1
|
Liu Q, Yang Y, Pan M, Shi K, Mo D, Li Y, Wang M, Guo L, Qian Z. Camptothecin multifunctional nanoparticles effectively achieve a balance between the efficacy of breast cancer treatment and the preservation of intestinal homeostasis. Bioact Mater 2024; 41:413-426. [PMID: 39184827 PMCID: PMC11342206 DOI: 10.1016/j.bioactmat.2024.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 08/27/2024] Open
Abstract
Camptothecin (CPT) exhibits potent antitumor activity; however, its clinical application is limited by significant gastrointestinal adverse effects (GAEs). Although the severity of GAEs associated with CPT derivatives has decreased, the incidence rate of these adverse effects has remained high. CPT multifunctional nanoparticles (PCRHNs) have the potential to increase the efficacy of CPT while reducing side effects in major target organs; however, the impact of PCRHNs on the GAEs from CPT remains uncertain. Here, we investigated the therapeutic effects of PCRHNs and different doses of CPT and examined their impacts on the intestinal barrier and the intestinal microbiota. We found that the therapeutic efficacy of PCRHNs + Laser treatment was superior to that of high-dose CPT, and PCRHNs + Laser treatment also provided greater benefits by helping maintain intestinal barrier integrity, intestinal microbiota diversity, and intestinal microbiota abundance. In summary, compared to high-dose CPT treatment, PCRHNs + Laser treatment can effectively balance therapeutic effects and GAEs. A high dose of CPT promotes the enrichment of the pathogenic bacteria Escherichia-Shigella, which may be attributed to diarrhea caused by CPT, thus leading to a reduction in microbial burden; additionally, Escherichia-Shigella rapidly grows and occupies niches previously occupied by other bacteria that are lost due to diarrhea. PCRHNs + Laser treatment increased the abundance of Lactobacillus (probiotics), possibly due to the photothermal effect of the PCRHNs. This effect increased catalase activity, thus facilitating the conversion of hydrogen peroxide into oxygen within tumors and increasing oxygen levels in the body, which is conducive to the growth of facultative anaerobic bacteria.
Collapse
Affiliation(s)
- Qingya Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yun Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Pan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kun Shi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dong Mo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yicong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linfeng Guo
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
2
|
Mishra P, Badiyani VM, Jain S, Subramanian S, Maharaj SV, Kumar A, Singh BN. Prebiotics: Ignored player in the fight against cancer. Cancer Rep (Hoboken) 2023; 6:e1870. [PMID: 37458148 PMCID: PMC10644333 DOI: 10.1002/cnr2.1870] [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: 03/22/2023] [Revised: 05/24/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Prebiotics is a relatively neglected area in cancer research, despite evidence suggesting that it plays a key role in suppressing tumour growth and improving immune function. RECENT FINDINGS Including prebiotics in the diet has been shown to strengthen the immune system and can better slow down or prevent the growth of tumours. It has also been strongly indicated in various scientific studies that prebiotics can contribute to the sustenance of a healthy microbiome, which in turn plays an important role in increasing the effectiveness and reducing the side effects of cancer treatments. CONCLUSION In the present review article we highlight the mechanisms by which prebiotics like inulin, fructooligosaccharide (FOS), β-glucan, pectin, and xylooligosaccharide (XOS) function. Furthermore, the beneficial effect of incorporating prebiotics during cancer therapy to improvise gut health and prevent/reverse the damage caused to patients due to chemotherapy has also been elaborated.
Collapse
Affiliation(s)
- Parichita Mishra
- Department of Ageing Research, Manipal School of Life SciencesManipal Academy of Higher EducationManipalKarnatakaIndia
| | - Vidhi Manish Badiyani
- Department of Ageing Research, Manipal School of Life SciencesManipal Academy of Higher EducationManipalKarnatakaIndia
| | - Sakshi Jain
- Biotechnology and Bioinformatics AreaNIIT UniversityNeemranaRajasthanIndia
| | - Sruti Subramanian
- Biotechnology and Bioinformatics AreaNIIT UniversityNeemranaRajasthanIndia
| | | | - Ashwini Kumar
- Biotechnology and Bioinformatics AreaNIIT UniversityNeemranaRajasthanIndia
| | - Bhisham Narayan Singh
- Department of Ageing Research, Manipal School of Life SciencesManipal Academy of Higher EducationManipalKarnatakaIndia
| |
Collapse
|
3
|
Munteanu R, Feder RI, Onaciu A, Munteanu VC, Iuga CA, Gulei D. Insights into the Human Microbiome and Its Connections with Prostate Cancer. Cancers (Basel) 2023; 15:cancers15092539. [PMID: 37174009 PMCID: PMC10177521 DOI: 10.3390/cancers15092539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
The human microbiome represents the diversity of microorganisms that live together at different organ sites, influencing various physiological processes and leading to pathological conditions, even carcinogenesis, in case of a chronic imbalance. Additionally, the link between organ-specific microbiota and cancer has attracted the interest of numerous studies and projects. In this review article, we address the important aspects regarding the role of gut, prostate, urinary and reproductive system, skin, and oral cavity colonizing microorganisms in prostate cancer development. Various bacteria, fungi, virus species, and other relevant agents with major implications in cancer occurrence and progression are also described. Some of them are assessed based on their values of prognostic or diagnostic biomarkers, while others are presented for their anti-cancer properties.
Collapse
Affiliation(s)
- Raluca Munteanu
- Department of In Vivo Studies, Research Center for Advanced Medicine-MEDFUTURE, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Department of Hematology, "Iuliu Hațieganu" University of Medicine and Pharmacy Cluj-Napoca, Victor Babes Street 8, 400012 Cluj-Napoca, Romania
| | - Richard-Ionut Feder
- Department of In Vivo Studies, Research Center for Advanced Medicine-MEDFUTURE, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Anca Onaciu
- Department of NanoBioPhysics, Research Center for Advanced Medicine-MEDFUTURE, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Department of Pharmaceutical Physics and Biophysics, "Iuliu Hațieganu" University of Medicine and Pharmacy, Louis Pasteur Street 6, 400349 Cluj-Napoca, Romania
| | - Vlad Cristian Munteanu
- Department of Urology, The Oncology Institute "Prof Dr. Ion Chiricuta", 400015 Cluj-Napoca, Romania
- Department of Urology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Cristina-Adela Iuga
- Department of Proteomics and Metabolomics, Research Center for Advanced Medicine-MEDFUTURE, "Iuliu Hațieganu" University of Medicine and Pharmacy Cluj-Napoca, Louis Pasteur Street 6, 400349 Cluj-Napoca, Romania
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, "Iuliu Hațieganu" University of Medicine and Pharmacy, Louis Pasteur Street 6, 400349 Cluj-Napoca, Romania
| | - Diana Gulei
- Department of In Vivo Studies, Research Center for Advanced Medicine-MEDFUTURE, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
4
|
Chen Y, Liao X, Li Y, Cao H, Zhang F, Fei B, Bao C, Cao H, Mao Y, Chen X, Gao X, Zhao W, Xu J. Effects of prebiotic supplement on gut microbiota, drug bioavailability, and adverse effects in patients with colorectal cancer at different primary tumor locations receiving chemotherapy: study protocol for a randomized clinical trial. Trials 2023; 24:268. [PMID: 37046334 PMCID: PMC10091326 DOI: 10.1186/s13063-023-07137-y] [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: 09/14/2021] [Accepted: 02/05/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND The prevalence of colorectal cancer (CRC) worldwide is a huge challenge to human health. Primary tumor locations found to impact prognosis and response to therapy. The important role of gut microbiota in the progression and treatment of CRC has led to many attempts of alleviating chemotherapy-induced adverse effects using microecologics. However, the underlying mechanism of the difference in the prognosis of different primary tumor locations and the synergistic effect of prebiotics on chemotherapy need to be further elucidated. This study aims to explore the differences in tumor microbiota and examine the effectiveness of xylooligosaccharides (XOS) on gut microbiota, adverse effects, and bioavailability of chemotherapy drugs in CRC patients at different primary tumor locations. METHODS This is a double-blinded, randomized, parallel controlled clinical trial. Participants with left-sided CRC (LSCRC, n = 50) and right-sided CC (RSCC, n = 50) will randomly allocated to prebiotic group (n = 25) or control group (n = 25) and will receive either a daily XOS (3 g/day) or placebo, respectively, for 12 weeks. The primary outcomes will be the differences in the mucosa microbiota composition at different tumor locations and differences in gut microbiota composition, adverse effects, and blood concentration of capecitabine posttreatment. The secondary outcomes will include other blood indicators, short-chain fatty acids (SCFAs) concentration, quality of life, and mental health. DISCUSSION This study will reveal the potential benefits of prebiotic for improving the gut microbiota composition, alleviating the adverse effects, and improving the efficacy of chemotherapy in patients with CRC. In addition, this study will provide data on the different distribution of tumor microbiota and the different changes of gut microbiota during treatment in LSCRC and RSCC, which may provide novel insights into personalized cancer treatment strategies based on primary tumor locations and gut microbiota in the future. TRIAL REGISTRATION Chinese Clinical Trial Registry ( www.chictr.org.cn ): ChiCTR2100046237. Registered on 12 May 2021.
Collapse
Affiliation(s)
- Ya Chen
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Xiaowei Liao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yanmin Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Hong Cao
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Feng Zhang
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Bojian Fei
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Chuanqing Bao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Huaxiang Cao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Xiaoping Chen
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Xiang Gao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| | - Jianmin Xu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
5
|
Son YM, Kim J. The Microbiome-Immune Axis Therapeutic Effects in Cancer Treatments. J Microbiol Biotechnol 2022; 32:1086-1097. [PMID: 36116940 PMCID: PMC9628962 DOI: 10.4014/jmb.2208.08002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 12/15/2022]
Abstract
During the last decades, research and therapeutic methods in cancer treatment have been evolving. As the results, nowadays, cancer patients are receiving several types of treatments, ranging from chemotherapy and radiation therapy to surgery and immunotherapy. In fact, most cancer patients take a combination of current anti-cancer therapies to improve the efficacy of treatment. However, current strategies still cause some side effects to patients, such as pain and depression. Therefore, there is the need to discover better ways to eradicate cancer whilst minimizing side effects. Recently, immunotherapy, particularly immune checkpoint blockade, is rising as an effective anti-cancer treatment. Unlike chemotherapy or radiation therapy, immunotherapy has few side effects and a higher tumor cell removal efficacy depend on cellular immunological mechanisms. Moreover, recent studies suggest that tissue immune responses are regulated by their microbiome composition. Each tissue has their specific microenvironment, which makes their microbiome composition different, particularly in the context of different types of cancer, such as breast, colorectal, kidney, lung, and skin. Herein, we review the current understanding of the relationship of immune responses and tissue microbiome in cancer in both animal and human studies. Moreover, we discuss the cancermicrobiome-immune axis in the context of cancer development and treatment. Finally, we speculate on strategies to control tissue microbiome alterations that may synergistically affect the immune system and impact cancer treatment outcomes.
Collapse
Affiliation(s)
- Young Min Son
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea,Corresponding author Phone: +82-31-670-4792 E-mail:
| | - Jihwan Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea
| |
Collapse
|
6
|
Liu L, Tian F, Li GY, Xu W, Xia R. The effects and significance of gut microbiota and its metabolites on the regulation of osteoarthritis: Close coordination of gut-bone axis. Front Nutr 2022; 9:1012087. [PMID: 36204373 PMCID: PMC9530816 DOI: 10.3389/fnut.2022.1012087] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a common chronic degenerative disease of articular cartilage in middle-aged and older individuals, which can result in the joint pain and dysfunction, and even cause the joint deformity or disability. With the enhancing process of global aging, OA has gradually become a major public health problem worldwide. Explaining pathogenesis of OA is critical for the development of new preventive and therapeutic interventions. In recent years, gut microbiota (GM) has been generally regarded as a “multifunctional organ,” which is closely relevant with a variety of immune, metabolic and inflammatory functions. Meanwhile, more and more human and animal researches have indicated the existence of gut-bone axis and suggested that GM and its metabolites are closely involved in the pathogenic process of OA, which might become a potential and promising intervention target. Based on the close coordination of gut-bone axis, this review aims to summarize and discuss the mechanisms of GM and its metabolites influencing OA from the aspects of the intestinal mucosal barrier modulation, intestinal metabolites modulation, immune modulation and strategies for the prevention or treatment of OA based on perspectives of GM and its metabolites, thus providing a profound knowledge and recognition of it.
Collapse
|
7
|
Alam Z, Shang X, Effat K, Kanwal F, He X, Li Y, Xu C, Niu W, War AR, Zhang Y. The potential role of prebiotics, probiotics, and synbiotics in adjuvant cancer therapy especially colorectal cancer. J Food Biochem 2022; 46:e14302. [PMID: 35816322 DOI: 10.1111/jfbc.14302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 12/13/2022]
Abstract
Cancer is a global health issue that is rising swiftly with younger people and an increased number of patients. The role of human microbiota in the pathophysiology of tumors has been paid more and more attention. Microecologics including prebiotics, probiotics, and synbiotics are among the best validated/proven resources for the application of microbiological prophylaxis and therapy. There is strong evidence that microecologics have anti-cancer activity and their potential association with cancer is significant. In this review, we will focus on the role of prebiotics, probiotics, and synbiotics in tumor suppression in maintaining the colon barrier, metabolism, immune regulation, inhibition of host tumor cell proliferation, and epidemiological-based recommendations. Besides, other signs illuminate the role of microecological agents to adjunct the cancer treatment and counter the toxic side effects of cancer drugs. In addition, we will explore their role in chemotherapy, where these probiotics can be used as an adjunct to chemotherapy, counteracting the toxic side effects of chemotherapy drugs to minimize or optimize the therapeutic effect. In the treatment of cancer, we can see the role of prebiotics, probiotics, synbiotics, and their application in cancer patients, and the effectiveness effect can be considered as a clinical benefit. PRACTICAL APPLICATIONS: A large number of studies have shown that microecologics including prebiotics, probiotics, and synbiotics play an important role in regulating intestinal microecology and contribute to the prevention and treatment of cancer, indicating that prebiotics, probiotics, and synbiotics have the potential to be used as microecological modulators in the adjuvant therapy of cancer. However, it is not clear what is the anti-tumor mechanism of these microecologics and how they antagonize the side effects of cancer chemotherapy and protect normal cells. This paper reviews the role of prebiotics, probiotics, and synbiotics in tumor suppression in maintaining the colon barrier, metabolism, immune regulation, and prevention of rapid growth of host cells, as well as their potential role in cancer chemotherapy. This review helps to better understand the relationship between prebiotics, probiotics, and synbiotics with immune regulation, intestinal microecology, metabolic regulation, and cell proliferation and provides strong evidence for their potential application as microecologics in cancer adjuvant therapy.
Collapse
Affiliation(s)
- Zahoor Alam
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiaoya Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Khansa Effat
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Freeha Kanwal
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiaoqin He
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yanye Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Chunlan Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Weining Niu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Abdul Rouf War
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yong Zhang
- Department of Surgical Oncology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Maioli TU, Trindade LM, Souza A, Torres L, Andrade MER, Cardoso VN, Generoso SV. Non-pharmacologic strategies for the management of intestinal inflammation. Biomed Pharmacother 2021; 145:112414. [PMID: 34808552 DOI: 10.1016/j.biopha.2021.112414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases, irritable bowel syndrome, and mucositis are characterized by intestinal inflammation, but vary according to their pathological mechanisms, severity, location, and etiology. Significant intestinal inflammation that occurs in these diseases induces weight loss, nutritional depletion, and gastrointestinal tract dysfunction. Nutritional support is important in alleviating symptoms and improving patients' quality of life. In this review, we summarize some nutritional components used to manage intestinal disorders. These include fatty acids, probiotics, parabiotics, postbiotics, prebiotics, synbiotics, and low FODMAP (LFD) diets. These components and LFD diets have been studied and clinical trials have been designed to develop new strategies to alleviate intestinal inflammation and improve the quality of life. Clinical trials on their use in intestinal inflammation do not allow firm conclusions to be drawn mainly because of the heterogeneity of the dose used and the study design or their inconclusive results. However, in the majority of cases, the use of omega-3, probiotics, parabiotics, postbiotics, prebiotics, synbiotics, and LFD improve the health.
Collapse
Affiliation(s)
- Tatiani Uceli Maioli
- Programa de Pós-Graduação em Nutrição e Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Brazil
| | - Luísa Martins Trindade
- Programa de Pós-Graduação em Ciência de Alimentos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Aline Souza
- Programa de Pós-Graduação em Nutrição e Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Brazil
| | - Lícia Torres
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | | | | | - Simone Vasconcelos Generoso
- Programa de Pós-Graduação em Nutrição e Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
9
|
Wu Y, Wu J, Lin Z, Wang Q, Li Y, Wang A, Shan X, Liu J. Administration of a Probiotic Mixture Ameliorates Cisplatin-Induced Mucositis and Pica by Regulating 5-HT in Rats. J Immunol Res 2021; 2021:9321196. [PMID: 34568500 PMCID: PMC8461230 DOI: 10.1155/2021/9321196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022] Open
Abstract
Probiotic-based therapies have been shown to be beneficial for chemotherapy-induced mucositis. Previous research has demonstrated that a probiotic mixture (Bifidobacterium brevis, Lactobacillus acidophilus, Lactobacillus casei, and Streptococcus thermophilus) can ameliorate chemotherapy-induced mucositis and dysbiosis in rats, but the underlying mechanism has not been completely elucidated. We aimed to determine the inhibitory effects of the probiotic mixture on cisplatin-induced mucositis and pica and the underlying mechanism, focusing on the levels of 5-hydroxytryptamine (5-HT, serotonin) regulated by the gut microbiota. A rat model of mucositis and pica was established by daily intraperitoneal injection of cisplatin (6 mg/kg) for 3 days. In the probiotic+cisplatin group, predaily intragastric injection of the probiotic mixture (1 × 109 CFU/kg BW) was administrated for 1 week before cisplatin injection. This was then followed by further daily probiotic injections for 6 days. Histopathology, pro-/anti-inflammatory cytokines, oxidative status, and 5-HT levels were assessed on days 3 and 6. The structure of the gut microbiota was analyzed by 16S rRNA gene sequencing and quantitative PCR. Additionally, 5-HT levels in enterochromaffin (EC) cells (RIN-14B cell line) treated with cisplatin and/or various probiotic bacteria were also determined. The probiotic mixture significantly attenuated kaolin consumption, inflammation, oxidative stress, and the increase in 5-HT concentrations in rats with cisplatin-induced intestinal mucositis and pica. Cisplatin markedly increased the relative abundances of Enterobacteriaceae_other, Blautia, Clostridiaceae_other, and members of Clostridium clusters IV and XIVa. These levels were significantly restored by the probiotic mixture. Importantly, most of the genera increased by cisplatin were significantly positively correlated with colonic 5-HT. Furthermore, in vitro, the probiotic mixture had direct inhibitory effects on the 5-HT secretion by EC cells. The probiotic mixture protects against cisplatin-induced intestine injury, exhibiting both anti-inflammatory and antiemetic properties. These results were closely related to the reestablishment of intestinal microbiota ecology and normalization of the dysbiosis-driven 5-HT overproduction.
Collapse
Affiliation(s)
- Yuanhang Wu
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jianlin Wu
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zhikun Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qian Wang
- Liaoning CapitalBio Technology Co., Ltd., Dalian, China
| | - Ying Li
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Aman Wang
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiu Shan
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
10
|
Alvarenga L, Cardozo LF, Borges NA, Lindholm B, Stenvinkel P, Shiels PG, Fouque D, Mafra D. Can nutritional interventions modulate the activation of the NLRP3 inflammasome in chronic kidney disease? Food Res Int 2020; 136:109306. [DOI: 10.1016/j.foodres.2020.109306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/28/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
|
11
|
Li S, Zhuo M, Huang X, Huang Y, Zhou J, Xiong D, Li J, Liu Y, Pan Z, Li H, Chen J, Li X, Xiang Z, Wu F, Wu K. Altered gut microbiota associated with symptom severity in schizophrenia. PeerJ 2020; 8:e9574. [PMID: 32821537 PMCID: PMC7395597 DOI: 10.7717/peerj.9574] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/29/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The gut microbiome and microbiome-gut-brain (MGB) axis have been receiving increasing attention for their role in the regulation of mental behavior and possible biological basis of psychiatric disorders. With the advance of next-generation sequencing technology, characterization of the gut microbiota in schizophrenia (SZ) patients can provide rich clues for the diagnosis and prevention of SZ. METHODS In this study, we compared the differences in the fecal microbiota between 82 SZ patients and 80 demographically matched normal controls (NCs) by 16S rRNA sequencing and analyzed the correlations between altered gut microbiota and symptom severity. RESULTS The alpha diversity showed no significant differences between the NC and SZ groups, but the beta diversity revealed significant community-level separation in microbiome composition between the two groups (pseudo-F =3.337, p < 0.001, uncorrected). At the phylum level, relatively more Actinobacteria and less Firmicutes (p < 0.05, FDR corrected) were found in the SZ group. At the genus level, the relative abundances of Collinsella, Lactobacillus, Succinivibrio, Mogibacterium, Corynebacterium, undefined Ruminococcus and undefined Eubacterium were significantly increased, whereas the abundances of Adlercreutzia, Anaerostipes, Ruminococcus and Faecalibacterium were decreased in the SZ group compared to the NC group (p < 0.05, FDR corrected). We performed PICRUSt analysis and found that several metabolic pathways differed significantly between the two groups, including the Polyketide sugar unit biosynthesis, Valine, Leucine and Isoleucine biosynthesis, Pantothenate and CoA biosynthesis, C5-Branched dibasic acid metabolism, Phenylpropanoid biosynthesis, Ascorbate and aldarate metabolism, Nucleotide metabolism and Propanoate metabolism pathways (p < 0.05, FDR corrected). Among the SZ group, the abundance of Succinivibrio was positively correlated with the total Positive and Negative Syndrome Scale (PANSS) scores (r = 0.24, p < 0.05, uncorrected) as well as the general PANSS scores (r = 0.22, p < 0.05, uncorrected); Corynebacterium was negatively related to the negative scores of PANSS (r = 0.22, p < 0.05, uncorrected). CONCLUSIONS Our findings provided evidence of altered gut microbial composition in SZ group. In addition, we found that Succinvibrio and Corynebacterium were associated with the severity of symptoms for the first time, which may provide some new biomarkers for the diagnosis of SZ.
Collapse
Affiliation(s)
- Shijia Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Min Zhuo
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Xia Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Yuanyuan Huang
- The Affifiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Jing Zhou
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Dongsheng Xiong
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Jiahui Li
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Ya Liu
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Zhilin Pan
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Hehua Li
- The Affifiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Jun Chen
- Guangdong Engineering Technology Research Center for Diagnosis and Rehabilitation of Dementia, Guangzhou, Guangdong, China
| | - Xiaobo Li
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NY, United States
| | - Zhiming Xiang
- Guangdong Engineering Technology Research Center for Diagnosis and Rehabilitation of Dementia, Guangzhou, Guangdong, China
- Department of Radiology, Panyu Central Hospital of Guangzhou, Guangzhou, Guangdong, China
| | - Fengchun Wu
- The Affifiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
| | - Kai Wu
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- The Affifiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, Guangdong, China
- Guangdong Engineering Technology Research Center for Diagnosis and Rehabilitation of Dementia, Guangzhou, Guangdong, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, Guangdong, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China
- Department of Nuclear Medicine and Radiology/Institute of Development/Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
12
|
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: 1.6] [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
|
13
|
Wu J, Gan Y, Li M, Chen L, Liang J, Zhuo J, Luo H, Xu N, Wu X, Wu Q, Lin Z, Su Z, Liu Y. Patchouli alcohol attenuates 5-fluorouracil-induced intestinal mucositis via TLR2/MyD88/NF-kB pathway and regulation of microbiota. Biomed Pharmacother 2020; 124:109883. [PMID: 32004938 DOI: 10.1016/j.biopha.2020.109883] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 02/09/2023] Open
Abstract
Intestinal mucositis causes great suffering to cancer patients who undergo chemotherapy and radiotherapy. Owing to the uncertain side effects of anticancer drugs to attenuate patients' intestinal mucositis, many studies focused on traditional Chinese medicine (TCM). Patchouli alcohol (PA) is an active compound extracted from Pogostemon cablin, and has potent gastrointestinal protective effect. However, whether PA has an effect on intestinal mucositis is still unknown. Therefore, we established a rat model of intestinal mucositis via intraperitoneal injection of 5-fluorouracil, and intragastrically administrated PA (10, 20, and 40 mg/kg) to evaluate the effect of PA on intestinal mucositis. The routine observation (body weight, food intake, and diarrhea) in rats was used to detect whether PA had an effect on intestinal mucositis. Levels of inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-10, and MPO), mucosal barrier proteins (zonula occludens -1 (ZO-1), claudin-1, occludin, myosin light chain (MLC), and mucin-2) and intestinal microbiota were determined to elucidate the underlying mechanism of PA action on intestinal mucositis in rats. The results showed that PA could effectively improve body weight, food intake, and diarrhea in intestinal mucositis rats, preliminary confirming PA efficacy. Further experiments revealed that PA not only decreased the levels of TNF-α, IL-1β, IL-6, and MPO but also increased the level of IL-10 significantly. In addition, the expression of mucosal barrier proteins and microbiota community were also improved after PA treatment in diseased rats. Hence, PA may prevent the development and progression of intestinal mucositis by improving inflammation, protecting mucosal barrier, and regulating intestinal microbiota.
Collapse
Affiliation(s)
- Jiazhen Wu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuxuan Gan
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Muxia Li
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Liping Chen
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Jiali Liang
- Faculty of Science and Engineering, Macquarie University, Balaclava Road, Macquarie Park, NSW, Sydney, 2109, Australia
| | - Jianyi Zhuo
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huijuan Luo
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Nan Xu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xue Wu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Qiduan Wu
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhixiu Lin
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ziren Su
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan, 523808, China.
| | - Yuhong Liu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
14
|
Chemotherapy-induced neuroinflammation is associated with disrupted colonic and bacterial homeostasis in female mice. Sci Rep 2019; 9:16490. [PMID: 31712703 PMCID: PMC6848141 DOI: 10.1038/s41598-019-52893-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy treatment negatively affects the nervous and immune systems and alters gastrointestinal function and microbial composition. Outside of the cancer field, alterations in commensal bacteria and immune function have been implicated in behavioral deficits; however, the extent to which intestinal changes are related to chemotherapy-associated behavioral comorbidities is not yet known. Thus, this study identified concurrent changes in behavior, central and peripheral immune activation, colon histology, and bacterial community structure in mice treated with paclitaxel chemotherapy. In paclitaxel-treated mice, increased fatigue and decreased cognitive performance occurred in parallel with reduced microglia immunoreactivity, increased circulating chemokine expression (CXCL1), as well as transient increases in pro-inflammatory cytokine/chemokine (Il-1β, Tnfα, Il-6, and Cxcl1) gene expression in the brain. Furthermore, mice treated with paclitaxel had altered colonic bacterial community composition and increased crypt depth. Relative abundances of multiple bacterial taxa were associated with paclitaxel-induced increases in colon mass, spleen mass, and microglia activation. Although microbial community composition was not directly related to available brain or behavioral measures, structural differences in colonic tissue were strongly related to microglia activation in the dentate gyrus and the prefrontal cortex. These data indicate that the chemotherapeutic paclitaxel concurrently affects the gut microbiome, colonic tissue integrity, microglia activation, and fatigue in female mice, thus identifying a novel relationship between colonic tissue integrity and behavioral responses that is not often assessed in studies of the brain-gut-microbiota axis.
Collapse
|
15
|
Yuan X, Kang Y, Zhuo C, Huang XF, Song X. The gut microbiota promotes the pathogenesis of schizophrenia via multiple pathways. Biochem Biophys Res Commun 2019; 512:373-380. [PMID: 30898321 DOI: 10.1016/j.bbrc.2019.02.152] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a severe mental disorder with unknown etiology. Many mechanisms, including dysregulation of neurotransmitters, immune disturbance, and abnormal neurodevelopment, are proposed for the pathogenesis of schizophrenia. The significance of communication between intestinal flora and the central nervous system through the gut-brain axis is increasingly being recognized. The intestinal microbiota plays an important role in regulating neurotransmission, immune homeostasis, and brain development. We hypothesize that an imbalance in intestinal flora causes immune activation and dysfunction in the gut-brain axis, contributing to schizophrenia. In this review, we examine recent studies that explore the intestinal flora and immune-mediated neurodevelopment of schizophrenia. We conclude that an imbalance in intestinal flora may reduce protectants and increase neurotoxin and inflammatory mediators, causing neuronal and synaptic damage, which induces schizophrenia.
Collapse
Affiliation(s)
- Xiuxia Yuan
- The First Affiliated Hospital/Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Yulin Kang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Chuanjun Zhuo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, NSW, 2522, Australia.
| | - Xueqin Song
- The First Affiliated Hospital/Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
16
|
Current Challenges in Cancer Immunotherapy: Multimodal Approaches to Improve Efficacy and Patient Response Rates. JOURNAL OF ONCOLOGY 2019; 2019:4508794. [PMID: 30941175 PMCID: PMC6420990 DOI: 10.1155/2019/4508794] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 02/12/2019] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is a promising innovative treatment for many forms of cancer, particularly melanoma. Although immunotherapy has been shown to be efficacious, patient response rates vary and, more often than not, only a small subset of the patients within a large cohort respond favourably to the treatment. This issue is particularly concerning and becomes a challenge of immunotherapy to improve the effectiveness and patient response rates. Here, we review the specific types of available immunotherapy options, their proposed mechanism(s) of action, and the reasons why the patient response to this treatment is variable. The potential favourable options to improve response rates to immunotherapy will be discussed with an emphasis on adopting a multimodal approach on the novel role that the gut microbiota may play in modulating the efficacy of cancer immunotherapy.
Collapse
|
17
|
Trindade L, Martins V, Rodrigues N, Souza E, Martins F, Costa G, Almeida-Leite C, Faria A, Cardoso V, Maioli T, Generoso S. Oral administration of Simbioflora® (synbiotic) attenuates intestinal damage in a mouse model of 5-fluorouracil-induced mucositis. Benef Microbes 2018; 9:477-486. [DOI: 10.3920/bm2017.0082] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The use of probiotics to prevent or treat mucosal inflammation has been studied; however, the combined effect of probiotics and prebiotics is unclear. The aim of this study was to test whether oral administration of a synbiotic (Simbioflora®) preparation containing Lactobacillus paracasei, Lactobacillus rhamnosus, Lactobacillus acidophilus and Bifidobacterium lactis plus fructooligosaccharide could help control mucosal inflammation in experimental mucositis induced by 5-fluorouracil (5-FU). Male BALB/c mice were randomly divided into six groups: control (CTL), control + prebiotic (CTL+P), control + synbiotic (CTL+S), mucositis (MUC), mucositis + prebiotic (MUC+P), and mucositis + synbiotic (MUC+S). Mice from the CTL+S, MUC+S, CTL+P, and MUC+P groups received synbiotic or prebiotic daily by oral gavage for 13 days. Mice in the CTL and MUC groups received the same volume of saline. On day 11, mice in the MUC, MUC+P, and MUC+S groups received an intraperitoneal injection of 300 mg/kg 5-FU to induce mucositis. After 72 h, all mice were euthanised. Intestinal permeability, intestinal histology, and biochemical parameters were analysed. Group MUC showed a greater weight loss and increased intestinal permeability (0.020 counts per min [cpm]/g) compared to the CTL group (0.01 cpm/g) P<0.05. Both treatments attenuated weight loss compared to the MUC group. Nonetheless, the synbiotic caused a greater reduction in intestinal permeability (0.012 cpm/g) compared to the MUC (0.020 cpm/g) and MUC+P (0.016 cpm/g) groups P<0.05. Mice in groups MUC+P and MUC+S displayed significant recovery of lesions and maintenance of the mucus layer. There were no differences in the short-chain fatty acid concentrations in the faeces between the MUC and CTL groups (P>0.05). Increased acetate and propionate concentrations were evidenced in the faeces of the MUC+P and MUC+S groups. Only the synbiotic treatment increased the butyrate concentration (P<0.05). The results indicate that administration of synbiotic can decrease mucosal damage caused by mucositis.
Collapse
Affiliation(s)
- L.M. Trindade
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - V.D. Martins
- Departamento de Análises Clínicas e Toxicológicas, Escola de Farmácia, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - N.M. Rodrigues
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - E.L.S. Souza
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - F.S. Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - G.M.F. Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - C.M. Almeida-Leite
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - A.M.C. Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - V.N. Cardoso
- Departamento de Análises Clínicas e Toxicológicas, Escola de Farmácia, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil
| | - T.U. Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Av Alfredo Balena 190, Belo Horizonte, MG 30130-100, Brazil
| | - S.V. Generoso
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Av Alfredo Balena 190, Belo Horizonte, MG 30130-100, Brazil
| |
Collapse
|
18
|
Loupakis F, Stein A, Ychou M, Hermann F, Salud A, Österlund P. A Review of Clinical Studies and Practical Guide for the Administration of Triplet Chemotherapy Regimens with Bevacizumab in First-line Metastatic Colorectal Cancer. Target Oncol 2017; 11:293-308. [PMID: 26687849 PMCID: PMC4901088 DOI: 10.1007/s11523-015-0400-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Colorectal cancer is the third most common cancer worldwide. A significant proportion of patients presents with unresectable metastatic disease or develops metachronous metastases following surgical resection of the primary tumor. The prognosis of the disease has improved over the past two decades, with extended multimodality treatment options and the development of increasingly intensified chemotherapy regimens that now typically include targeted biologics. A recent advance in therapy is a treatment regimen composed of three chemotherapeutic agents (i.e., triplet chemotherapy: 5-fluorouracil [5-FU]/leucovorin [LV], oxaliplatin, and irinotecan; FOLFOXIRI) in combination with the vascular endothelial growth factor inhibitor bevacizumab. This regimen has been shown to elicit significantly improved objective response rates and median progression-free survival compared with 5-FU/LV and irinotecan in combination with bevacizumab. However, triplet chemotherapy has been associated with increased rates of chemotherapy-related adverse events, and treatment-emergent adverse events should be properly managed to minimize treatment interruption or discontinuation. We present herein a review of clinical studies evaluating the safety and efficacy of FOLFOXIRI with bevacizumab in metastatic colorectal cancer, and propose a practical guide for the management of adverse events associated with the regimen. ![]()
Collapse
Affiliation(s)
- Fotios Loupakis
- Istituto Toscano Tumori, U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliero-Universitaria Pisana, Via Roma, 67, 56126, Pisa, Italy.
| | - Alexander Stein
- University Cancer Center Hamburg, University of Hamburg, Hamburg, Germany
| | - Marc Ychou
- ICM Val d'Aurelle, 34000, Montpellier, France
| | | | - Antonieta Salud
- Department of Medical Oncology, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Pia Österlund
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
19
|
Kuchay RAH. A review of complementary therapies for chemotherapy induced gastrointestinal mucositis. Drug Discov Ther 2017; 10:292-299. [PMID: 27746417 DOI: 10.5582/ddt.2016.01059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Administration of chemotherapy often leads to gastrointestinal mucositis (GIM). GIM manifests as nausea, abdominal pain and diarrhoea in recipients of chemotherapy. GIM is a major complication occurring in approximately 80% of patients receiving 5-flurouracil treatment. These side-effects may become so severe that significant dose reductions are required, ultimately affecting treatment efficacy and patient survival. Complementary and alternative medicine (CAM) is a growing area of public interest. This review will provide an overview of current knowledge of complementary medicinal therapies for chemotherapy induced GIM. An understanding of this evolving literature is useful in discussing these therapies with patients who are considering using them.
Collapse
|