YN, Cui ML, Zhang LM, Lu N, Quan X, Yin K, Li AN, Zhang MX. Gut microbiota in gastric cancer: A determinant of etiology or a therapeutic approach? Shijie Huaren Xiaohua Zazhi 2023; 31:933-939. [DOI: 10.11569/wcjd.v31.i22.933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/28/2023] Open
Abstract
The impact of the gut microbiota on the well-being and pathology of the host has garnered growing interest. In recent times, there has been a surge in understanding the mechanistic connections between the gut microbiota and cancer, particularly in relation to the genesis, progression, and therapeutic approaches for gastric cancer. The dysbiosis of the intestinal microbiome stands as a significant determinant in the etiology of gastric cancer. Currently, a preliminary consensus exists, although the precise mechanism remains incompletely understood. As research progresses, it becomes increasingly evident that intestinal flora significantly contributes to the therapeutic approach for gastric cancer. This paper gives a comprehensive review of the impact of intestinal flora on gastric cancer, examines the role of the intestinal microbiome in the management of gastric cancer, and elucidates the potential of utilizing the intestinal microbiome as an anti-tumor therapy, with an aim to furnish a point of reference and stimulate future research endeavors.
Collapse
Affiliation(s)
- Ya-Nan Zhang
- Xi'an Medical University, Xi'an 710000, Shaanxi Province, China
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Man-Li Cui
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Ling-Min Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Ning Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Xin Quan
- Xi'an Medical University, Xi'an 710000, Shaanxi Province, China
| | - Kun Yin
- Xi'an Medical University, Xi'an 710000, Shaanxi Province, China
| | - An-Na Li
- Xi'an Medical University, Xi'an 710000, Shaanxi Province, China
| | - Ming-Xin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| |
Collapse
|
11
|
Zheng Y, Chen M, Zhang Y, Wang G, Zhao H. Lead exposure disrupted ileal barrier of developmental Japanese quails(Coturnix japonica): Histopathological damages, microbiota dysbiosis and immune disorder. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115488. [PMID: 37717353 DOI: 10.1016/j.ecoenv.2023.115488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/09/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
The gut barrier plays an essential role in maintaining homeostasis and is usually composed of a mechanical barrier, a chemical barrier, an immune barrier, and a biological barrier. However, the impacts of lead (Pb) exposure on avian gut barrier are still unclear. Therefore, the present study tried to determine the toxic effects of Pb on ileal barrier of a biological model-Japanese quail (Coturnix japonica). One-week old quails were exposed to 0, 50, 500 and 1000 ppm Pb in drinking water for 5 weeks. The results showed mechanic barrier in the ileum was disrupted with microstructural deformation featured by epithelial cell abscission, villi contractions and goblet cells reduction as well as ultrastructural changes characterized by swollen mitochondria, blurry tight junctions and microvilli subtraction. Meanwhile, the expression of genes associated with intestinal tight junctions was downregulated in Pb-treated groups indicating tight junction malfunction. Moreover, less mucus and downregulation of expression of mucin2 (Muc2) and Krüppel-like factor 4 (Klf4) indicated chemical barrier disturbance by Pb. In addition, the alteration of microbial diversity and emergence of pathogen bacteria suggested ileal biological barrier disruption by Pb. Furthermore, Pb caused immune dysfunction in the ileum through promoting the expression of pro-inflammatory factors including interleukin 1 beta (IL-1β), interleukin 6 (IL-6), Interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α) and nuclear factor kappa B (NF-κB) and inhibiting the expression of anti-inflammatory factor interleukin 10 (IL-10). The present study demonstrated that Pb may pose health risks to birds through gut barrier damages.
Collapse
Affiliation(s)
- Ying Zheng
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Mingcun Chen
- AP Center, Changzhou Senior High School of Jiangsu Province, Changzhou 213000, China
| | - Yuxin Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Gang Wang
- AP Center, Changzhou Senior High School of Jiangsu Province, Changzhou 213000, China
| | - Hongfeng Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
12
|
Luisa Valerio de Mello Braga L, Simão G, Silva Schiebel C, Caroline Dos Santos Maia A, Mulinari Turin de Oliveira N, Barbosa da Luz B, Rita Corso C, Soares Fernandes E, Maria Ferreira D. Rodent models for anticancer toxicity studies: contributions to drug development and future perspectives. Drug Discov Today 2023:103626. [PMID: 37224998 DOI: 10.1016/j.drudis.2023.103626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023]
Abstract
Antineoplastic treatment induces a type of gastrointestinal toxicity known as mucositis. Findings in animal models are usually easily reproducible, and standardized treatment regimens are often used, thus supporting translational science. Essential characteristics of mucositis, including intestinal permeability, inflammation, immune and oxidative responses, and tissue repair mechanisms, can be easily investigated in these models. Given the effects of mucositis on the quality of life of patients with cancer, and the importance of experimental models in the development of more effective new therapeutic alternatives, this review discusses progress and current challenges in using experimental models of mucositis in translational pharmacology research. Teaser Experimental models for studying gastrointestinal mucositis have provided a wealth of information improving the understanding of antineoplastic toxicity.
Collapse
Affiliation(s)
- Lara Luisa Valerio de Mello Braga
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Carolina Silva Schiebel
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Andressa Caroline Dos Santos Maia
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Bruna Barbosa da Luz
- Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Claudia Rita Corso
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Elizabeth Soares Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Daniele Maria Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil.
| |
Collapse
|
13
|
Roggiani S, Mengoli M, Conti G, Fabbrini M, Brigidi P, Barone M, D'Amico F, Turroni S. Gut microbiota resilience and recovery after anticancer chemotherapy. MICROBIOME RESEARCH REPORTS 2023; 2:16. [PMID: 38046820 PMCID: PMC10688789 DOI: 10.20517/mrr.2022.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 12/05/2023]
Abstract
Although research on the role of the gut microbiota (GM) in human health has sharply increased in recent years, what a "healthy" gut microbiota is and how it responds to major stressors is still difficult to establish. In particular, anticancer chemotherapy is known to have a drastic impact on the microbiota structure, potentially hampering its recovery with serious long-term consequences for patients' health. However, the distinguishing features of gut microbiota recovery and non-recovery processes are not yet known. In this narrative review, we first investigated how gut microbiota layouts are affected by anticancer chemotherapy and identified potential gut microbial recovery signatures. Then, we discussed microbiome-based intervention strategies aimed at promoting resilience, i.e., the rapid and complete recovery of a healthy gut microbial network associated with a better prognosis after such high-impact pharmacological treatments.
Collapse
Affiliation(s)
- Sara Roggiani
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Mariachiara Mengoli
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Gabriele Conti
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Marco Fabbrini
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Monica Barone
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Federica D'Amico
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| |
Collapse
|
14
|
Chrysostomou D, Roberts LA, Marchesi JR, Kinross JM. Gut Microbiota Modulation of Efficacy and Toxicity of Cancer Chemotherapy and Immunotherapy. Gastroenterology 2023; 164:198-213. [PMID: 36309208 DOI: 10.1053/j.gastro.2022.10.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/02/2022] [Accepted: 10/16/2022] [Indexed: 01/31/2023]
Abstract
Accumulating evidence supports not only the functional role of the gut microbiome in cancer development and progression but also its role in defining the efficacy and toxicity of chemotherapeutic agents (5-fluorouracil, cyclophosphamide, irinotecan, oxaliplatin, gemcitabine, methotrexate) and immunotherapeutic compounds (anti-programmed death-ligand 1/anti-programmed cell death protein 1 and anti-cytotoxic T-lymphocyte-associated antigen 4). This evidence is supported in numerous in vitro, animal, and clinical studies that highlight the importance of microbial mechanisms in defining therapeutic responses. The microbiome therefore shapes oncologic outcomes and is now being leveraged for the development of novel personalized therapeutic approaches in cancer treatment. However, if the microbiome is to be successfully translated into next-generation oncologic treatments, a new multimodal model of the oncomicrobiome must be conceptualized that incorporates gut microbial cometabolism of pharmacologic agents into cancer care. The objective of this review is therefore to outline the current knowledge of oncologic pharmacomicrobiomics and to describe how the multiparametric functions of the gut microbiome influence treatment response across cancer types. The secondary objective is to propose innovative approaches for modulating the gut microbiome in clinical environments that improve therapy efficacy and diminish toxic effects derived from antineoplastic agents for patient benefit.
Collapse
Affiliation(s)
- Despoina Chrysostomou
- Centre for Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Lauren A Roberts
- Centre for Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Julian R Marchesi
- Centre for Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - James M Kinross
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom.
| |
Collapse
|
15
|
Ren Z, Hong Y, Huo Y, Peng L, Lv H, Chen J, Wu Z, Wan C. Prospects of Probiotic Adjuvant Drugs in Clinical Treatment. Nutrients 2022; 14:4723. [PMID: 36432410 PMCID: PMC9697729 DOI: 10.3390/nu14224723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
In modern society, where new diseases and viruses are constantly emerging, drugs are still the most important means of resistance. However, adverse effects and diminished efficacy remain the leading cause of treatment failure and a major determinant of impaired health-related quality of life for patients. Clinical studies have shown that the disturbance of the gut microbial structure plays a crucial role in the toxic and side effects of drugs. It is well known that probiotics have the ability to maintain the balance of intestinal microecology, which implies their potential as an adjunct to prevent and alleviate the adverse reactions of drugs and to make medicines play a better role. In addition, in the past decade, probiotics have been found to have excellent prevention and alleviation effects in drug toxicity side effects, such as liver injury. In this review, we summarize the development history of probiotics, discuss the impact on drug side effects of probiotics, and propose the underlying mechanisms. Probiotics will be a new star in the world of complementary medicine.
Collapse
Affiliation(s)
- Zhongyue Ren
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Yan Hong
- Jiangxi Institution for Drug Control, Nanchang 330024, China
| | - Yalan Huo
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, 575 W Stadium Ave., West Lafayette, IN 47907, USA
| | - Lingling Peng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Huihui Lv
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Jiahui Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Zhihua Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Cuixiang Wan
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang 330047, China
| |
Collapse
|
16
|
Bullard BM, McDonald SJ, Cardaci TD, VanderVeen BN, Murphy EA. Nonpharmacological approaches for improving gut resilience to chemotherapy. Curr Opin Support Palliat Care 2022; 16:151-160. [PMID: 35862879 DOI: 10.1097/spc.0000000000000599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Mucositis of the gastrointestinal tract is a debilitating side effect of chemotherapy that negatively influences treatment tolerance and patient life quality. This review will evaluate the recent literature on nonpharmacological strategies that have the potential to improve chemotherapy-induced mucositis (CIM). RECENT FINDINGS Alternatives to pharmacological approaches have shown great promise in preventing CIM. Natural products, including curcumin, ginseng, quercetin, and patchouli all show potential in mitigating CIM. In addition, dietary patterns, such as the elemental diet, high fiber diet, and diets high in amino acids have documented benefits in preventing CIM. Perhaps the greatest advancement coming to this arena in recent years is in the field of probiotics. Indeed, research on single species as well as probiotic mixtures show potential in reducing CIM insofar as probiotics are now being suggested for treatment of CIM by governing bodies. Although behavioral interventions including psychological interventions and exercise interventions have shown promise in reducing cancer therapy-related side effects, more work in this domain is warranted and particularly in the context of CIM. SUMMARY Alternatives to pharmacological approaches show great potential for use in prevention and treatment of CIM and should be further developed for use in the clinic.
Collapse
Affiliation(s)
- Brooke M Bullard
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| | | | | | | | | |
Collapse
|
17
|
Schemczssen-Graeff Z, Pileggi M. Probiotics and live biotherapeutic products aiming at cancer mitigation and patient recover. Front Genet 2022; 13:921972. [PMID: 36017495 PMCID: PMC9395637 DOI: 10.3389/fgene.2022.921972] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Molecular biology techniques allowed access to non-culturable microorganisms, while studies using analytical chemistry, as Liquid Chromatography and Tandem Mass Spectrometry, showed the existence of a complex communication system among bacteria, signaled by quorum sensing molecules. These approaches also allowed the understanding of dysbiosis, in which imbalances in the microbiome diversity, caused by antibiotics, environmental toxins and processed foods, lead to the constitution of different diseases, as cancer. Colorectal cancer, for example, can originate by a dysbiosis configuration, which leads to biofilm formation, production of toxic metabolites, DNA damage in intestinal epithelial cells through the secretion of genotoxins, and epigenetic regulation of oncogenes. However, probiotic strains can also act in epigenetic processes, and so be use for recovering important intestinal functions and controlling dysbiosis and cancer mitigation through the metabolism of drugs used in chemotherapy, controlling the proliferation of cancer cells, improving the immune response of the host, regulation of cell differentiation and apoptosis, among others. There are still gaps in studies on the effectiveness of the use of probiotics, therefore omics and analytical chemistry are important approaches to understand the role of bacterial communication, formation of biofilms, and the effects of probiotics and microbiome on chemotherapy. The use of probiotics, prebiotics, synbiotics, and metabiotics should be considered as a complement to other more invasive and hazard therapies, such chemotherapy, surgery, and radiotherapy. The study of potential bacteria for cancer treatment, as the next-generation probiotics and Live Biotherapeutic Products, can have a controlling action in epigenetic processes, enabling the use of these bacteria for the mitigation of specific diseases through changes in the regulation of genes of microbiome and host. Thus, it is possible that a path of medicine in the times to come will be more patient-specific treatments, depending on the environmental, genetic, epigenetic and microbiome characteristics of the host.
Collapse
Affiliation(s)
- Zelinda Schemczssen-Graeff
- Comparative Immunology Laboratory, Department of Microbiology, Parasitology and Pathology, Federal University of Paraná, Curitiba, Brazil
| | - Marcos Pileggi
- Environmental Microbiology Laboratory, Structural and Molecular Biology and Genetics Department, Life Sciences and Health Institute, Ponta Grossa State University, Ponta Grossa, Brazil
- *Correspondence: Marcos Pileggi,
| |
Collapse
|
18
|
Inatomi T, Honma M. Effects of probiotics on loperamide-induced constipation in rats. Sci Rep 2021; 11:24098. [PMID: 34916548 PMCID: PMC8677781 DOI: 10.1038/s41598-021-02931-7] [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: 06/14/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022] Open
Abstract
The role of probiotics in mitigating constipation, gut immunity, and gut microbiota has not been well studied. We aimed to evaluate the effects of probiotics on loperamide (LP)-induced constipation in Sprague-Dawley rats. Altogether, 150 male Sprague-Dawley rats (age 8 weeks) were used in the experiments following a 12-day acclimatisation period and were randomly divided into three treatment groups (groups 1, 2, and 3). Spastic constipation was induced via oral LP administration (3 mg/kg) for 6 days, 1 h before administering each test compound in groups 1 and 2. A probiotic solution (4 mL/kg body weight) was orally administered once a day for 6 days in group 2. In group 1, a phosphate buffer solution was orally administered once a day for 6 days, 1 h after each LP administration. In group 3, a phosphate buffer solution was orally administered once a day for 6 days. In the probiotic group, faecal parameters improved; faecal n-butyric acid, acetic acid, and IgA concentrations were increased; intestinal transit time was shortened; and disturbance of intestinal microbiota was inhibited. Our findings suggest that this probiotic was useful in improving various symptoms caused by constipation.
Collapse
Affiliation(s)
- Takio Inatomi
- Inatomi Animal Hospital, 1-1-24 Denenchofu, Ota-ku, Tokyo, 145-0071, Japan.
| | - Mihoko Honma
- Kusama Animal Health Laboratory, 2240 Tsunehiro, Kashima-shi, Saga, 849-1301, Japan
| |
Collapse
|
19
|
A Perspective on the Role of Microbiome for Colorectal Cancer Treatment. Cancers (Basel) 2021; 13:cancers13184623. [PMID: 34572850 PMCID: PMC8468110 DOI: 10.3390/cancers13184623] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Colorectal cancer is the third most diagnosed cancer worldwide and contributes significantly to global mortality and morbidity. The gut microbiome, composed of the trillions of microbes endemic to the human gastrointestinal tract, has been shown to be implicated in colorectal cancer oncogenesis; however, the roles of microbiota and dysbiosis in CRC treatment remain poorly understood. This review sought to characterize this relationship and in doing so, identify how these interactions may inform future treatments in the form of synbiotics designed to alter the host microbiota to achieve optimized treatment outcomes. Abstract In healthy hosts, trillions of microbes colonise the gut and oral cavity in a well-balanced state, maintaining a mutually beneficial relationship. Loss of this balance, termed dysbiosis, is strongly implicated in the pathogenesis of colorectal cancer (CRC). However, the roles of microbiota and dysbiosis in CRC treatment remain poorly understood. Recent studies suggest that the gut microbiota has the ability to affect the host response to chemotherapeutic agents by enhancing drug efficacy, promoting chemoresistance and mediating chemotherapy-induced toxicity and side effects via a variety of mechanisms. Several other studies have also proposed manipulation of the microbiota to optimise CRC treatment. In this review, we summarise the current advancement of knowledge on how microbiota and CRC treatments interact with each other and how this interaction may shed some light on the development of personalised microbiota manipulations that improve CRC treatment outcomes.
Collapse
|