1
|
Lee TS. Are Probiotics Beneficial or Harmful for Pancreatic Cancer Outcomes? Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10437-7. [PMID: 39714574 DOI: 10.1007/s12602-024-10437-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Pancreatic cancer is influenced by interactions between cancer cells and the tumor microenvironment (TME), including tumor-infiltrating lymphocytes (TILs). Specifically, CD8 + T cells impact prognosis by eliminating cancer cells. Recent studies have revealed that microbiomes are present in pancreatic tissues and may affect tumor growth and immune responses. Additionally, recent studies revealed that the abundance of Bacteroides, Lactobacillus, and Peptoniphilus are associated with poor pancreatic cancer prognosis. This study investigates the role of oral probiotics in influencing pancreatic cancer outcomes. We retrospectively reviewed patients aged ≥ 18 years with pathologically confirmed pancreatic cancer from Seoul National University Hospital between January 2011 and January 2023. We investigated progression-free survival and overall survival between the control group and the probiotics group. Among pancreatic cancer patients undergoing palliative chemotherapy without radiotherapy and resection, there was a significant difference in overall survival (OS) when comparing the control group to the probiotics group (median: 10 months (9-11) vs. 12 months (9-19), p = 0.026). Regardless of the type of probiotics, oral probiotics may have a positive impact, but further research is still needed to understand the underlying immunological mechanisms.
Collapse
Affiliation(s)
- Tae Seung Lee
- Division of Gastroenterology, Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101, Daehak-Ro, Jongno-Gu, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Qureshi Z, Jamil A, Altaf F, Siddique R. Efficacy and Safety of Probiotics as Adjunctive Therapy in Cancer Treatment: A Comprehensive Systematic Review and Meta-Analysis. Am J Clin Oncol 2024:00000421-990000000-00245. [PMID: 39659264 DOI: 10.1097/coc.0000000000001158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
OBJECTIVES The gut microbiome is crucial in influencing cancer progression and response to treatment. We evaluate the efficacy and safety of probiotics and synbiotics in cancer treatment, focusing on the incidence of diarrhea, significant complications, surgical site infections, length of hospital stay, progression-free survival (PFS), and overall survival (OS). METHODS Following PRISMA guidelines, a comprehensive literature search was conducted in PubMed, Scopus, Web of Science, and Cochrane CENTRAL up to June 2024. The risk of bias was assessed using the Cochrane Risk-of-Bias tool. Meta-analysis was performed using a random-effects model. RESULTS Fifteen studies involving 2197 participants were included. Probiotic use was associated with a significant reduction in the incidence of diarrhea (OR=0.39, 95% CI: 0.15-1.00, P=0.049) with moderate heterogeneity (I2=64%). No significant differences were found in major complications (OR=0.50, 95% CI: 0.05-4.92, P=0.4053, I2=73%), surgical site infections (OR=0.36, 95% CI: 0.12-1.09, P=0.058, I2=0%), length of hospital stay (SMD=-0.30, 95% CI: -1.00 to 0.41, P=0.2726, I2=62%), PFS (HR=0.61, 95% CI: 0.03-10.82, P=0.2715, I2=0%), or OS (HR=0.52, 95% CI: 0.00-58.82, P=0.3298, I2=0%). CONCLUSIONS Probiotics significantly reduced the incidence of chemotherapy-induced diarrhea, highlighting their potential as supportive care agents in oncology. However, their impact on significant complications, surgical site infections, length of hospital stay, and survival outcomes remains inconclusive.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | - Abdur Jamil
- Department of Medicine, Samaritan Medical Centre
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| | | |
Collapse
|
3
|
Li C, Li J. Dysregulation of systemic immunity in colorectal cancer and its clinical applications as biomarkers and therapeutics. Crit Rev Oncol Hematol 2024; 204:104543. [PMID: 39454739 DOI: 10.1016/j.critrevonc.2024.104543] [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: 07/31/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024] Open
Abstract
The immune system plays critical roles in the initiation and progression of colorectal cancer (CRC), and the majority of studies have focused on immune perturbations within the tumor microenvironment. In recent years, systemic immunity, which mainly occurs in the periphery, has attracted much attention. In CRC, both the tumor itself and treatments have extensive effects on systemic immunity, characterized by alterations in circulating cytokines and immune cells. In addition, intact systemic immunity is critical for the efficacy of therapies for CRC, especially immunotherapy. Therefore, various strategies aimed at alleviating the detrimental effects of traditional therapies or directly harnessing the components of systemic immunity for CRC treatment have been developed. However, whether these improvements can translate to survival benefits requires further study. This review aims to comprehensively outline the current knowledge of systemic immunity in CRC.
Collapse
Affiliation(s)
- Changqin Li
- Department of Clinical Laboratory, the Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan, China
| | - Jian Li
- Department of General Surgery, the Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan, China.
| |
Collapse
|
4
|
Wanitsuwan W, Pahumunto N, Surachat K, Thananimit S, Wonglapsuwan M, Laohawiriyakamol S, Teanpaisan R. Comparison of the effects of postbiotics and live-probiotics containing Lacticaseibacillus paracasei SD1 and Lacticaseibacillus rhamnosus SD11 in patients with previous colorectal cancer: A randomized controlled trial. J Funct Foods 2024; 123:106576. [DOI: 10.1016/j.jff.2024.106576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
5
|
Ciernikova S, Sevcikova A, Mego M. Targeting the gut and tumor microbiome in cancer treatment resistance. Am J Physiol Cell Physiol 2024; 327:C1433-C1450. [PMID: 39437444 DOI: 10.1152/ajpcell.00201.2024] [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: 03/28/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Therapy resistance represents a significant challenge in oncology, occurring in various therapeutic approaches. Recently, animal models and an increasing set of clinical trials highlight the crucial impact of the gut and tumor microbiome on treatment response. The intestinal microbiome contributes to cancer initiation, progression, and formation of distant metastasis. In addition, tumor-associated microbiota is considered a critical player in influencing tumor microenvironments and regulating local immune processes. Intriguingly, numerous studies have successfully identified pathogens within the gut and tumor microbiome that might be linked to a poor response to different therapeutic modalities. The unfavorable microbial composition with the presence of specific microbes participates in cancer resistance and progression via several mechanisms, including upregulation of oncogenic pathways, macrophage polarization reprogramming, metabolism of chemotherapeutic compounds, autophagy pathway modulation, enhanced DNA damage repair, inactivation of a proapoptotic cascade, and bacterial secretion of extracellular vesicles, promoting the processes in the metastatic cascade. Targeted elimination of specific intratumoral bacteria appears to enhance treatment response. However, broad-spectrum antibiotic pretreatment is mostly connected to reduced efficacy due to gut dysbiosis and lower diversity. Mounting evidence supports the potential of microbiota modulation by probiotics and fecal microbiota transplantation to improve intestinal dysbiosis and increase microbial diversity, leading to enhanced treatment efficacy while mitigating adverse effects. In this context, further research concerning the identification of clinically relevant microbiome signatures followed by microbiota-targeted strategies presents a promising approach to overcoming immunotherapy and chemotherapy resistance in refractory patients, improving their outcomes.
Collapse
Affiliation(s)
- Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
6
|
Yan J, Yang L, Ren Q, Zhu C, Du H, Wang Z, Qi Y, Xian X, Chen D. Gut microbiota as a biomarker and modulator of anti-tumor immunotherapy outcomes. Front Immunol 2024; 15:1471273. [PMID: 39669573 PMCID: PMC11634861 DOI: 10.3389/fimmu.2024.1471273] [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: 07/27/2024] [Accepted: 10/25/2024] [Indexed: 12/14/2024] Open
Abstract
Although immune-checkpoint inhibitors (ICIs) have significantly improved cancer treatment, their effectiveness is limited by primary or acquired resistance in many patients. The gut microbiota, through its production of metabolites and regulation of immune cell functions, plays a vital role in maintaining immune balance and influencing the response to cancer immunotherapies. This review highlights evidence linking specific gut microbial characteristics to increased therapeutic efficacy in a variety of cancers, such as gastrointestinal cancers, melanoma, lung cancer, urinary system cancers, and reproductive system cancers, suggesting the gut microbiota's potential as a predictive biomarker for ICI responsiveness. It also explores the possibility of enhancing ICI effectiveness through fecal microbiota transplantation, probiotics, prebiotics, synbiotics, postbiotics, and dietary modifications. Moreover, the review underscores the need for extensive randomized controlled trials to confirm the gut microbiota's predictive value and to establish guidelines for microbiota-targeted interventions in immunotherapy. In summary, the article suggests that a balanced gut microbiota is key to maximizing immunotherapy benefits and calls for further research to optimize microbiota modulation strategies for cancer treatment. It advocates for a deeper comprehension of the complex interactions between gut microbiota, host immunity, and cancer therapy, aiming for more personalized and effective treatment options.
Collapse
Affiliation(s)
- Jiexi Yan
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lu Yang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chan Zhu
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Haiyun Du
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhouyu Wang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Yaya Qi
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaohong Xian
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Dongsheng Chen
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Yao B, Wei W, Zhang H. Efficacy of probiotics or synbiotics supplementation on chemotherapy-induced complications and gut microbiota dysbiosis in gastrointestinal cancer: a systematic review and meta-analysis. Eur J Clin Nutr 2024:10.1038/s41430-024-01542-5. [PMID: 39562823 DOI: 10.1038/s41430-024-01542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024]
Abstract
This study aimed to systematically review the clinical efficacy of probiotics or synbiotics supplementation in the treatment of chemotherapy-induced complications and gut microbiota dysbiosis in patients with gastrointestinal cancer. A literature search was performed systematically using PubMed, Embase, Cochrane, Web of Science, Wanfang Data, and CNKI for randomized controlled trials of probiotics or synthetic supplementation on chemotherapy-induced complications and gut microbiota dysbiosis in gastrointestinal cancer up to December 2023. The outcome measures included chemotherapy-related complications and the the incidence of gut microbiotas. Fifteen studies were finally eligible for meta-analysis, involving 1356 patients. Meta-analysis results showed that the the incidence rates of chemotherapy-related complications such as nausea and vomiting [RR = 0.61, 95% CI (0.46,0.82), P = 0.0008] and diarrhea [RR = 0.47, 95% CI (0.32,0.68), P < 0.001] were significantly reduced after probiotic intervention. The number of intestinal flora changed significantly after intervention, such as bifidobacterium [SMD = 1.33, 95% CI (0.52,2.31), P = 0.001], Escherichia coli [SMD = -0.82, 95% CI (-1.26, -0.38), P = 0.0003], and the difference was statistically significant. Probiotics or synbiotics supplementation can reduce chemotherapy-induced complications in patients with gastrointestinal cancer and regulate the number of gut microbiotas to balance the intestinal microecology of the body.
Collapse
Affiliation(s)
- Bei Yao
- First school of clinical medicine, Shandong Traditional Chinese Medicine University, Jinan Shandong, 250014, China
| | - Wei Wei
- Acupuncture rehabilitation Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, GuangDong, 510006, China
| | - Huiping Zhang
- Oncology department, Jinan Hospital of Traditional Chinese Medicine, Jinan Shandong, 250012, China.
| |
Collapse
|
8
|
Dan X, Tian YL, Huang Y, He YL, Ren JH. Nutritional status and daily habits as determinants of hospitalization duration in ovarian cancer patients undergoing chemotherapy. Sci Rep 2024; 14:27841. [PMID: 39537687 PMCID: PMC11561155 DOI: 10.1038/s41598-024-78941-y] [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: 02/21/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Adequate nutrition in a hospital setting is essential for achieving optimal health outcomes in oncology patients. This study specifically investigated the interrelationships between nutritional status, daily habits, and hospital length of stay (LOS) in ovarian cancer patients undergoing chemotherapy. A prospective longitudinal study was conducted from August 2019 to January 2022 in a tertiary hospital. Throughout the study, nutritional status, biochemical indicators, diet, and physical activity were meticulously recorded at different stages of chemotherapy: before chemotherapy (T0), the first course (T1), third course (T2), and fifth course (T3). To determine the factors influencing LOS, a generalized estimation equation (GEE) was employed. A total of 460 patients completed the follow-up period. The findings revealed a decline in nutritional risk among patients by 9.90% at T1, 17.62% at T2 and 18.26% at T3 (χ2 = 79.220, P < 0.001). The proportion of people receiving enteral nutrition showed an upward trend (χ2 = 15.202, P < 0.001). Notably, the proportion of patients adhering to a healthier diet increased by 40.44% by the study's conclusion, while the number of patients abstaining from physical activity or engaging in solely low-intensity activities decreased by 21.08%. Moreover, as the chemotherapy cycle progressed, daily activity steps exhibited an upward trajectory (F = 5.986, P < 0.001), while the LOS experienced a significant reduction (F = 21.298, P < 0.001). This study identified hypoproteinemia (protein level < 34 g/L), a high nutritional risk (NRS 2002 score ≥ 3), a short duration of sleep (≤ 7 h/day), and a lower daily activity level as risk factors for LOS. Receiving enteral nutrition support is a protective factor for LOS. Significant improvements in nutritional status, diet, and physical activity have been observed among ovarian cancer patients during their chemotherapy cycles. Reduced nutritional risks, implementation of nutritional support, good physical activity, and adequate sleep were associated with a shorter LOS.
Collapse
Affiliation(s)
- Xin Dan
- Department of Radiation Therapy and Chemotherapy for Cancer Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, Sichuan, China
| | - Ya-Lin Tian
- Department of Radiation Therapy and Chemotherapy for Cancer Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, Sichuan, China
| | - Yan Huang
- Department of Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, Sichuan, China
| | - Ya-Lin He
- Department of Radiation Therapy and Chemotherapy for Cancer Nursing, West China Second University Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, Sichuan, China.
| | - Jian-Hua Ren
- Department of Obstetrics and Gynecology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
9
|
Kim S, Na GH, Yim DJ, Liu CF, Lin TH, Shih TW, Pan TM, Lee CL, Koo YK. Lactobacillus paracasei subsp. paracasei NTU 101 prevents obesity by regulating AMPK pathways and gut microbiota in obese rat. Biochem Biophys Res Commun 2024; 731:150279. [PMID: 39018972 DOI: 10.1016/j.bbrc.2024.150279] [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: 03/27/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 07/19/2024]
Abstract
This study assessed the anti-obesity effects of Lactobacillus paracasei subsp. paracasei NTU 101 (NTU 101) both in vitro and in vivo. Initially, the cytotoxicity and lipid accumulation inhibitory effects of NTU 101 on 3T3-L1 cells were evaluated using the MTT assay and oil red O assay, respectively. Subsequently, the anti-obesity effects of NTU 101 were investigated in high-fat diet-induced obese rat. Moreover, western blotting was performed to measure the obesity-related protein expression of PPARα, PPARβ, PPARγ, C/EBPα, C/EBPβ, ATGL, p-p38 MAPK, p-ERK1/2, p-AMPK and CPT-1 in both 3T3-L1 adipocytes and adipose and liver tissues. Treatment with 16 × 108 CFU/mL NTU 101 reduced lipid accumulation in 3T3-L1 adipocytes by more than 50 %. Oral administration of NTU 101 significantly attenuated body weight gain, as well as adipose tissue weight. NTU 101 administration enhanced fatty acid oxidation increasing expression levels of PPARα, CPT-1, and p-AMPK proteins in liver tissue, while simultaneously inhibited adipogenesis by reducing PPARγ and C/EBPα proteins in adipose tissue. Furthermore, NTU 101 supplementation positively modulated the composition of gut microbiota, notably increasing the abundance of Akkermansiaceae. This present study suggests that NTU 101 exerts anti-obesity effects by regulating gut microbiota, fatty acid oxidation, lipolysis and adipogenesis.
Collapse
Affiliation(s)
- SukJin Kim
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Gwi Hwan Na
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Dong Joon Yim
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Chin-Feng Liu
- Continuing Education Program of Food Biotechnology Applications, National Taitung University, Taitung, Taiwan, ROC
| | - Tse-Han Lin
- Department of Life Science, National Taitung University, 369, Sec. 2, University Rd., Taitung, 95092, Taitung, Taiwan, ROC
| | | | - Tzu-Ming Pan
- SunWay Biotech Co. LTD., Taipei, Taiwan, ROC; Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Chun-Lin Lee
- Continuing Education Program of Food Biotechnology Applications, National Taitung University, Taitung, Taiwan, ROC.
| | - Yean Kyoung Koo
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea.
| |
Collapse
|
10
|
Xu Y, Wu X, Li Y, Liu X, Fang L, Jiang Z. Probiotics and the Role of Dietary Substrates in Maintaining the Gut Health: Use of Live Microbes and Their Products for Anticancer Effects against Colorectal Cancer. J Microbiol Biotechnol 2024; 34:1933-1946. [PMID: 39210613 PMCID: PMC11540615 DOI: 10.4014/jmb.2403.03056] [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: 03/27/2024] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 09/04/2024]
Abstract
The gut microbiome is an important and the largest endocrine organ linked to the microbes of the GI tract. The bacterial, viral and fungal communities are key regulators of the health and disease status in a host at hormonal, neurological, immunological, and metabolic levels. The useful microbes can compete with microbes exhibiting pathogenic behavior by maintaining resistance against their colonization, thereby maintaining eubiosis. As diagnostic tools, metagenomic, proteomic and genomic approaches can determine various microbial markers in clinic for early diagnosis of colorectal cancer (CRC). Probiotics are live non-pathogenic microorganisms such as lactic acid bacteria, Bifidobacteria, Firmicutes and Saccharomyces that can help maintain eubiosis when administered in appropriate amounts. In addition, the type of dietary intake contributes substantially to the composition of gut microbiome. The use of probiotics has been found to exert antitumor effects at preclinical levels and promote the antitumor effects of immunotherapeutic drugs at clinical levels. Also, modifying the composition of gut microbiota by Fecal Microbiota Transplantation (FMT), and using live lactic acid producing bacteria such as Lactobacillus, Bifidobacteria and their metabolites (termed postbiotics) can contribute to immunomodulation of the tumor microenvironment. This can lead to tumor-preventive effects at early stages and antitumor effects after diagnosis of CRC. To conclude, probiotics are presumably found to be safe to use in humans and are to be studied further to promote their appliance at clinical levels for management of CRC.
Collapse
Affiliation(s)
- Yi Xu
- Phase I Clinical Cancer Trial Center, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, P.R. China
| | - Xiahui Wu
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang 222002, P.R. China
- Department of Oncology, The First People’s Hospital of Lianyungang, Lianyungang 222002, P.R. China
| | - Yan Li
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang 222002, P.R. China
- Department of Oncology, The First People’s Hospital of Lianyungang, Lianyungang 222002, P.R. China
| | - Xuejie Liu
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang 222002, P.R. China
- Department of Oncology, The First People’s Hospital of Lianyungang, Lianyungang 222002, P.R. China
| | - Lijian Fang
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang 222002, P.R. China
- Department of Oncology, The First People’s Hospital of Lianyungang, Lianyungang 222002, P.R. China
| | - Ziyu Jiang
- Phase I Clinical Cancer Trial Center, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, P.R. China
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang 222002, P.R. China
- Department of Oncology, The First People’s Hospital of Lianyungang, Lianyungang 222002, P.R. China
| |
Collapse
|
11
|
Zhong Y, Liu Z, Wang Y, Cai S, Qiao Z, Hu X, Wang T, Yi J. Preventive Methods for Colorectal Cancer Through Dietary Interventions: A Focus on Gut Microbiota Modulation. FOOD REVIEWS INTERNATIONAL 2024:1-29. [DOI: 10.1080/87559129.2024.2414908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Yujie Zhong
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory for Plateau Food Advanced Manufacturing, Kunming University of Science and Technology, Kunming, China
- International Green Food Processing Research and Development Center of Kunming City, Kunming University of Science and Technology, Kunming, China
| | - Zhijia Liu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory for Plateau Food Advanced Manufacturing, Kunming University of Science and Technology, Kunming, China
- International Green Food Processing Research and Development Center of Kunming City, Kunming University of Science and Technology, Kunming, China
| | - Yanfei Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory for Plateau Food Advanced Manufacturing, Kunming University of Science and Technology, Kunming, China
- International Green Food Processing Research and Development Center of Kunming City, Kunming University of Science and Technology, Kunming, China
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory for Plateau Food Advanced Manufacturing, Kunming University of Science and Technology, Kunming, China
- International Green Food Processing Research and Development Center of Kunming City, Kunming University of Science and Technology, Kunming, China
| | - Zhu Qiao
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian, Henan Province, China
| | - Xiaosong Hu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory for Plateau Food Advanced Manufacturing, Kunming University of Science and Technology, Kunming, China
- International Green Food Processing Research and Development Center of Kunming City, Kunming University of Science and Technology, Kunming, China
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Tao Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory for Plateau Food Advanced Manufacturing, Kunming University of Science and Technology, Kunming, China
- International Green Food Processing Research and Development Center of Kunming City, Kunming University of Science and Technology, Kunming, China
| | - Junjie Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory for Plateau Food Advanced Manufacturing, Kunming University of Science and Technology, Kunming, China
- International Green Food Processing Research and Development Center of Kunming City, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
12
|
Hu Y, Zhou P, Deng K, Zhou Y, Hu K. Targeting the gut microbiota: a new strategy for colorectal cancer treatment. J Transl Med 2024; 22:915. [PMID: 39379983 PMCID: PMC11460241 DOI: 10.1186/s12967-024-05671-0] [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: 11/18/2023] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND How to reduce the high incidence rate and mortality of colorectal cancer (CRC) effectively is the focus of current research. Endoscopic treatment of early-stage CRC and colorectal adenomas (CAC) has a high success rate, but although several treatments are available for advanced CRC, such as surgery, radiotherapy, chemotherapy, and immunotherapy, the 5-year survival rate remains low. In view of the high incidence rate and mortality of CRC, early rational drug prevention for high-risk groups and exploration of alternative treatment modalities are particularly warranted. Gut microbiota is the target of and interacts with probiotics, prebiotics, aspirin, metformin, and various Chinese herbal medicines (CHMs) for the prevention of CRC. In addition, the anti-cancer mechanisms of probiotics differ widely among bacterial strains, and both bacterial strains and their derivatives and metabolites have been found to have anti-cancer effects. Gut microbiota plays a significant role in early drug prevention of CRC and treatment of CRC in its middle and late stages, targeting gut microbiota may be a new strategy for colorectal cancer treatment.
Collapse
Affiliation(s)
- Yue Hu
- Health Science Center, Ningbo University, Ningbo, China
| | - Peng Zhou
- Health Science Center, Ningbo University, Ningbo, China
| | - Kaili Deng
- Health Science Center, Ningbo University, Ningbo, China
| | - Yuping Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China.
- Institute of Digestive Disease of Ningbo University, Ningbo, China.
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Ningbo, China.
| | - Kefeng Hu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
13
|
Islam MM, Mahbub NU, Hong ST, Chung HJ. Gut bacteria: an etiological agent in human pathological conditions. Front Cell Infect Microbiol 2024; 14:1291148. [PMID: 39439902 PMCID: PMC11493637 DOI: 10.3389/fcimb.2024.1291148] [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: 09/08/2023] [Accepted: 08/12/2024] [Indexed: 10/25/2024] Open
Abstract
Through complex interactions with the host's immune and physiological systems, gut bacteria play a critical role as etiological agents in a variety of human diseases, having an impact that extends beyond their mere presence and affects the onset, progression, and severity of the disease. Gaining a comprehensive understanding of these microbial interactions is crucial to improving our understanding of disease pathogenesis and creating tailored treatment methods. Correcting microbial imbalances may open new avenues for disease prevention and treatment approaches, according to preliminary data. The gut microbiota exerts an integral part in the pathogenesis of numerous health conditions, including metabolic, neurological, renal, cardiovascular, and gastrointestinal problems as well as COVID-19, according to recent studies. The crucial significance of the microbiome in disease pathogenesis is highlighted by this role, which is comparable to that of hereditary variables. This review investigates the etiological contributions of the gut microbiome to human diseases, its interactions with the host, and the development of prospective therapeutic approaches. To fully harness the benefits of gut microbiome dynamics for improving human health, future research should address existing methodological challenges and deepen our knowledge of microbial interactions.
Collapse
Affiliation(s)
- Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
14
|
Wu YJ, Xiong JF, Zhan CN, Xu H. Gut microbiota alterations in colorectal adenoma-carcinoma sequence based on 16S rRNA gene sequencing: A systematic review and meta-analysis. Microb Pathog 2024; 195:106889. [PMID: 39197689 DOI: 10.1016/j.micpath.2024.106889] [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: 04/29/2024] [Revised: 08/12/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Most sporadic colorectal cancers (CRC) develop through the adenoma-carcinoma sequence. While dysbiosis of the intestinal flora contributes to CRC's pathogenesis, precise microbial taxa closely associated with the colorectal adenoma-carcinoma sequence remain elusive. This meta-analysis aimed to summarize the features of intestinal flora in patients with AD and CRC. METHODS PubMed, Embase, Cochrane Library, and Web of Science were searched for case-control studies comparing the relative abundance of gut microbiota in the feces of patients with AD, CRC, and healthy controls (HC) from inception to January 2024. The weighted mean difference (WMD) with a 95 % confidence interval (CI) was used to display the results. The Newcastle-Ottawa Scale (NOS) was used to assess the quality of the entailed literature. Publication bias was evaluated with the Egger's and Begg's tests. RESULTS Eleven studies were included, involving 477 CRC patients, 628 AD patients, and 864 healthy controls. Compared with HC, the patients with AD had a significantly lower Chao 1 index (WMD = -30.17, 95 % CI [-41.10, -19.23], P < 0.001) and Shannon index (WMD = -0.11 95 % CI [-0.18, -0.04], P = 0.002). Compared with AD, the CRC patients had a significantly higher Chao1 index (WMD = 22.09, 95 % CI [7.59, 36.00], P = 0.003) and Shannon index (WMD = 0.08, 95 % CI [0.00, 0.15], P = 0.037). Enterobacteriaceae (WMD = 0.03 95 % CI [0.00,0.05], P = 0.047; WMD = 0.02 95 % CI [0.00,0.04], P = 0.027) significantly increased in the order of Control-AD-CRC, while that of Blautia (WMD = -0.00 95 % CI [-0.01, -0.00], P = 0.001; WMD = -0.00 95 % CI [-0.00, -0.00], P = 0.002) was reduced. Compared with HC, the relative abundance of Proteobacteria (WMD = 0.05 95 % CI [0.03,0.07], P < 0.001), Fusobacteria (WMD = 0.02 95 % CI [0.00,0.03], P = 0.042), Streptococcaceae (WMD = 0.03 95 % CI [0.01,0.05], P = 0.017), Prevotellaceae (WMD = 0.02 95 % CI [0.00,0.04], P = 0.040), and Escherichia-Shigella (WMD = 0.06 95 % CI [0.01, 0.11], P = 0.021) was enriched in the CRC group. The relative abundance of Alistipes (WMD = 0.00 95 % CI [0.00,0.01], P = 0.032) and Streptococcus (WMD = 0.00 95 % CI [0.00,0.00], P = 0.001) was increased in the AD vs HC. The relative abundance of Firmicutes (WMD = -0.07 95 % CI [-0.12, -0.03], P = 0.003), Bifidobacteria (WMD = -0.03 95 % CI [-0.05, -0.01], P = 0.016), and Klebsiella (WMD = -0.01 95 % CI [-0.01, -0.00], P = 0.001) was decreased in the CRC vs HC. Compared with AD, the relative abundance of Firmicutes (WMD = -0.04 95 % CI [-0.07, -0.02], P = 0.002), Peptostreptococcaceae (WMD = -0.03 95 % CI [-0.05, -0.00], P = 0.021), Lachnospiraceae (WMD = -0.04 95 % CI [-0.08,-0.00], P = 0.037), Ruminococcaceae (WMD = -0.06 95 % CI [-0.09,-0.03], P < 0.001), Faecalibacterium (WMD = -0.01 95 % CI [-0.02, -0.01], P = 0.001), and Lachnoclostridium (WMD = -0.02 95 % CI [-0.03, -0.00], P = 0.040) was decreased in the CRC group, while Proteobacteria (WMD = 0.04 95 % CI [0.02,0.05], P < 0.001) was increased. CONCLUSIONS The dysbiosis characterized by reduced levels of short-chain fatty acid (SCFA)-producing bacteria, decreased anti-inflammatory bacteria, increased pro-inflammatory bacteria, and an elevation of bacteria with cytotoxic effects damaging to DNA may represent the specific microbial signature of colorectal adenoma/carcinoma. Further research is required to elucidate the mechanisms by which gut dysbiosis leads to the progression from AD to CRC and to explore the potential of specific microbiota markers in clinical treatment and non-invasive screening.
Collapse
Affiliation(s)
- Yi-Jun Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing-Fang Xiong
- Department of Geriatrics, Hangzhou Red Cross Hospital, Hangzhou, China
| | - Cheng-Nan Zhan
- Medical Service Community, Hangzhou Xiaoshan Hospital of TCM, Hangzhou, China
| | - Hong Xu
- Department of Gastroenterology and Hepatology, Hangzhou Red Cross Hospital, Hangzhou, China.
| |
Collapse
|
15
|
Sudaarsan ASK, Ghosh AR. Appraisal of postbiotics in cancer therapy. Front Pharmacol 2024; 15:1436021. [PMID: 39372197 PMCID: PMC11449718 DOI: 10.3389/fphar.2024.1436021] [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: 05/21/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Cancer remains a multifactorial disease with an increased mortality rate around the world for the past several decades. Despite advancements in treatment strategies, lower survival rates, drug-associated side effects, and drug resistance create a need for novel anticancer agents. Ample evidence shows that imbalances in the gut microbiota are associated with the formation of cancer and its progression. Altering the gut microbiota via probiotics and their metabolites has gained attention among the research community as an alternative therapy to treat cancer. Probiotics exhibit health benefits as well as modulate the immunological and cellular responses in the host. Apart from probiotics, their secreted products like bacteriocins, exopolysaccharides, short-chain fatty acids, conjugated linoleic acid, peptidoglycan, and other metabolites are found to possess anticancer activity. The beneficiary role of these postbiotic compounds is widely studied for characterizing their mechanism and mode of action that reduces cancer growth. The present review mainly focuses on the postbiotic components that are employed against cancer with their reported mechanism of action. It also describes recent research works carried out so far with specific strain and anticancer activity of derived compounds both in vitro and in vivo, validating that the probiotic approach would pave an alternative way to reduce the burden of cancer.
Collapse
|
16
|
Le Ngoc K, Pham TTH, Nguyen TK, Huong PT. Pharmacomicrobiomics in precision cancer therapy: bench to bedside. Front Immunol 2024; 15:1428420. [PMID: 39315107 PMCID: PMC11416994 DOI: 10.3389/fimmu.2024.1428420] [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: 05/08/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The burgeoning field of pharmacomicrobiomics offers promising insights into the intricate interplay between the microbiome and cancer, shaping responses to diverse treatment modalities. This review aims to analyze the molecular mechanisms underlying interactions between distinct microbiota types and cancer, as well as their influence on treatment outcomes. We explore how the microbiome impacts antitumor immunity, and response to chemotherapy, immunotherapy, and radiation therapy, unveiling its multifaceted roles in cancer progression and therapy resistance. Moreover, we discuss the challenges hindering the development of microbiome-based interventions in cancer therapy, including standardization, validation, and clinical translation. By synthesizing clinical evidence, we underscore the transformative potential of harnessing pharmacomicrobiomics in guiding cancer treatment decisions, paving the way for improved patient outcomes in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Phung Thanh Huong
- Faculty of Biotechnology, Hanoi University of Pharmacy,
Hanoi, Vietnam
| |
Collapse
|
17
|
Deckey DG, Boddu SP, Verhey JT, Doxey SA, Spangehl MJ, Clarke HD, Bingham JS. Clostridium difficile Infection Prior to Total Hip Arthroplasty Independently Increases the Risk of Periprosthetic Joint Infection. J Arthroplasty 2024; 39:S444-S448.e1. [PMID: 38548233 DOI: 10.1016/j.arth.2024.03.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Periprosthetic joint infection (PJI) following total hip arthroplasty (THA) is associated with major morbidity. There may be a link between the gut microbiome and an individual's overall immune system. A Clostridium difficile (C. difficile) infection portends poor gut microbiome health and has been previously associated with increased 90-day complication rates in total joint arthroplasty (TJA). The purpose of this study was to determine the effect of a previous history of C. difficile infection within 2 years of undergoing THA on PJI within 2 years postoperatively. METHODS Patients undergoing THA from 2010 to 2021 were identified in a patient claims database (n = 770,075). Patients who had active records 2 years before and after THA as well as a history of C. difficile infection within 2 years prior to THA (n = 1,836) were included and propensity matched to a control group using age, sex, and Elixhauser comorbidity index. The primary outcome was the 2-year incidence of postoperative PJI. The exposed C. difficile infection cohort was stratified into 4 groups based on the time proximity of the C. difficile infection. Chi-square tests and logistic regressions were used to compare the groups. RESULTS A C. difficile infection anytime within 2 years prior to total hip arthroplasty was independently associated with higher odds of PJI (OR [odds ratio]: 1.49 [95% CI (confidence interval) 1.09 to 2.02, P = .014]). Proximity of C. difficile infection to arthroplasty was associated with increased risk of PJI (infection 0 to 3 months before THA: OR 2.01 [95% CI 1.23 to 3.20], infection 3 to 6 months before THA: OR 1.84 [95% CI 1.06 to 3.04], infection 6 to 12 months before THA: OR 1.10 [95% CI 0.65 to 1.77], infection 1 to 2 years before THA: OR 1.40 [95% CI 0.94 to 2.06]). CONCLUSIONS A C. difficile infection prior to THA is an independent risk factor for PJI. Proximity of C. difficile infection is associated with increased risk of PJI. Future investigations should evaluate how to adequately optimize patients prior to THA and pursue strategies to determine appropriate timing for proceeding with THA.
Collapse
Affiliation(s)
- David G Deckey
- Department of Orthopaedic Surgery, Mayo Clinic Arizona, Phoenix, Arizona
| | - Sayi P Boddu
- Alix School of Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Jens T Verhey
- Department of Orthopaedic Surgery, Mayo Clinic Arizona, Phoenix, Arizona
| | - Stephen A Doxey
- Department of Orthopaedic Surgery, TRIA Orthopaedic Institute, Bloomington, Minnesota
| | - Mark J Spangehl
- Department of Orthopaedic Surgery, Mayo Clinic Arizona, Phoenix, Arizona
| | - Henry D Clarke
- Department of Orthopaedic Surgery, Mayo Clinic Arizona, Phoenix, Arizona
| | - Joshua S Bingham
- Department of Orthopaedic Surgery, Mayo Clinic Arizona, Phoenix, Arizona
| |
Collapse
|
18
|
Cheraghpour M, Fatemi N, Shadnoush M, Talebi G, Tierling S, Bermúdez-Humarán LG. Immunomodulation aspects of gut microbiome-related interventional strategies in colorectal cancer. Med Oncol 2024; 41:231. [PMID: 39162936 DOI: 10.1007/s12032-024-02480-9] [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] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
Colorectal cancer (CRC), the third most common cancer worldwide, develops mainly due to the accumulation of genetic and epigenetic changes over many years. Substantial evidence suggests that gut microbiota plays a significant role in the initiation, progression, and control of CRC, depending on the balance between beneficial and pathogenic microorganisms. Nonetheless, gut microbiota composition by regulating the host immune response may either promote or inhibit CRC. Thus, modification of gut microbiota potentially impacts clinical outcomes of immunotherapy. Previous studies have indicated that therapeutic strategies such as probiotics, prebiotics, and postbiotics enhance the intestinal immune system and improve the efficacy of immunotherapeutic agents, potentially serving as a complementary strategy in cancer immunotherapy. This review discusses the role of the gut microbiota in the onset and development of CRC in relation to the immune response. Additionally, we focus on the effect of strategies manipulating gut microbiome on the immune response and efficacy of immunotherapy against CRC. We demonstrate that manipulation of gut microbiome can enhance immune response and outcomes of immunotherapy through downregulating Treg cells and other immunosuppressive cells while improving the function of T cells within the tumor; however, further research, especially clinical trials, are needed to evaluate its efficacy in cancer treatment.
Collapse
Affiliation(s)
- Makan Cheraghpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Shadnoush
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Talebi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sascha Tierling
- Department of Genetics/Epigenetics, Faculty NT, Life Sciences, Saarland University, Saarbrücken, Germany
| | - Luis G Bermúdez-Humarán
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350, Jouy-en-Josas, France.
| |
Collapse
|
19
|
Su ACY, Ding X, Lau HCH, Kang X, Li Q, Wang X, Liu Y, Jiang L, Lu Y, Liu W, Ding Y, Cheung AHK, To KF, Yu J. Lactococcus lactis HkyuLL 10 suppresses colorectal tumourigenesis and restores gut microbiota through its generated alpha-mannosidase. Gut 2024; 73:1478-1488. [PMID: 38599786 PMCID: PMC11347254 DOI: 10.1136/gutjnl-2023-330835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 03/29/2024] [Indexed: 04/12/2024]
Abstract
OBJECTIVE Probiotic Lactococcus lactis is known to confer health benefits to humans. Here, we aimed to investigate the role of L. lactis in colorectal cancer (CRC). DESIGN L. lactis abundance was evaluated in patients with CRC (n=489) and healthy individuals (n=536). L. lactis was isolated from healthy human stools with verification by whole genome sequencing. The effect of L. lactis on CRC tumourigenesis was assessed in transgenic Apc Min/+ mice and carcinogen-induced CRC mice. Faecal microbiota was profiled by metagenomic sequencing. Candidate proteins were characterised by nano liquid chromatography-mass spectrometry. Biological function of L. lactis conditioned medium (HkyuLL 10-CM) and functional protein was studied in human CRC cells, patient-derived organoids and xenograft mice. RESULTS Faecal L. lactis was depleted in patients with CRC. A new L. lactis strain was isolated from human stools and nomenclated as HkyuLL 10. HkyuLL 10 supplementation suppressed CRC tumourigenesis in Apc Min/+ mice, and this tumour-suppressing effect was confirmed in mice with carcinogen-induced CRC. Microbiota profiling revealed probiotic enrichment including Lactobacillus johnsonii in HkyuLL 10-treated mice. HkyuLL 10-CM significantly abrogated the growth of human CRC cells and patient-derived organoids. Such protective effect was attributed to HkyuLL 10-secreted proteins, and we identified that α-mannosidase was the functional protein. The antitumourigenic effect of α-mannosidase was demonstrated in human CRC cells and organoids, and its supplementation significantly reduced tumour growth in xenograft mice. CONCLUSION HkyuLL 10 suppresses CRC tumourigenesis in mice through restoring gut microbiota and secreting functional protein α-mannosidase. HkyuLL 10 administration may serve as a prophylactic measure against CRC.
Collapse
Affiliation(s)
- Anthony Chin Yang Su
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao Ding
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xing Kang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Qing Li
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xueliang Wang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Yali Liu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Lanping Jiang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Yinghong Lu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Weixin Liu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Yanqiang Ding
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Bakrim S, El Hachlafi N, Khalid A, Abdalla AN, El Omari N, Aboulaghras S, Sakran AM, Goh KW, Ming LC, Razi P, Bouyahya A. Recent advances and molecular mechanisms of TGF-β signaling in colorectal cancer, with focus on bioactive compounds targeting. Biomed Pharmacother 2024; 177:116886. [PMID: 38945700 DOI: 10.1016/j.biopha.2024.116886] [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: 03/03/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most significant forms of human cancer. It is characterized by its heterogeneity because several molecular factors are involved in contiguity and can link it to others without having a linear correlation. Among the factors influencing tumor transformation in CRC, transforming growth factor-beta (TGF-β) plays a key promoter role. This factor is associated with human colorectal tumors with a very high prognosis: it increases the survival, invasion, and metastasis of CRC cells, thus functioning as an oncogene. The inhibition of this factor can constitute a major therapeutic route for CRC treatment. Various chemical drugs including synthetic molecules and biotherapies have been developed as TGF-β inhibitors. Moreover, the scientific community has recently shown a major interest in screening natural drugs inhibiting TGF-β in CRC. In this context, we carried out this review article using computerized databases, such as PubMed, Google Scholar, Springer Link, Science Direct, Cochrane Library, Embase, Web of Science, and Scopus, to highlight the molecular mechanism of TGF-β in CRC induction and progression and current advances in the pharmacodynamic effects of natural bioactive substances targeting TGF-β in CRC.
Collapse
Affiliation(s)
- Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Naoufal El Hachlafi
- Microbial Biotechnology and Bioactive Molecules Laboratory, Sciences and Technologies Faculty, Sidi Mohmed Ben Abdellah University, P.O.Box-2002, Imouzzer Road, Fez, Morocco
| | - Asaad Khalid
- Health Research Center, Jazan University, P.O. Box: 114, Jazan 45142, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| | - Sara Aboulaghras
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Ashraf M Sakran
- Department of Anatomy, Faculty of Medicine, Umm Alqura University, Makkah 21955, Saudi Arabia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia.
| | - Pakhrur Razi
- Center of Disaster Monitoring and Earth Observation, Universitas Negeri Padang, Padang, Indonesia.
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| |
Collapse
|
21
|
Kalam N, Balasubramaniam VRMT. Crosstalk between COVID-19 and the gut-brain axis: a gut feeling. Postgrad Med J 2024; 100:539-554. [PMID: 38493312 DOI: 10.1093/postmj/qgae030] [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: 12/10/2023] [Accepted: 02/15/2024] [Indexed: 03/18/2024]
Abstract
The microbes in the gut are crucial for maintaining the body's immune system and overall gut health. However, it is not fully understood how an unstable gut environment can lead to more severe cases of SARS-CoV-2 infection. The gut microbiota also plays a role in the gut-brain axis and interacts with the central nervous system through metabolic and neuroendocrine pathways. The interaction between the microbiota and the host's body involves hormonal, immune, and neural pathways, and any disruption in the balance of gut bacteria can lead to dysbiosis, which contributes to pathogen growth. In this context, we discuss how dysbiosis could contribute to comorbidities that increase susceptibility to SARS-CoV-2. Probiotics and fecal microbiota transplantation have successfully treated infectious and non-infectious inflammatory-related diseases, the most common comorbidities. These treatments could be adjuvant therapies for COVID-19 infection by restoring gut homeostasis and balancing the gut microbiota.
Collapse
Affiliation(s)
- Nida Kalam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Malaysia
| | - Vinod R M T Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Malaysia
| |
Collapse
|
22
|
Ermolenko E, Baryshnikova N, Alekhina G, Zakharenko A, Ten O, Kashchenko V, Novikova N, Gushchina O, Ovchinnikov T, Morozova A, Ilina A, Karaseva A, Tsapieva A, Gladyshev N, Dmitriev A, Suvorov A. Autoprobiotics in the Treatment of Patients with Colorectal Cancer in the Early Postoperative Period. Microorganisms 2024; 12:980. [PMID: 38792809 PMCID: PMC11124500 DOI: 10.3390/microorganisms12050980] [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: 02/23/2024] [Revised: 04/26/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Despite great advances in the treatment of oncological diseases, the development of medical technologies to prevent or reduce complications of therapy, in particular, those associated with surgery and the introduction of antibiotics, remains relevant. The aim of this study is to evaluate the effectiveness of the use of autoprobiotics based on indigenous non-pathogenic strains of Enterococcus faecium and Enterococcus hirae as a personalized functional food product (PFFP) in the complex therapy of colorectal cancer (CRC) in the early postoperative period. A total of 36 patients diagnosed with CRC were enrolled in the study. Study group A comprised 24 CRC patients who received autoprobiotic therapy in the early postoperative period, while the control group C included 12 CRC patients without autoprobiotic therapy. Prior to surgery and between days 14 and 16 post-surgery, comprehensive evaluations were conducted on all patients, encompassing the following: stool and gastroenterological complaints analysis, examination of the gut microbiota (bacteriological study, quantitative polymerase chain reaction, metagenome analysis), and analysis of interleukins in the serum. Results: The use of autoprobiotics led to a decrease in dyspeptic complaints after surgery. It was also associated with the absence of postoperative complications, did not cause any side effects, and led to a decrease in the level of pro-inflammatory cytokines (IL-6 and IL-18) in the blood serum. The use of autoprobiotics led to positive changes in the structure of escherichia and enterococci populations, the elimination of Parvomonas micra and Fusobacterium nucleatum, and a decrease in the quantitative content of Clostridium perfringens and Akkermansia muciniphila. Metagenomic analysis (16S rRNA) revealed an increase in alpha diversity. Conclusion: The introduction of autoprobiotics in the postoperative period is a highly effective and safe approach in the complex treatment of CRC. Future studies will allow the discovery of additional fine mechanisms of autoprobiotic therapy and its impact on the digestive, immune, endocrine, and neural systems.
Collapse
Affiliation(s)
- Elena Ermolenko
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Natalia Baryshnikova
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
- Department of Internal Disease of Stomatology Faculty, Pavlov First St-Petersburg State Medical University, 197022 St-Petersburg, Russia
- Laboratory of Medico-Social Problems of Pediatry, St-Petersburg State Pediatric Medical University, 194100 St-Petersburg, Russia
| | - Galina Alekhina
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Alexander Zakharenko
- Oncology Department, Pavlov First St-Petersburg State Medical University, 197022 St-Petersburg, Russia;
| | - Oleg Ten
- North-Western District Scientific and Clinical Center Named after L. G. Sokolov, 194291 St-Petersburg, Russia (O.G.)
| | - Victor Kashchenko
- Department of Faculty Surgery, St-Petersburg State University, 199034 St-Petersburg, Russia;
- Beloostrov High Technology Clinic (MMC VT LLC), 188652 Leningrad Region, Russia
| | - Nadezhda Novikova
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Olga Gushchina
- North-Western District Scientific and Clinical Center Named after L. G. Sokolov, 194291 St-Petersburg, Russia (O.G.)
| | - Timofey Ovchinnikov
- North-Western District Scientific and Clinical Center Named after L. G. Sokolov, 194291 St-Petersburg, Russia (O.G.)
| | - Anastasia Morozova
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Anastasia Ilina
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Alena Karaseva
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
- Microbiology Department, St-Petersburg State University, 199034 St-Petersburg, Russia
| | - Anna Tsapieva
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Nikita Gladyshev
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Alexander Dmitriev
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
- Department of Molecular Biotechnology, Saint-Petersburg State Institute of Technology, 190013 St-Petersburg, Russia
| | - Alexander Suvorov
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| |
Collapse
|
23
|
Profir M, Roşu OA, Creţoiu SM, Gaspar BS. Friend or Foe: Exploring the Relationship between the Gut Microbiota and the Pathogenesis and Treatment of Digestive Cancers. Microorganisms 2024; 12:955. [PMID: 38792785 PMCID: PMC11124004 DOI: 10.3390/microorganisms12050955] [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: 03/11/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Digestive cancers are among the leading causes of cancer death in the world. However, the mechanisms of cancer development and progression are not fully understood. Accumulating evidence in recent years pointing to the bidirectional interactions between gut dysbiosis and the development of a specific type of gastrointestinal cancer is shedding light on the importance of this "unseen organ"-the microbiota. This review focuses on the local role of the gut microbiota imbalance in different digestive tract organs and annexes related to the carcinogenic mechanisms. Microbiota modulation, either by probiotic administration or by dietary changes, plays an important role in the future therapies of various digestive cancers.
Collapse
Affiliation(s)
- Monica Profir
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Surgery Clinic, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania;
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
24
|
Su L, Yang R, Sheng Y, Ullah S, Zhao Y, Shunjiayi H, Zhao Z, Wang Q. Insights into the oral microbiota in human systemic cancers. Front Microbiol 2024; 15:1369834. [PMID: 38756728 PMCID: PMC11098135 DOI: 10.3389/fmicb.2024.1369834] [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: 01/13/2024] [Accepted: 04/11/2024] [Indexed: 05/18/2024] Open
Abstract
The oral cavity stands as one of the pivotal interfaces facilitating the intricate interaction between the human body and the external environment. The impact of diverse oral microorganisms on the emergence and progression of various systemic cancers, typified by oral cancer, has garnered increasing attention. The potential pathogenicity of oral bacteria, notably the anaerobic Porphyromonas gingivalis and Fusobacterium nucleatum, has been extensively studied and exhibits obvious correlation with different carcinoma types. Furthermore, oral fungi and viruses are closely linked to oropharyngeal carcinoma. Multiple potential mechanisms of oral microbiota-induced carcinogenesis have been investigated, including heightened inflammatory responses, suppression of the host immune system, influence on the tumor microenvironment, anti-apoptotic activity, and promotion of malignant transformation. The disturbance of microbial equilibrium and the migration of oral microbiota play a pivotal role in facilitating oncogenic functions. This review aims to comprehensively outline the pathogenic mechanisms by which oral microbiota participate in carcinogenesis. Additionally, this review delves into their potential applications in cancer prevention, screening, and treatment. It proves to be a valuable resource for researchers investigating the intricate connection between oral microbiota and systemic cancers.
Collapse
Affiliation(s)
- Lan Su
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Rui Yang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Yanan Sheng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Saif Ullah
- Department of Microbiology School of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Yuheng Zhao
- College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Hu Shunjiayi
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuo Zhao
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Qingjing Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Dahab M, Idris H, Zhang P, Aladhadh M, Alatawi EA, Ming LC, Goh KW, Ser HL. Influence of Maqian essential oil on gut microbiota and immunoresponses in type 1 diabetes: In silico study. Heliyon 2024; 10:e29490. [PMID: 38655301 PMCID: PMC11035065 DOI: 10.1016/j.heliyon.2024.e29490] [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: 01/01/2024] [Revised: 03/10/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024] Open
Abstract
Diversity and homeostasis of gut bacterial composition is highly associated with the pathogenesis of insulin dysfunction and type 1 diabetes melittus (T1D), hence emerged in parallel with the activation of autoimmunity. We aimed to study the bioactive potential of essential oil from Zanthoxylum myriacanthum var. pubescens Huang (Maqian) through computational approaches. Twelve chemical constituents derived from Maqian essential oil were docked with selected proteins (i.e., 3pig, 1kho, 7dmq, 4m4d, 2z65, 4glp, and 3fxi) in which are involved in gut microbiota modulation in T1D. Subsequently, the prediction of bioavailability properties of the small molecules were evaluated. Among all chemical constituents, the post-docking interaction analysis demonstrated that α-phellandrene exhibits the strongest binding affinity and induces gut microbiota modulation with β-fructofuranosidase from Bifidobacterium longum. The current result revealed the potential of 3-Carene and α-Pinene in inducing specific changes in gut microbiota downregulating Clostridium perfringens and quenching Leptotrichia shahii respectively. β-Pinene possess exceptionally strong binding affinity that effectively disrupt the interaction between lipopolysaccharide and its cognate receptors, while α-Phellandrene was exhibited the uppermost binding affinity with TLR4/MD2 and could likely target TLR4 stimulating lipopolysaccharide. Our results are the first to report on the gut microbiota modulation effects of α-Phellandrene and β-Phellandrene via actions on LPS binding to CD14 and the TLR4 co-receptor signaling. In conclusion, our findings based on computational approaches, small molecules from Maqian present as promising agents which could regulate inflammatory response and modulate gut microbiota in type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Mahmoud Dahab
- Department of Microbiology, Faculty of Pure and Applied Sciences, International University of Africa, P.O. Box 2469, Khartoum, Sudan
| | - Hajo Idris
- Department of Physics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Saudi Arabia
| | - Ping Zhang
- Center for Integrative Conservation, Yunnan Key Laboratory for the Conservation of Tropical Rainforests and Asian Elephants, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Mengla, 666303, China
| | - Mohammed Aladhadh
- Department of Food Science and Human Nutrition, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia
| | - Eid A Alatawi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, 71491, Saudi Arabia
| | - Long Chiau Ming
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Hooi-Leng Ser
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Sunway City, 47500, Malaysia
| |
Collapse
|
26
|
Loyala JV, Down B, Wong E, Tan B. Treatment of Cachexia in Gastric Cancer: Exploring the Use of Anti-Inflammatory Natural Products and Their Derivatives. Nutrients 2024; 16:1246. [PMID: 38674936 PMCID: PMC11053965 DOI: 10.3390/nu16081246] [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: 03/22/2024] [Revised: 04/12/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
(1) Background: Gastric cancer is a significant cause of cancer-related mortality worldwide. Weight loss and malnutrition associated with cancer are linked with increased mortality rates and reduced quality of life. Cancer cachexia, characterised by the loss of skeletal muscle, is associated with approximately 20% of cancer-related deaths and differs from malnutrition in that it cannot be fully reversed by nutritional support alone. It is now recognised that the primary pathophysiological process underlying cancer cachexia is chronic inflammation leading to increased calorie consumption. Current treatments that focus on nutritional supplementation, psychological counselling, appetite stimulation and reducing inflammation are lacking in efficacy. This review focuses on the evidence supporting the potential roles of natural anti-inflammatory products and their derivatives including fatty acids, probiotics, amino acids, curcumin, fucoidan, epigallocatechin-3-gallate, ginger, resveratrol and Boswellia serrata in the management of gastric cancer cachexia. (2) Results: While natural anti-inflammatory products show promise in a number of in vitro and in vivo studies, there are only a small number of human studies available. Where present, the evidence base is heterogeneous, with varying study methodologies and outcomes. (3) Conclusions: Natural anti-inflammatory products represent a potential adjunctive therapy for gastric cancer cachexia. Further research, particularly well-designed clinical trials, is needed to elucidate their optimal role, dosing and safety profiles in the management of gastric cancer cachexia.
Collapse
Affiliation(s)
- Jerocin Vishani Loyala
- Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, UK
| | - Billy Down
- High Wycombe Hospital, Buckinghamshire Healthcare NHS Trust, High Wycombe HP11 2TT, UK;
| | - Enoch Wong
- Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2GW, UK
| | - Benjamin Tan
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
27
|
Fan S, Zhou L, Zhang W, Wang D, Tang D. Role of imbalanced gut microbiota in promoting CRC metastasis: from theory to clinical application. Cell Commun Signal 2024; 22:232. [PMID: 38637851 PMCID: PMC11025274 DOI: 10.1186/s12964-024-01615-9] [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: 02/12/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024] Open
Abstract
Metastasis poses a major challenge in colorectal cancer (CRC) treatment and remains a primary cause of mortality among patients with CRC. Recent investigations have elucidated the involvement of disrupted gut microbiota homeostasis in various facets of CRC metastasis, exerting a pivotal influence in shaping the metastatic microenvironment, triggering epithelial-mesenchymal transition (EMT), and so on. Moreover, therapeutic interventions targeting the gut microbiota demonstrate promise in enhancing the efficacy of conventional treatments for metastatic CRC (mCRC), presenting novel avenues for mCRC clinical management. Grounded in the "seed and soil" hypothesis, this review consolidates insights into the mechanisms by which imbalanced gut microbiota promotes mCRC and highlights recent strides in leveraging gut microbiota modulation for the clinical prevention and treatment of mCRC. Emphasis is placed on the considerable potential of manipulating gut microbiota within clinical settings for managing mCRC.
Collapse
Affiliation(s)
- Shiying Fan
- Clinical Medical College, Yangzhou University, 225000, Yangzhou, P. R. China
| | - Lujia Zhou
- Clinical Medical College, Yangzhou University, 225000, Yangzhou, P. R. China
| | - Wenjie Zhang
- School of Medicine, Chongqing University, 400030, Chongqing, P. R. China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, 225000, Yangzhou, P. R. China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, 225000, Yangzhou, P. R. China.
| |
Collapse
|
28
|
Wu H, Ma W, Wang Y, Wang Y, Sun X, Zheng Q. Gut microbiome-metabolites axis: A friend or foe to colorectal cancer progression. Biomed Pharmacother 2024; 173:116410. [PMID: 38460373 DOI: 10.1016/j.biopha.2024.116410] [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: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
An expanding corpus of research robustly substantiates the complex interrelation between gut microbiota and the onset, progression, and metastasis of colorectal cancer. Investigations in both animal models and human subjects have consistently underscored the role of gut bacteria in a variety of metabolic activities, driven by dietary intake. These activities include amino acid metabolism, carbohydrate fermentation, and the generation and regulation of bile acids. These metabolic derivatives, in turn, have been identified as significant contributors to the progression of colorectal cancer. This thorough review meticulously explores the dynamic interaction between gut bacteria and metabolites derived from the breakdown of amino acids, fatty acid metabolism, and bile acid synthesis. Notably, bile acids have been recognized for their potential carcinogenic properties, which may expedite tumor development. Extensive research has revealed a reciprocal influence of gut microbiota on the intricate spectrum of colorectal cancer pathologies. Furthermore, strategies to modulate gut microbiota, such as dietary modifications or probiotic supplementation, may offer promising avenues for both the prevention and adjunctive treatment of colorectal cancer. Nevertheless, additional research is imperative to corroborate these findings and enhance our comprehension of the underlying mechanisms in colorectal cancer development.
Collapse
Affiliation(s)
- Hao Wu
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Wenmeng Ma
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yiyao Wang
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yuanyuan Wang
- Department of anesthesiology, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province, PR China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
29
|
Han H, Zhang Y, Tang H, Zhou T, Khan A. A Review of the Use of Native and Engineered Probiotics for Colorectal Cancer Therapy. Int J Mol Sci 2024; 25:3896. [PMID: 38612706 PMCID: PMC11011422 DOI: 10.3390/ijms25073896] [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: 01/20/2024] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Colorectal cancer (CRC) is a serious global health concern, and researchers have been investigating different strategies to prevent, treat, or support conventional therapies for CRC. This review article comprehensively covers CRC therapy involving wild-type bacteria, including probiotics and oncolytic bacteria as well as genetically modified bacteria. Given the close relationship between CRC and the gut microbiota, it is crucial to compile and present a comprehensive overview of bacterial therapies used in the context of colorectal cancer. It is evident that the use of native and engineered probiotics for colorectal cancer therapy necessitates research focused on enhancing the therapeutic properties of probiotic strains.. Genetically engineered probiotics might be designed to produce particular molecules or to target cancer cells more effectively and cure CRC patients.
Collapse
Affiliation(s)
- Huawen Han
- State Key Laboratory of Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Yifan Zhang
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK;
| | - Haibo Tang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou 730000, China; (H.T.); (T.Z.)
| | - Tuoyu Zhou
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou 730000, China; (H.T.); (T.Z.)
| | - Aman Khan
- College of Life Sciences, Northeast Forestry University, Harbin 150040, China
| |
Collapse
|
30
|
Mohammed C, Fuego JP, Garcia KV, Jamil H, Rajesh RY, Escobar AS, Hassan MJ, Rai M. A Mini Literature Review of Probiotics: Transforming Gastrointestinal Health Through Evidence-Based Insights. Cureus 2024; 16:e57055. [PMID: 38681263 PMCID: PMC11051678 DOI: 10.7759/cureus.57055] [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] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
As our understanding of the intricate interaction between gut bacteria and human health continues to expand, so too has interest in the ability of probiotics to manage gut microbiota and confer multiple health benefits to the host. The mini literature review focuses on the expanding potential of the use of probiotics in GI health, with a focus on probiotics' potential therapeutic advantages in a variety of gastrointestinal (GI) illnesses. Probiotics play a significant role in managing diarrhea and symptoms of irritable bowel syndrome with diarrhea (IBS-D) by modulating gut microbial communities. Specific probiotic strains have been found to reduce the abundance of harmful bacteria, regulate inflammatory markers like interleukin 6, and improve GI symptoms such as abdominal discomfort and stool consistency. Additionally, probiotic blends have shown potential for preventing GI infections and alleviating GI pain in IBS-D patients. Studies have demonstrated that certain multi-strain probiotics, including Bifidobacterium and Lactobacillus species, can significantly increase the frequency of bowel movements and reduce the proportion of individuals experiencing constipation. It has also been found that probiotic supplementation may reduce the incidence of postoperative complications and mortality, particularly in patients undergoing colorectal adenocarcinoma surgery. Additionally, probiotics have been associated with decreased levels of pro-inflammatory cytokines and improved clinical outcomes in patients with colorectal cancer. Furthermore, probiotics have been associated with enhanced digestive tolerance, reduced GI inflammation, and prolonged clinical remission in certain UC patients. Studies have also shown that probiotics, administered either directly to infants or pregnant women during the perinatal stage, can alleviate symptoms such as inconsolable crying and irritation associated with infant colic, improve bowel movement frequency in cases of functional constipation, and enhance overall conditions in premature infants, including reducing regurgitation and improving feeding tolerance. The review addresses both encouraging results and challenges with probiotic therapy, while also arguing for more studies to elucidate underlying mechanisms and enhance therapeutic techniques. As we traverse the complex field of probiotic therapy in the treatment of GI illnesses, researchers, physicians, and other healthcare professionals can benefit from the informative information provided by this study.
Collapse
Affiliation(s)
- Cara Mohammed
- Orthopedics, East Regional Health Authority, Port of Spain, TTO
| | - Jhon P Fuego
- Internal Medicine, West Visayas State University College of Medicine, Iloilo City, PHL
| | - Karina V Garcia
- Internal Medicine, National Autonomous University of Mexico, Mexico City, MEX
| | - Hira Jamil
- Medicine, University Medical and Dental College Faisalabad, Faisalabad, PAK
| | - Rahul Y Rajesh
- Internal Medicine, Tbilisi State Medical University, Tbilisi, GEO
| | | | | | - Manju Rai
- Immunology, Shri Venkateshwara University, Gajraula, IND
| |
Collapse
|
31
|
Pandey M, Bhattacharyya J. Gut microbiota and epigenetics in colorectal cancer: implications for carcinogenesis and therapeutic intervention. Epigenomics 2024; 16:403-418. [PMID: 38410915 DOI: 10.2217/epi-2023-0382] [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] [Indexed: 02/28/2024] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. The occurrence of CRC is associated with various genetic and epigenetic mutations in intestinal epithelial cells that transform them into adenocarcinomas. There is increasing evidence indicating the gut microbiota plays a crucial role in the regulation of host physiological processes. Alterations in gut microbiota composition are responsible for initiating carcinogenesis through diverse epigenetic modifications, including histone modifications, ncRNAs and DNA methylation. This work was designed to comprehensively review recent findings to provide insight into the associations between the gut microbiota and CRC at an epigenetic level. These scientific insights can be used in the future to develop effective strategies for early detection and treatment of CRC.
Collapse
Affiliation(s)
- Monu Pandey
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, 110016, India
- Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, 110608, India
| | - Jayanta Bhattacharyya
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, 110016, India
- Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, 110608, India
| |
Collapse
|
32
|
Bahitham W, Alghamdi S, Omer I, Alsudais A, Hakeem I, Alghamdi A, Abualnaja R, Sanai FM, Rosado AS, Sergi CM. Double Trouble: How Microbiome Dysbiosis and Mitochondrial Dysfunction Drive Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. Biomedicines 2024; 12:550. [PMID: 38540163 PMCID: PMC10967987 DOI: 10.3390/biomedicines12030550] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/12/2024] [Accepted: 02/18/2024] [Indexed: 11/22/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are closely related liver conditions that have become more prevalent globally. This review examines the intricate interplay between microbiome dysbiosis and mitochondrial dysfunction in the development of NAFLD and NASH. The combination of these two factors creates a synergistic situation referred to as "double trouble", which promotes the accumulation of lipids in the liver and the subsequent progression from simple steatosis (NAFLD) to inflammation (NASH). Microbiome dysbiosis, characterized by changes in the composition of gut microbes and increased intestinal permeability, contributes to the movement of bacterial products into the liver. It triggers metabolic disturbances and has anti-inflammatory effects. Understanding the complex relationship between microbiome dysbiosis and mitochondrial dysfunction in the development of NAFLD and NASH is crucial for advancing innovative therapeutic approaches that target these underlying mechanisms.
Collapse
Affiliation(s)
- Wesam Bahitham
- King Abdullah International Medical Research Center-WR, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard for Health Affairs, Riyadh 11426, Saudi Arabia
- Bioscience, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Siraj Alghamdi
- King Abdullah International Medical Research Center-WR, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard for Health Affairs, Riyadh 11426, Saudi Arabia
| | - Ibrahim Omer
- King Abdullah International Medical Research Center-WR, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard for Health Affairs, Riyadh 11426, Saudi Arabia
| | - Ali Alsudais
- King Abdullah International Medical Research Center-WR, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard for Health Affairs, Riyadh 11426, Saudi Arabia
| | - Ilana Hakeem
- King Abdullah International Medical Research Center-WR, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard for Health Affairs, Riyadh 11426, Saudi Arabia
| | - Arwa Alghamdi
- King Abdullah International Medical Research Center-WR, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard for Health Affairs, Riyadh 11426, Saudi Arabia
| | - Reema Abualnaja
- King Abdullah International Medical Research Center-WR, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard for Health Affairs, Riyadh 11426, Saudi Arabia
| | - Faisal M Sanai
- Gastroenterology Unit, Department of Medicine, King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Alexandre S Rosado
- Bioscience, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Consolato M Sergi
- Anatomic Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2B7, Canada
| |
Collapse
|
33
|
Roy R, Singh SK. The Microbiome Modulates the Immune System to Influence Cancer Therapy. Cancers (Basel) 2024; 16:779. [PMID: 38398170 PMCID: PMC10886470 DOI: 10.3390/cancers16040779] [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: 01/22/2024] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The gut microbiota composition can affect the tumor microenvironment and its interaction with the immune system, thereby having implications for treatment predictions. This article reviews the studies available to better understand how the gut microbiome helps the immune system fight cancer. To describe this fact, different mechanisms and approaches utilizing probiotics to improve advancements in cancer treatment will be discussed. Moreover, not only calorie intake but also the variety and quality of diet can influence cancer patients' immunotherapy treatment because dietary patterns can impair immunological activities either by stimulating or suppressing innate and adaptive immunity. Therefore, it is interesting and critical to understand gut microbiome composition as a biomarker to predict cancer immunotherapy outcomes and responses. Here, more emphasis will be given to the recent development in immunotherapies utilizing microbiota to improve cancer therapies, which is beneficial for cancer patients.
Collapse
Affiliation(s)
- Ruchi Roy
- UICentre for Drug Discovery, The University of Illinois, Chicago, IL 60612, USA
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
34
|
Spalinger MR, Scharl M. Microbiota Manipulation as an Emerging Concept in Cancer Therapy. Visc Med 2024; 40:2-11. [PMID: 38312366 PMCID: PMC10836949 DOI: 10.1159/000534810] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/24/2023] [Indexed: 02/06/2024] Open
Abstract
Background The human body is colonized by billions of bacteria that provide nutrients to the host, train our immune system, and importantly affect our heath. It has long been suggested that microbes might play a role in tumor pathogenesis; however, compelling evidence was only provided in the past decades when novel detection methods that do not depend on culturing techniques had been developed. Summary The microbiome impacts tumor development and anti-tumor therapies on various levels. Bacteria can promote or suppress tumor growth via direct interactions with cancer cells, production of metabolites that promote or inhibit tumor growth, and via stimulation or suppression of the local and systemic immune response. Cancer patients harbor a distinct microbiome when compared to healthy controls, which could potentially be employed to detect, identify, and treat cancer. Manipulation of the microbiome either via supplementation of single strains, bacterial consortia, fecal microbiota transfer or the use of pre- and probiotics has been suggested as therapeutic approach to directly target tumor growth or to enhance the efficacy of current state-of-the-art treatment options. Key Messages (1) Bacteria have a tremendous impact on anti-cancer immune responses. (2) Cancer patients harbor a distinct microbiome when compared to healthy controls. (3) The microbiome seems to be cancer-type specific. (4) Exploitation of bacteria to promote anti-tumor therapy is a novel, very promising venue in cancer treatment.
Collapse
Affiliation(s)
| | - Michael Scharl
- Department for Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Nachum Z, Perlitz Y, Shavit LY, Magril G, Vitner D, Zipori Y, Weiner E, Alon AS, Ganor-Paz Y, Nezer M, Harel N, Soltsman S, Yefet E. The effect of oral probiotics on glycemic control of women with gestational diabetes mellitus-a multicenter, randomized, double-blind, placebo-controlled trial. Am J Obstet Gynecol MFM 2024; 6:101224. [PMID: 37956906 DOI: 10.1016/j.ajogmf.2023.101224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/29/2023] [Accepted: 11/08/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Gestational diabetes mellitus should be treated adequately to avoid maternal hyperglycemia-related complications. Previously, probiotic supplements were suggested to improve fasting blood glucose in women with gestational diabetes mellitus. However, a major limitation of previous studies was that preprandial and especially postprandial glucose values, which are important predictors of pregnancy outcomes, were not studied. OBJECTIVE This study aimed to examine the effect of a mixture of probiotic strains on maternal glycemic parameters, particularly preprandial and postprandial glucose values and pregnancy outcomes among women with gestational diabetes mellitus. STUDY DESIGN A multicenter prospective randomized, double-blind, placebo-controlled trial was conducted. Women newly diagnosed with gestational diabetes mellitus were randomly allocated into a research group, receiving 2 capsules of oral probiotic formula containing Bifidobacterium bifidum, B lactis, Lactobacillus acidophilus, L paracasei, L rhamnosus, and Streptococcus thermophilus (>6 × 109/capsule), and a control group, receiving a placebo (2 capsules/day) until delivery. Glycemic control was evaluated by daily glucose charts. After 2 weeks, pharmacotherapy was started in case of poor glycemic control. The primary outcomes were the rate of women requiring medications for glycemic control and mean daily glucose charts after 2 weeks of treatment with the study products. RESULTS Forty-one and 44 women were analyzed in the treatment and placebo cohorts, respectively. Mean daily glucose during the first 2 weeks in the probiotics and placebo groups was 99.7±7.9 and 98.0±9.3 mg/dL, respectively (P=.35). The rate of women needing pharmacotherapy because of poor glycemic control after 2 weeks of treatment in the probiotics and placebo groups was 24 (59%) and 18 (41%), respectively (P=.10). Mean preprandial and postprandial glucose levels throughout the study period were similar between the groups (P>.05). There were no differences in maternal and neonatal outcomes, including birthweight and adverse effect profile between the groups. CONCLUSION The oral probiotic product tested in this study did not affect glycemic control of women with gestational diabetes mellitus.
Collapse
Affiliation(s)
- Zohar Nachum
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, Israel (Dr Nachum); Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel (Drs Nachum, Vitner, and Zipori)
| | - Yuri Perlitz
- Department of Obstetrics and Gynecology, Tzafon Medical Center, Poriya, Israel (Drs Perlitz, Harel, Soltsman, and Yefet); Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel (Drs Perlitz and Yefet)
| | - Lilach Yacov Shavit
- Diabetes in Pregnancy Clinic, Tzafon Medical Center, Poriya, Israel (Ms Shavit)
| | - Galit Magril
- Nutrition Division, Tzafon Medical Center Poriya, Israel (Ms Magril)
| | - Dana Vitner
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel (Drs Nachum, Vitner, and Zipori); Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel (Drs Vitner and Zipori)
| | - Yaniv Zipori
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel (Drs Nachum, Vitner, and Zipori); Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel (Drs Vitner and Zipori)
| | - Eran Weiner
- Department of Obstetrics and Gynecology, Wolfson Medical Center, Holon, Israel (Drs Weiner, Alon, and Ganor-Paz); Sackler Faculty of Medicine, Tel Aviv university, Tel Aviv, Israel (Drs Weiner, Alon, and Ganor-Paz)
| | - Ayala Shevach Alon
- Department of Obstetrics and Gynecology, Wolfson Medical Center, Holon, Israel (Drs Weiner, Alon, and Ganor-Paz); Sackler Faculty of Medicine, Tel Aviv university, Tel Aviv, Israel (Drs Weiner, Alon, and Ganor-Paz)
| | - Yael Ganor-Paz
- Department of Obstetrics and Gynecology, Wolfson Medical Center, Holon, Israel (Drs Weiner, Alon, and Ganor-Paz); Sackler Faculty of Medicine, Tel Aviv university, Tel Aviv, Israel (Drs Weiner, Alon, and Ganor-Paz)
| | - Meirav Nezer
- Department of Obstetrics and Gynecology, Samson Assuta Ashdod University Hospital, Ashdod, Israel (Dr Nezer)
| | - Noa Harel
- Department of Obstetrics and Gynecology, Tzafon Medical Center, Poriya, Israel (Drs Perlitz, Harel, Soltsman, and Yefet)
| | - Sofia Soltsman
- Department of Obstetrics and Gynecology, Tzafon Medical Center, Poriya, Israel (Drs Perlitz, Harel, Soltsman, and Yefet)
| | - Enav Yefet
- Department of Obstetrics and Gynecology, Tzafon Medical Center, Poriya, Israel (Drs Perlitz, Harel, Soltsman, and Yefet); Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel (Drs Perlitz and Yefet); Women's Health Center, Clalit Health Services, Afula, Israel (Dr Yefet).
| |
Collapse
|
36
|
Nenu I, Baldea I, Coadă CA, Crăciun RC, Moldovan R, Tudor D, Petrushev B, Toma VA, Ştefanescu H, Procopeţ B, Spârchez Z, Vodnar D, Lenghel M, Clichici S, Filip GA. Lactobacillus rhamnosus probiotic treatment modulates gut and liver inflammatory pathways in a hepatocellular carcinoma murine model. A preliminary study. Food Chem Toxicol 2024; 183:114314. [PMID: 38052407 DOI: 10.1016/j.fct.2023.114314] [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: 07/23/2023] [Revised: 09/18/2023] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) is a growing global concern with an increasing incidence rate. The intestinal microbiota has been identified as a potential culprit in modulating the effects of antitumoral drugs. We aimed to assess the impact of adding Lactobacillus rhamnosus probiotic to regorafenib in mice with HCC. METHODS Cirrhosis and HCCs were induced in 56 male Swiss mice via diethylnitrosamine injection and carbon tetrachloride administration. Mice were divided into four groups: treated with vehicle (VC), regorafenib (Rego), L. rhamnosus probiotic, and a combination of regorafenib and probiotic (Rego-Pro). After 3 weeks of treatment, liver and intestinal fragments were collected for analysis. RESULTS Regorafenib elevated gut permeability, an effect mitigated by probiotic intervention, which exhibited a notable correlation with reduced inflammation (p < 0.01). iNOS levels were also reduced by adding the probiotic with respect to the mice treated with regorafenib only (p < 0.001). Notably, regorafenib substantially increased IL-6, TNF-a and TLR4 in intestinal fragments (p < 0.01). The administration of the probiotic effectively restored IL-6 to its initial levels (p < 0.001). CONCLUSION Reducing systemic and intestinal inflammation by administering L. rhamnosus probiotic may alleviate tumoral resistance and systemic adverse effects.
Collapse
Affiliation(s)
- Iuliana Nenu
- Department of Functional Sciences, Physiology Discipline, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania; Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania.
| | - Ioana Baldea
- Department of Functional Sciences, Physiology Discipline, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | | | - Rareş Călin Crăciun
- Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania.
| | - Remus Moldovan
- Department of Functional Sciences, Physiology Discipline, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Diana Tudor
- Department of Functional Sciences, Physiology Discipline, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Bobe Petrushev
- Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania.
| | - Vlad Alexandru Toma
- Department of Molecular Biology and Biotechnologies, Faculty of Biology and Geology, Babeș-Bolyai University, Cluj-Napoca, Romania; Department of Experimental Biology and Biochemistry, Institute of Biological Research, Branch of NIRDBS, Cluj-Napoca, Romania; Department of Molecular and Biomolecular Physics, NIRD for Isotopic and Molecular Technologies, Cluj-Napoca, Romania.
| | - Horia Ştefanescu
- Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania.
| | - Bogdan Procopeţ
- Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania.
| | - Zeno Spârchez
- Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania.
| | - Dan Vodnar
- Department of Food Science University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania.
| | - Manuela Lenghel
- Radiology Department, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Simona Clichici
- Department of Functional Sciences, Physiology Discipline, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Gabriela Adriana Filip
- Department of Functional Sciences, Physiology Discipline, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
37
|
Esposito A, Bossi P. How can we prevent diarrhea induced by systemic treatments in oncological patients? An evaluation of phase II trials. Expert Opin Emerg Drugs 2023; 28:213-216. [PMID: 37750491 DOI: 10.1080/14728214.2023.2264182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/25/2023] [Indexed: 09/27/2023]
Affiliation(s)
- Andrea Esposito
- Department of Medical and Surgical Specialities, Radiological Sciences and Public Health, ASST-Spedali Civili Brescia, University of Brescia, Oncology Unit, Piazzale Spedali Civili, Brescia, Italy
| | - Paolo Bossi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
38
|
Thet D, Areepium N, Siritientong T. Effects of Probiotics on Chemotherapy-induced Diarrhea. Nutr Cancer 2023; 75:1811-1821. [PMID: 37908158 DOI: 10.1080/01635581.2023.2267779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 11/02/2023]
Abstract
Chemotherapy-induced diarrhea (CID) is a common adverse event in cancer patients, which, unless treated, may lead to drug discontinuation and treatment failure. Some probiotics such as Lactobacillus, Bifidobacterium, and Saccharomyces species have been gaining clinical attention in alleviating chemotherapy-induced adverse events including diarrhea. This comprehensive review provides an overview and discusses preventive approaches of probiotics with respect to CID in several types of cancers. The potential mechanisms of probiotics may comprise regulation of intestinal microbiota, modulation of immune functions, or reduction of proinflammatory cytokines. The efficacy and safety precautions of probiotics in immunocompromised cancer patients are discussed. The non-pharmacological strategy using probiotics could reduce the use of anti-diarrheal or antibiotic agents. Some individuals experienced shorter length of hospital stay, better gastrointestinal function, and reduced incidence of chemotherapy dose reduction after probiotic administration. Nonetheless, some studies failed to report the benefits of probiotics in certain patients. This review also highlights preventive protocols and therapeutic implications by considering the potential influencing factors, particularly types of probiotic strains, dosages of probiotics, duration of their administration, patients' tolerability, and variations in probiotic effects over the cancer stages.
Collapse
Affiliation(s)
- Daylia Thet
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Nutthada Areepium
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tippawan Siritientong
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
39
|
Ciernikova S, Sevcikova A, Drgona L, Mego M. Modulating the gut microbiota by probiotics, prebiotics, postbiotics, and fecal microbiota transplantation: An emerging trend in cancer patient care. Biochim Biophys Acta Rev Cancer 2023; 1878:188990. [PMID: 37742728 DOI: 10.1016/j.bbcan.2023.188990] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/26/2023]
Abstract
Treatment resistance, together with acute and late adverse effects, represents critical issues in the management of cancer patients. Promising results from preclinical and clinical research underline the emerging trend of a microbiome-based approach in oncology. Favorable bacterial species and higher gut diversity are associated with increased treatment efficacy, mainly in chemo- and immunotherapy. On the other hand, alterations in the composition and activity of gut microbial communities are linked to intestinal dysbiosis and contribute to high treatment-induced toxicity. In this Review, we provide an overview of studies concerning gut microbiota modulation in patients with solid and hematologic malignancies with a focus on probiotics, prebiotics, postbiotics, and fecal microbiota transplantation. Targeting the gut microbiome might bring clinical benefits and improve patient outcomes. However, a deeper understanding of mechanisms and large clinical trials concerning microbiome and immunological profiling is warranted to identify safe and effective ways to incorporate microbiota-based interventions in routine clinical practice.
Collapse
Affiliation(s)
- Sona Ciernikova
- Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Aneta Sevcikova
- Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lubos Drgona
- Department of Oncohematology, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Michal Mego
- 2nd Department of Oncology, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
40
|
Guo P, Wang S, Yue H, Zhang X, Ma G, Li X, Wei W. Advancement of Engineered Bacteria for Orally Delivered Therapeutics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302702. [PMID: 37537714 DOI: 10.1002/smll.202302702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/06/2023] [Indexed: 08/05/2023]
Abstract
The use of bacteria and their biotic components as therapeutics has shown great potential in the treatment of diseases. Orally delivered bacteria improve patient compliance compared with injection-administered bacteria and are considered the preferred mode. However, due to the harsh gastrointestinal environment, the viability and therapeutic efficacy of orally delivered bacteria are significantly reduced in vivo. In recent years, with the rapid development of synthetic biology and nanotechnology, bacteria and biotic components have been engineered to achieve directed genetic reprogramming for construction and precise spatiotemporal control in the gastrointestinal tract, which can improve viability and therapeutic efficiency. Herein, a state-of-the-art review on the current progress of engineered bacterial systems for oral delivery is provided. The different types of bacterial and biotic components for oral administration are first summarized. The engineering strategies of these bacteria and biotic components and their treatment of diseases are next systematically summarized. Finally, the current challenges and prospects of these bacterial therapeutics are highlighted that will contribute to the development of next-generation orally delivered bacteriotherapy.
Collapse
Affiliation(s)
- Peilin Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Hua Yue
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiao Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xin Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
41
|
Sharma D, Gajjar D, Seshadri S. Understanding the role of gut microfloral bifidobacterium in cancer and its potential therapeutic applications. MICROBIOME RESEARCH REPORTS 2023; 3:3. [PMID: 38455077 PMCID: PMC10917622 DOI: 10.20517/mrr.2023.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/28/2023] [Accepted: 10/30/2023] [Indexed: 03/09/2024]
Abstract
Gut microbiota research has gained a tremendous amount of attention from the scientific community because of its contribution to gut homeostasis, human health, and various pathophysiological conditions. The early colonizer of the human gut, i.e., bifidobacteria, has emerged as an efficient probiotic in various diseased conditions, including cancer. This review explores the pros and cons of Bifidobacterium in various malignancies and various therapeutic strategies. We have illustrated the controversial role of bifidobacteria participating in various malignancies as well as described the current knowledge regarding its use in anticancer therapies. Ultimately, this article also addresses the need for further extensive research in elucidating the mechanism of how bifidobacteria is involved and is indirectly affecting the tumor microenvironment. Exhaustive and large-scale research is also required to solve the controversial questions regarding the involvement of bifidobacteria in cancer research.
Collapse
Affiliation(s)
| | | | - Sriram Seshadri
- Institute of Science, Nirma University, 382481 Ahmedabad, Gujarat, India
| |
Collapse
|
42
|
Martinelli S, Lamminpää I, Dübüş EN, Sarıkaya D, Niccolai E. Synergistic Strategies for Gastrointestinal Cancer Care: Unveiling the Benefits of Immunonutrition and Microbiota Modulation. Nutrients 2023; 15:4408. [PMID: 37892482 PMCID: PMC10610426 DOI: 10.3390/nu15204408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Gastrointestinal (GI) cancers are a group of highly prevalent malignant tumors affecting the gastrointestinal tract. Globally, one in four cancer cases and one in three cancer deaths are estimated to be GI cancers. They can alter digestive and absorption functions, leading to severe malnutrition which may worsen the prognosis of the patients. Therefore, nutritional intervention and monitoring play a fundamental role in managing metabolic alterations and cancer symptoms, as well as minimizing side effects and increasing the effectiveness of chemotherapy. In this scenario, the use of immunonutrients that are able to modulate the immune system and the modification/regulation of the gut microbiota composition have gained attention as a possible strategy to improve the conditions of these patients. The complex interaction between nutrients and microbiota might contribute to maintaining the homeostasis of each individual's immune system; therefore, concurrent use of specific nutrients in combination with traditional cancer treatments may synergistically improve the overall care of GI cancer patients. This work aims to review and discuss the role of immunonutrition and microbiota modulation in improving nutritional status, postoperative recovery, and response to therapies in patients with GI cancer.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| | - Ingrid Lamminpää
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| | - Eda Nur Dübüş
- Department of Nutrition and Dietetics, Gazi University, 06560 Ankara, Turkey; (E.N.D.); (D.S.)
| | - Dilara Sarıkaya
- Department of Nutrition and Dietetics, Gazi University, 06560 Ankara, Turkey; (E.N.D.); (D.S.)
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| |
Collapse
|
43
|
Wang Z, Zhou Y, Luo A, Heng X, Liu J, Wang H, Chu W. Lactobacillus salivarius CPU-01 Ameliorates Temozolomide-Induced Intestinal Mucositis by Modulating Gut Microbiota, Maintaining Intestinal Barrier, and Blocking Pro-inflammatory Cytokines. Probiotics Antimicrob Proteins 2023; 15:1079-1091. [PMID: 35639268 DOI: 10.1007/s12602-022-09955-z] [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] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Chemotherapy-induced intestinal mucositis is one of the major toxic side effects in the treatment of cancer patients. The purpose of this study is to screen lactic acid bacteria which could alleviate intestinal inflammation and damage induced by chemotherapeutic agents and explore the possible underlying mechanisms. Lactobacillus salivarius CPU-01 was selected from traditional Chinese fermented foods due to its protective effects on the toxicity of temozolomide in Caenorhabditis elegans. Eighteen ICR mice were randomly divided into 3 groups including control group, temozolomide-induced intestinal mucositis group, and temozolomide + L. salivarius CPU-01 group, and were used to investigate the effect of L. salivarius CPU-01 on chemotherapy-induced intestinal mucositis. It has been demonstrated that the administration of L. salivarius CPU-01 can prevent colon shortening and alleviate colon tissue damage caused by temozolomide-induced intestinal mucositis in mice. L. salivarius CPU-01 relieved the intestinal microbiota disorders caused by temozolomide and contributed to the growth of beneficial bacteria, such as Lactobacillus, Clostridia UCG - 014_norank, and Akkermansia. In vivo experiments also indicated that L. salivarius CPU-01 can suppress the level of temozolomide-induced pro-inflammatory cytokines in serum and mRNA expression in the small intestine tissues. It was also found that L. salivarius CPU-01 significantly increased the expressions of intestinal tight junction (TJ) proteins, ZO-1, and Occludin proteins in mice treated with temozolomide. These findings suggest that L. salivarius CPU-01 can ameliorate temozolomide-induced intestinal mucositis by modulating gut microbiota, blocking pro-inflammatory cytokines, and repairing the intestinal barrier. These findings suggest probiotics may serve as a potential alternative therapeutic strategy for the prevention of chemotherapy-induced intestinal mucositis in the future.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuhong Zhou
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Aoxiang Luo
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Xing Heng
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Jinqiu Liu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Huafu Wang
- Lishui People's Hospital, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Weihua Chu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
44
|
Cornista AM, Giolito MV, Baker K, Hazime H, Dufait I, Datta J, Khumukcham SS, De Ridder M, Roper J, Abreu MT, Breckpot K, Van der Jeught K. Colorectal Cancer Immunotherapy: State of the Art and Future Directions. GASTRO HEP ADVANCES 2023; 2:1103-1119. [PMID: 38098742 PMCID: PMC10721132 DOI: 10.1016/j.gastha.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Cancer immunotherapy has become an indispensable mode of treatment for a multitude of solid tumor cancers. Colorectal cancer (CRC) has been one of the many cancer types to benefit from immunotherapy, especially in advanced disease where standard treatment fails to prevent recurrence or results in poor survival. The efficacy of immunotherapy in CRC has not been without challenge, as early clinical trials observed dismal responses in unselected CRC patients treated with checkpoint inhibitors. Many studies and clinical trials have since refined immunotherapies available for CRC, solidifying immunotherapy as a powerful asset for CRC treatment. This review article examines CRC immunotherapies, from their foundation, through emerging avenues for improvement, to future directions.
Collapse
Affiliation(s)
- Alyssa Mauri Cornista
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
| | - Maria Virginia Giolito
- Department of Biomedical Sciences, Vrije Universiteit Brussel, Laboratory for Molecular and Cellular Therapy, Brussels, Belgium
| | - Kristi Baker
- Department of Oncology, University of Alberta, Edmonton, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Hajar Hazime
- Division of Gastroenterology, University of Miami Miller School of Medicine, Miami, Florida
| | - Inès Dufait
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jashodeep Datta
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
- Division of Surgical Oncology, Dewitt Daughtry Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Saratchandra Singh Khumukcham
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Mark De Ridder
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jatin Roper
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Maria T. Abreu
- Division of Gastroenterology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Karine Breckpot
- Department of Biomedical Sciences, Vrije Universiteit Brussel, Laboratory for Molecular and Cellular Therapy, Brussels, Belgium
| | - Kevin Van der Jeught
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
45
|
Zhuang YP, Zhou HL, Chen HB, Zheng MY, Liang YW, Gu YT, Li WT, Qiu WL, Zhou HG. Gut microbiota interactions with antitumor immunity in colorectal cancer: From understanding to application. Biomed Pharmacother 2023; 165:115040. [PMID: 37364479 DOI: 10.1016/j.biopha.2023.115040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Colorectal cancer (CRC) is one of highly prevalent cancer. Immunotherapy with immune checkpoint inhibitors (ICIs) has dramatically changed the landscape of treatment for many advanced cancers, but CRC still exhibits suboptimal response to immunotherapy. The gut microbiota can affect both anti-tumor and pro-tumor immune responses, and further modulate the efficacy of cancer immunotherapy, particularly in the context of therapy with ICIs. Therefore, a deeper understanding of how the gut microbiota modulates immune responses is crucial to improve the outcomes of CRC patients receiving immunotherapy and to overcome resistance in nonresponders. The present review aims to describe the relationship between the gut microbiota, CRC, and antitumor immune responses, with a particular focus on key studies and recent findings on the effect of the gut microbiota on the antitumor immune activity. We also discuss the potential mechanisms by which the gut microbiota influences host antitumor immune responses as well as the prospective role of intestinal flora in CRC treatment. Furthermore, the therapeutic potential and limitations of different modulation strategies for the gut microbiota are also discussed. These insights may facilitate to better comprehend the interplay between the gut microbiota and the antitumor immune responses of CRC patients and provide new research pathways to enhance immunotherapy efficacy and expand the patient population that could be benefited by immunotherapy.
Collapse
Affiliation(s)
- Yu-Pei Zhuang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong-Li Zhou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hai-Bin Chen
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming-Yue Zheng
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu-Wei Liang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu-Tian Gu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen-Ting Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Wen-Li Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Hong-Guang Zhou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
46
|
Bai L, Yan X, Lv J, Qi P, Song X, Zhang L. Intestinal Flora in Chemotherapy Resistance of Biliary Pancreatic Cancer. BIOLOGY 2023; 12:1151. [PMID: 37627035 PMCID: PMC10452461 DOI: 10.3390/biology12081151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Biliary pancreatic malignancy has an occultic onset, a high degree of malignancy, and a poor prognosis. Most clinical patients miss the opportunity for surgical resection of the tumor. Systemic chemotherapy is still one of the important methods for the treatment of biliary pancreatic malignancies. Many chemotherapy regimens are available, but their efficacy is not satisfactory, and the occurrence of chemotherapy resistance is a major reason leading to poor prognosis. With the advancement of studies on intestinal flora, it has been found that intestinal flora is correlated with and plays an important role in chemotherapy resistance. The application of probiotics and other ways to regulate intestinal flora can improve this problem. This paper aims to review and analyze the research progress of intestinal flora in the chemotherapy resistance of biliary pancreatic malignancies to provide new ideas for treatment.
Collapse
Affiliation(s)
- Liuhui Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiangdong Yan
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jin Lv
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Ping Qi
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiaojing Song
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
47
|
Li Q, Chan H, Liu WX, Liu CA, Zhou Y, Huang D, Wang X, Li X, Xie C, Liu WYZ, Wang XS, Ng SK, Gou H, Zhao LY, Fong W, Jiang L, Lin Y, Zhao G, Bai F, Liu X, Chen H, Zhang L, Wong SH, Chan MTV, Wu WKK, Yu J. Carnobacterium maltaromaticum boosts intestinal vitamin D production to suppress colorectal cancer in female mice. Cancer Cell 2023; 41:1450-1465.e8. [PMID: 37478851 DOI: 10.1016/j.ccell.2023.06.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/03/2023] [Accepted: 06/29/2023] [Indexed: 07/23/2023]
Abstract
Carnobacterium maltaromaticum was found to be specifically depleted in female patients with colorectal cancer (CRC). Administration of C. maltaromaticum reduces intestinal tumor formation in two murine CRC models in a female-specific manner. Estrogen increases the attachment and colonization of C. maltaromaticum via increasing the colonic expression of SLC3A2 that binds to DD-CPase of this bacterium. Metabolomic and transcriptomic profiling unveils the increased gut abundance of vitamin D-related metabolites and the mucosal activation of vitamin D receptor (VDR) signaling in C. maltaromaticum-gavaged mice in a gut microbiome- and VDR-dependent manner. In vitro fermentation system confirms the metabolic cross-feeding of C. maltaromaticum with Faecalibacterium prausnitzii to convert C. maltaromaticum-produced 7-dehydrocholesterol into vitamin D for activating the host VDR signaling. Overall, C. maltaromaticum colonizes the gut in an estrogen-dependent manner and acts along with other microbes to augment the intestinal vitamin D production to activate the host VDR for suppressing CRC.
Collapse
Affiliation(s)
- Qing Li
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hung Chan
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wei-Xin Liu
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chang-An Liu
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yunfei Zhou
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dan Huang
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xueliang Wang
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiaoxing Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Chuan Xie
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing Ying-Zhi Liu
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xian-Song Wang
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Siu Kin Ng
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hongyan Gou
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liu-Yang Zhao
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Winnie Fong
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lanping Jiang
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yufeng Lin
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Guijun Zhao
- Department of Endoscopy Center, Inner Mongolia Key Laboratory of Endoscopic Digestive Disease, Inner Mongolia people's Hospital, Hohhot, China
| | - Feihu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarong Chen
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sunny Hei Wong
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Matthew Tak Vai Chan
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - William Ka Kei Wu
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jun Yu
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
48
|
Eze OC, Berebon DP, Emencheta SC, Evurani SA, Okorie CN, Balcão VM, Vila MMDC. Therapeutic Potential of Marine Probiotics: A Survey on the Anticancer and Antibacterial Effects of Pseudoalteromonas spp. Pharmaceuticals (Basel) 2023; 16:1091. [PMID: 37631006 PMCID: PMC10458718 DOI: 10.3390/ph16081091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/17/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Due to the increasing limitations and negative impacts of the current options for preventing and managing diseases, including chemotherapeutic drugs and radiation, alternative therapies are needed, especially ones utilizing and maximizing natural products (NPs). NPs abound with diverse bioactive primary and secondary metabolites and compounds with therapeutic properties. Marine probiotics are beneficial microorganisms that inhabit marine environments and can benefit their hosts by improving health, growth, and disease resistance. Several studies have shown they possess potential bioactive and therapeutic actions against diverse disease conditions, thus opening the way for possible exploitation of their benefits through their application. Pseudoalteromonas spp. are a widely distributed heterotrophic, flagellated, non-spore-forming, rod-shaped, and gram-negative marine probiotic bacteria species with reported therapeutic capabilities, including anti-cancer and -bacterial effects. This review discusses the basic concepts of marine probiotics and their therapeutic effects. Additionally, a survey of the anticancer and antibacterial effects of Pseudoalteromonas spp. is presented. Finally, marine probiotic production, advances, prospects, and future perspectives is presented.
Collapse
Affiliation(s)
- Osita C. Eze
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Nigeria; (O.C.E.); (S.A.E.); (C.N.O.)
| | - Dinebari P. Berebon
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Nigeria; (O.C.E.); (S.A.E.); (C.N.O.)
| | - Stephen C. Emencheta
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Nigeria; (O.C.E.); (S.A.E.); (C.N.O.)
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil; (V.M.B.); (M.M.D.C.V.)
| | - Somtochukwu A. Evurani
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Nigeria; (O.C.E.); (S.A.E.); (C.N.O.)
| | - Chibundo N. Okorie
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Nigeria; (O.C.E.); (S.A.E.); (C.N.O.)
| | - Victor M. Balcão
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil; (V.M.B.); (M.M.D.C.V.)
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, P-3810-193 Aveiro, Portugal
| | - Marta M. D. C. Vila
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil; (V.M.B.); (M.M.D.C.V.)
| |
Collapse
|
49
|
Qu R, Zhang Y, Ma Y, Zhou X, Sun L, Jiang C, Zhang Z, Fu W. Role of the Gut Microbiota and Its Metabolites in Tumorigenesis or Development of Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205563. [PMID: 37263983 PMCID: PMC10427379 DOI: 10.1002/advs.202205563] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/20/2023] [Indexed: 06/03/2023]
Abstract
Colorectal cancer (CRC) is the most common cancer of the digestive system with high mortality and morbidity rates. Gut microbiota is found in the intestines, especially the colorectum, and has structured crosstalk interactions with the host that affect several physiological processes. The gut microbiota include CRC-promoting bacterial species, such as Fusobacterium nucleatum, Escherichia coli, and Bacteroides fragilis, and CRC-protecting bacterial species, such as Clostridium butyricum, Streptococcus thermophilus, and Lacticaseibacillus paracasei, which along with other microorganisms, such as viruses and fungi, play critical roles in the development of CRC. Different bacterial features are identified in patients with early-onset CRC, combined with different patterns between fecal and intratumoral microbiota. The gut microbiota may be beneficial in the diagnosis and treatment of CRC; some bacteria may serve as biomarkers while others as regulators of chemotherapy and immunotherapy. Furthermore, metabolites produced by the gut microbiota play essential roles in the crosstalk with CRC cells. Harmful metabolites include some primary bile acids and short-chain fatty acids, whereas others, including ursodeoxycholic acid and butyrate, are beneficial and impede tumor development and progression. This review focuses on the gut microbiota and its metabolites, and their potential roles in the development, diagnosis, and treatment of CRC.
Collapse
Affiliation(s)
- Ruize Qu
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Yi Zhang
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Yanpeng Ma
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Xin Zhou
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Lulu Sun
- State Key Laboratory of Women's Reproductive Health and Fertility PromotionPeking UniversityBeijing100191P. R. China
- Department of Endocrinology and MetabolismPeking University Third HospitalBeijing100191P. R. China
| | - Changtao Jiang
- Center of Basic Medical ResearchInstitute of Medical Innovation and ResearchThird HospitalPeking UniversityBeijing100191P. R. China
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesPeking University and the Key Laboratory of Molecular Cardiovascular Science (Peking University)Ministry of EducationBeijing100191P. R. China
- Center for Obesity and Metabolic Disease ResearchSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Zhipeng Zhang
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Wei Fu
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| |
Collapse
|
50
|
Sumiyoshi A, Fujii H, Okuma Y. Targeting microbiome, drug metabolism, and drug delivery in oncology. Adv Drug Deliv Rev 2023; 199:114902. [PMID: 37263544 DOI: 10.1016/j.addr.2023.114902] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
Recent emerging scientific evidence shows a relationship between gut microbiota (GM) and immunomodulation. In the recently published "Hallmarks of Cancer", the microbiome has been reported to play a crucial role in cancer research, and perspectives for its clinical implementation to improve the effectiveness of pharmacotherapy were explored. Several studies have shown that GM can affect the outcomes of pharmacotherapy in cancer, suggesting that GM may affect anti-tumor immunity. Thus, studies on GM that analyze big data using computer-based analytical methods are required. In order to successfully deliver GM to an environment conducive to the proliferation of immune cells both within and outside the tumor microenvironment (TME), it is crucial to address a variety of challenges associated with distinct delivery methods, specifically those pertaining to oral, endoscopic, and intravenous delivery. Clinical trials are in progress to evaluate the effects of targeting GM and whether it can enhance immunity or act on the TME, thereby to improve the clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Ai Sumiyoshi
- Department of Pharmacy, National Cancer Center Hospital 5-1-1 Tsukiji Chuo, Tokyo 104-0045, Japan
| | - Hiroyuki Fujii
- Department of Thoracic Oncology, National Cancer Center Hospital 5-1-1 Tsukiji Chuo, Tokyo 104-0045, Japan; Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital 5-1-1 Tsukiji Chuo, Tokyo 104-0045, Japan.
| |
Collapse
|