1
|
Xu Y, Gao Z, Liu J, Yang Q, Xu S. Role of gut microbiome in suppression of cancers. Gut Microbes 2025; 17:2495183. [PMID: 40254597 PMCID: PMC12013426 DOI: 10.1080/19490976.2025.2495183] [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: 12/14/2024] [Revised: 03/23/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025] Open
Abstract
The pathogenesis of cancer is closely related to the disruption of homeostasis in the human body. The gut microbiome plays crucial roles in maintaining the homeostasis of its host throughout lifespan. In recent years, a large number of studies have shown that dysbiosis of the gut microbiome is involved in the entire process of cancer initiation, development, and prognosis by influencing the host immune system and metabolism. Some specific intestinal bacteria promote the occurrence and development of cancers under certain conditions. Conversely, some other specific intestinal bacteria suppress the oncogenesis and progression of cancers, including inhibiting the occurrence of cancers, delaying the progression of cancers and boosting the therapeutic effect on cancers. The promoting effects of the gut microbiome on cancers have been comprehensively discussed in the previous review. This article will review the latest advances in the roles and mechanisms of gut microbiome in cancer suppression, providing a new perspective for developing strategies of cancer prevention and treatment.
Collapse
Affiliation(s)
- Yao Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Zhaoyu Gao
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Jiaying Liu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Qianqian Yang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Shunjiang Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| |
Collapse
|
2
|
Ding S, Alexander E, Liang H, Kulchar RJ, Singh R, Herzog RW, Daniell H, Leong KW. Synthetic and Biogenic Materials for Oral Delivery of Biologics: From Bench to Bedside. Chem Rev 2025; 125:4009-4068. [PMID: 40168474 DOI: 10.1021/acs.chemrev.4c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
The development of nucleic acid and protein drugs for oral delivery has lagged behind their production for conventional nonoral routes. Over the past decade, the evolution of DNA- and RNA-based technologies combined with the innovation of state-of-the-art delivery vehicles for nucleic acids has brought rapid advancements to the biopharmaceutical field. Nucleic acid therapies have the potential to achieve long-lasting effects, or even cures, by inhibiting or editing genes, which is not possible with conventional small-molecule drugs. However, challenges and limitations must be addressed before these therapies can provide cures for chronic conditions and rare diseases, rather than only offering temporary relief. Nucleic acids and proteins face premature degradation in the acidic, enzyme-rich stomach environment and are rapidly cleared by the liver. To overcome these challenges, various delivery vehicles have been developed to transport therapeutic compounds to the intestines, where the active compounds are released and gut microbiota and mucosal immune system also play an important role. This review provides a comprehensive overview of the promises and pitfalls associated with the oral route of administration of biologics, current delivery systems, applications of orally delivered therapeutics, and the challenges and considerations for translation of nucleic acid and protein therapeutics into clinical practice.
Collapse
Affiliation(s)
- Suwan Ding
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Elena Alexander
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Huiyi Liang
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Rachel J Kulchar
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Rahul Singh
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| |
Collapse
|
3
|
Grilc NK, Kristl J, Zupančič Š. Can polymeric nanofibers effectively preserve and deliver live therapeutic bacteria? Colloids Surf B Biointerfaces 2025; 245:114329. [PMID: 39486375 DOI: 10.1016/j.colsurfb.2024.114329] [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: 08/05/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
Probiotics and live therapeutic bacteria (LTB), their strictly regulated therapeutic counterpart, are increasingly important in treating and preventing biofilm-related diseases. This necessitates new approaches to (i) preserve bacterial viability during manufacturing and storage and (ii) incorporate LTB into delivery systems for enhanced therapeutic efficacy. This review explores advances in probiotic and LTB product development, focusing on preservation, protection, and improved delivery. Preservation of bacteria can be achieved by drying methods that decelerate metabolism. These methods introduce stresses affecting viability which can be mitigated with suitable excipients like polymeric or low molecular weight stabilizers. The review emphasizes the incorporation of LTB into polymer-based nanofibers via electrospinning, enabling simultaneous drying, encapsulation, and delivery system production. Optimization of bacterial survival during electrospinning and storage is discussed, as well as controlled LTB release achievable through formulation design using gel-forming, gastroprotective, mucoadhesive, and pH-responsive polymers. Evaluation of the presence of the actual therapeutic strains, bacterial viability and activity by CFU enumeration or alternative analytical techniques is presented as a key aspect of developing effective and safe formulations with LTB. This review offers insights into designing delivery systems, especially polymeric nanofibers, for preservation and delivery of LTB, guiding readers in developing innovative biotherapeutic delivery systems.
Collapse
Affiliation(s)
- Nina Katarina Grilc
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Julijana Kristl
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Špela Zupančič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia.
| |
Collapse
|
4
|
Papadopoulou D, Chrysikopoulou V, Rampaouni A, Tsoupras A. Antioxidant and anti-inflammatory properties of water kefir microbiota and its bioactive metabolites for health promoting bio-functional products and applications. AIMS Microbiol 2024; 10:756-811. [PMID: 39628717 PMCID: PMC11609422 DOI: 10.3934/microbiol.2024034] [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: 05/22/2024] [Revised: 08/05/2024] [Accepted: 08/28/2024] [Indexed: 12/06/2024] Open
Abstract
Inflammation and oxidative stress are implicated in several chronic disorders, while healthy foods and especially fermented beverages and those containing probiotics can provide anti-inflammatory and antioxidant protection against such manifestations and the associated disorders. Water kefir is such a beverage that is rich in both probiotic microbiota and anti-inflammatory bioactives, with an increasing demand as an alternative to a fermented product based on non-dairy matrix with potential health properties. Within this study, the health-promoting properties of the most representative species and strains of microorganisms present in water kefir grains, as well as the health benefits attributed to the bioactive metabolites produced by each individual strain in a series of their cultures, were thoroughly reviewed. Emphasis was given to the antioxidant, antithrombotic, and anti-inflammatory bio-functionalities of both the cultured microorganisms and the bioactive metabolites produced in each case. Moreover, an extensive presentation of the antioxidant and anti-inflammatory health benefits observed from the overall water kefir cultures and classic water kefir beverages obtained were also conducted. Finally, the use of water kefir for the production of several other bio-functional products, including fermented functional foods, supplements, nutraceuticals, nutricosmetics, cosmeceuticals, and cosmetic applications with anti-inflammatory and antioxidant health promoting potential was also thoroughly discussed. Limitations and future perspectives on the use of water kefir, its microorganisms, and their bioactive metabolites are also outlined.
Collapse
Affiliation(s)
| | | | | | - Alexandros Tsoupras
- Hephaestus Laboratory, School of Chemistry, Faculty of Science, Democritus University of Thrace, Kavala University Campus, 65404, Kavala, Greece
| |
Collapse
|
5
|
Chen A, Zhu J, Liu R, Mei Y, Li L, Fan Y, Ke Y, Liu B, Liu Q. Injectable thermo-sensitive hydrogel enhances anti-tumor potency of engineered Lactococcus lactis by activating dendritic cells and effective memory T cells. Bioact Mater 2024; 37:331-347. [PMID: 38694762 PMCID: PMC11061616 DOI: 10.1016/j.bioactmat.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 05/04/2024] Open
Abstract
Engineered bacteria have shown great potential in cancer immunotherapy by dynamically releasing therapeutic payloads and inducing sustained antitumor immune response with the crosstalk of immune cells. In previous studies, FOLactis was designed, which could secret an encoded fusion protein of Fms-related tyrosine kinase 3 ligand and co-stimulator OX40 ligand, leading to remarkable tumor suppression and exerting an abscopal effect by intratumoral injection. However, it is difficult for intratumoral administration of FOLactis in solid tumors with firm texture or high internal pressure. For patients without lesions such as abdominal metastatic tumors and orthotopic gastric tumors, intratumoral injection is not feasible and peritumoral maybe a better choice. Herein, an engineered bacteria delivery system is constructed based on in situ temperature-sensitive poloxamer 407 hydrogels. Peritumoral injection of FOLactis/P407 results in a 5-fold increase in the proportion of activated DC cells and a more than 2-fold increase in the proportion of effective memory T cells (TEM), playing the role of artificial lymph island. Besides, administration of FOLactis/P407 significantly inhibits the growth of abdominal metastatic tumors and orthotopic gastric tumors, resulting in an extended survival time. Therefore, these findings demonstrate the delivery approach of engineered bacteria based on in situ hydrogel will promote the efficacy and universality of therapeutics.
Collapse
Affiliation(s)
- Aoxing Chen
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, The Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Junmeng Zhu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Rui Liu
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yi Mei
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Lin Li
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yue Fan
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yaohua Ke
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, The Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Qin Liu
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, The Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| |
Collapse
|
6
|
Li Y, Chen G, Hu X, Bao Y, Wu C, Zeng N, Jiang F. Assessing causal relationships between gut microbiota and psoriasis: evidence from two sample Mendelian randomization analysis. Sci Rep 2024; 14:8831. [PMID: 38632320 PMCID: PMC11024213 DOI: 10.1038/s41598-024-59603-5] [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: 09/28/2023] [Accepted: 04/12/2024] [Indexed: 04/19/2024] Open
Abstract
Mounting data hints that the gut microbiota's role may be pivotal in understanding the emergence of psoriasis. However, discerning a direct causal link is yet elusive. In this exploration, we adopted a Mendelian randomization (MR) strategy to probe the prospective causal interplay between the gut's microbial landscape and the predisposition to psoriasis. Genetic markers acting as instrumental variables for gut microbiota were extrapolated from a genome-wide association study (GWAS) encompassing 18,340 individuals. A separate GWAS yielded summary data for psoriasis, which covered 337,159 patients and 433,201 control subjects. The primary analysis hinged on inverse variance weighting (IVW). Additional methods like the weighted median approach and MR-Egger regression were employed to validate the integrity of our findings. Intriguing correlations emerged between psoriasis risk and eight specific bacterial traits. To illustrate: Mollicutes presented an odds ratio (OR) of 1.003 with a 95% confidence interval (CI) spanning 1.001-1.005 (p = 0.016), while the family. Victivallaceae revealed an OR of 0.998 with CI values between 0.997 and 0.999 (p = 0.023). Eubacterium (coprostanoligenes group) revealed an OR of 0.997 with CI values between 0.994 and 0.999 (p = 0.027). Eubacterium (fissicatena group) revealed an OR of 0.997 with CI values between 0.996 and 0.999 (p = 0.005). Holdemania revealed an OR of 1.001 with CI values 1-1.003 (p = 0.034). Lachnospiraceae (NK4A136 group) revealed an OR of 0.997 with CI values between 0.995 and 0.999 (p = 0.046). Lactococcus revealed an OR of 0.998 with CI values between 0.996 and 0.999 (p = 0.008). Tenericutes revealed an OR of 1.003 with CI values between 1.001 and 1.006 (p = 0.016). Sensitivity analysis for these bacterial features yielded congruent outcomes, reinforcing statistically significant ties between the eight bacterial entities and psoriasis. This comprehensive probe underscores emerging evidence pointing towards a plausible causal nexus between diverse gut microbiota and the onset of psoriasis. It beckons further research to unravel the intricacies of how the gut's microbial constituents might sway psoriasis's pathogenesis.
Collapse
Affiliation(s)
- Yuan Li
- Department of Dermatology, The Fifth People's Hospital of Hainan Province, Haikou, China
| | - Gaihe Chen
- Department of Dermatology, The Fifth People's Hospital of Hainan Province, Haikou, China
| | - Xiaohuan Hu
- Department of Dermatology, The Fifth People's Hospital of Hainan Province, Haikou, China
| | - Yunlei Bao
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chuyan Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Ni Zeng
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, China.
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Campos GM, Américo MF, Dos Santos Freitas A, Barroso FAL, da Cruz Ferraz Dutra J, Quaresma LS, Cordeiro BF, Laguna JG, de Jesus LCL, Fontes AM, Birbrair A, Santos TM, Azevedo V. Lactococcus lactis as an Interleukin Delivery System for Prophylaxis and Treatment of Inflammatory and Autoimmune Diseases. Probiotics Antimicrob Proteins 2024; 16:352-366. [PMID: 36746838 PMCID: PMC9902259 DOI: 10.1007/s12602-023-10041-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2023] [Indexed: 02/08/2023]
Abstract
Target delivery of therapeutic agents with anti-inflammatory properties using probiotics as delivery and recombinant protein expression vehicles is a promising approach for the prevention and treatment of many diseases, such as cancer and intestinal immune disorders. Lactococcus lactis, a Lactic Acid Bacteria (LAB) widely used in the dairy industry, is one of the most important microorganisms with GRAS status for human consumption, for which biotechnological tools have already been developed to express and deliver recombinant biomolecules with anti-inflammatory properties. Cytokines, for example, are immune system communication molecules present at virtually all levels of the immune response. They are essential in cellular and humoral processes, such as hampering inflammation or adjuvating in the adaptive immune response, making them good candidates for therapeutic approaches. This review discusses the advances in the development of new therapies and prophylactic approaches using LAB to deliver/express cytokines for the treatment of inflammatory and autoimmune diseases in the future.
Collapse
Affiliation(s)
- Gabriela Munis Campos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Monique Ferrary Américo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Andria Dos Santos Freitas
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Joyce da Cruz Ferraz Dutra
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ludmila Silva Quaresma
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bárbara Fernandes Cordeiro
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana Guimarães Laguna
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luís Cláudio Lima de Jesus
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aparecida Maria Fontes
- Genetics Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Alexander Birbrair
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Tulio Marcos Santos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Uniclon Biotecnologia, Belo Horizonte, MG, Brazil
| | - Vasco Azevedo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
8
|
Kim MG, Kim S, Jeon JY, Moon SJ, Kwak YG, Na JY, Lee S, Park KM, Kim HJ, Lee SM, Choi SY, Shin KH. Profiling of endogenous metabolites and changes in intestinal microbiota distribution after GEN-001 ( Lactococcus lactis) administration. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:153-164. [PMID: 38414398 PMCID: PMC10902589 DOI: 10.4196/kjpp.2024.28.2.153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 02/29/2024]
Abstract
This study aimed to identify metabolic biomarkers and investigate changes in intestinal microbiota in the feces of healthy participants following administration of Lactococcus lactis GEN-001. GEN-001 is a single-strain L. lactis strain isolated from the gut of a healthy human volunteer. The study was conducted as a parallel, randomized, phase 1, open design trial. Twenty healthy Korean males were divided into five groups according to the GEN-001 dosage and dietary control. Groups A, B, C, and D1 received 1, 3, 6, and 9 GEN-001 capsules (1 × 1011 colony forming units), respectively, without dietary adjustment, whereas group D2 received 9 GEN-001 capsules with dietary adjustment. All groups received a single dose. Fecal samples were collected 2 days before GEN-001 administration to 7 days after for untargeted metabolomics and gut microbial metagenomic analyses; blood samples were collected simultaneously for immunogenicity analysis. Levels of phenylalanine, tyrosine, cholic acid, deoxycholic acid, and tryptophan were significantly increased at 5-6 days after GEN-001 administration when compared with predose levels. Compared with predose, the relative abundance (%) of Parabacteroides and Alistipes significantly decreased, whereas that of Lactobacillus and Lactococcus increased; Lactobacillus and tryptophan levels were negatively correlated. A single administration of GEN-001 shifted the gut microbiota in healthy volunteers to a more balanced state as evidenced by an increased abundance of beneficial bacteria, including Lactobacillus, and higher levels of the metabolites that have immunogenic properties.
Collapse
Affiliation(s)
- Min-Gul Kim
- Center for Clinical Pharmacology, Jeonbuk National University Hospital, Jeonju 54907, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea
- Department of Pharmacology, School of Medicine, Jeonbuk National University, Jeonju 54907, Korea
| | - Suin Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Ji-Young Jeon
- Center for Clinical Pharmacology, Jeonbuk National University Hospital, Jeonju 54907, Korea
| | - Seol Ju Moon
- Center for Clinical Pharmacology, Jeonbuk National University Hospital, Jeonju 54907, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea
| | - Yong-Geun Kwak
- Center for Clinical Pharmacology, Jeonbuk National University Hospital, Jeonju 54907, Korea
- Department of Pharmacology, School of Medicine, Jeonbuk National University, Jeonju 54907, Korea
| | - Joo Young Na
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | | | | | - Sang-Min Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Seo-Yeon Choi
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Kwang-Hee Shin
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
9
|
Chen Y, Wang X, Ye Y, Ren Q. Gut microbiota in cancer: insights on microbial metabolites and therapeutic strategies. Med Oncol 2023; 41:25. [PMID: 38129370 DOI: 10.1007/s12032-023-02249-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/11/2023] [Indexed: 12/23/2023]
Abstract
In recent years, the role of gut microbiota in cancer treatment has attracted substantial attention. It is now well established that gut microbiota and its metabolites significantly contribute to the incidence, treatment, and prognosis of various cancers. This review provides a comprehensive review on the pivotal role of gut microbiota and their metabolites in cancer initiation and progression. Furthermore, it evaluates the impact of gut microbiota on the efficacy and associated side effects of anticancer therapies, including radiotherapy, chemotherapy, and immunotherapy, thus emphasizing the clinical importance of gut microbiota reconstitution in cancer treatment.
Collapse
Affiliation(s)
- Yalan Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Xibin Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Yuwei Ye
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Gansu Province Clinical Research Center for Digestive Diseases, Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Qian Ren
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China.
- Gansu Province Clinical Research Center for Digestive Diseases, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
10
|
Romero Zoghbi SE, Martín Martín M, García JC, Valero M, Rincón D, Peña Huertas M, Fusco JP, López E, Zafra J, Fernández Luengas D, López Campos F, Couñago F. Total Neoadjuvant Therapy Approach for the Treatment of Locally Advanced Rectal Cancer: Where Do We Stand? Oncology 2023; 102:646-658. [PMID: 37935161 PMCID: PMC11216349 DOI: 10.1159/000534888] [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: 08/21/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND For the management of locally advanced rectal cancer (LARC), initial treatment with neoadjuvant chemoradiotherapy followed by surgery and chemotherapy in selected patients is considered one of the recommended options by the main international clinical guidelines. Nonetheless, the administration of all chemotherapy before definitive treatment (total neoadjuvant therapy [TNT]) is an optimal alternative with a growing level of evidence that must be evaluated in multidisciplinary boards. This review summarizes the available data and controversies in this scenario. SUMMARY We have analyzed the characteristics of the main published studies that assess the use of TNT in patients with LARC, evaluating their inclusion criteria and distinguishing between the employed radiotherapy fractionations, systemic agents, timing, and the implications of these treatments with regard to surgery and long-term oncological results. Our aim was to describe the evidence that supports the use of a specific regime in everyday clinical practice. KEY POINTS There is solid evidence for the use of TNT in patients with LARC. There are no data indicating the superiority of one specific TNT scheme among all the existing options. International clinical guidelines leave the door open to choose the most adequate treatment based on the clinical and pathological characteristics of each patient. This review shows the different approaches to TNT and assesses the best options based on clinical evidence.
Collapse
Affiliation(s)
| | | | - Juan Carlos García
- Department of General and Digestive Surgery, Hospital Universitario Ramón y Cajal, Madrid,Spain
| | - Mireia Valero
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Diego Rincón
- Department of Radiation Oncology, GenesisCare, Hospital Universitario Vithas Madrid La Milagrosa, Madrid, Spain
| | - Marina Peña Huertas
- Department of Radiation Oncology, Hospital Universitario Quirónsalud, Madrid, Spain
| | - Juan Pablo Fusco
- Department of Medical Oncology, Hospital Universitario La Luz, Madrid, Spain
| | - Escarlata López
- Department of Radiation Oncology, GenesisCare, Hospital Universitario Vithas Madrid La Milagrosa, Madrid, Spain
| | - Juan Zafra
- Department of Radiation Oncology, Hospital Clínico Universitario Virgen de la Victoria, Málaga, Spain
- Group of Translational Research in Cancer Immunotherapy (CIMO 2), Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - David Fernández Luengas
- Department of General and Digestive Surgery, Hospital Universitario Quirónsalud, Madrid, Spain
| | - Fernando López Campos
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Felipe Couñago
- Department of Radiation Oncology, GenesisCare, Hospital Universitario Vithas Madrid La Milagrosa, Madrid, Spain
| |
Collapse
|
11
|
Guo C, Kong L, Xiao L, Liu K, Cui H, Xin Q, Gu X, Jiang C, Wu J. The impact of the gut microbiome on tumor immunotherapy: from mechanism to application strategies. Cell Biosci 2023; 13:188. [PMID: 37828613 PMCID: PMC10571290 DOI: 10.1186/s13578-023-01135-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 09/15/2023] [Indexed: 10/14/2023] Open
Abstract
Immunotherapy is one of the fastest developing areas in the field of oncology. Many immunological treatment strategies for refractory tumors have been approved and marketed. Nevertheless, much clinical and preclinical experimental evidence has shown that the efficacy of immunotherapy in tumor treatment varies markedly among individuals. The commensal microbiome mainly colonizes the intestinal lumen in humans, is affected by a variety of factors and exhibits individual variation. Moreover, the gut is considered the largest immune organ of the body due to its influence on the immune system. In the last few decades, with the development of next-generation sequencing (NGS) techniques and in-depth research, the view that the gut microbiota intervenes in antitumor immunotherapy through the immune system has been gradually confirmed. Here, we review important studies published in recent years focusing on the influences of microbiota on immune system and the progression of malignancy. Furthermore, we discuss the mechanism by which microbiota affect tumor immunotherapy, including immune checkpoint blockade (ICB) and adoptive T-cell therapy (ACT), and strategies for modulating the microbial composition to facilitate the antitumor immune response. Finally, opportunity and some challenges are mentioned to enable a more systematic understanding of tumor treatment in the future and promote basic research and clinical application in related fields.
Collapse
Affiliation(s)
- Ciliang Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Lingkai Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Lingjun Xiao
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Kua Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Huawei Cui
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
| | - Qilei Xin
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Qingdao Road 3716#, Huaiyin District, Jinan, Shandong, China
| | - Xiaosong Gu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Qingdao Road 3716#, Huaiyin District, Jinan, Shandong, China
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Qingdao Road 3716#, Huaiyin District, Jinan, Shandong, China.
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, 22 Hankou Road, Nanjing, 210093, Jiangsu, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Qingdao Road 3716#, Huaiyin District, Jinan, Shandong, China.
| |
Collapse
|
12
|
Huang J, Gong C, Zhou A. Modulation of gut microbiota: a novel approach to enhancing the effects of immune checkpoint inhibitors. Ther Adv Med Oncol 2023; 15:17588359231204854. [PMID: 37841750 PMCID: PMC10571694 DOI: 10.1177/17588359231204854] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have greatly improved the prognosis of some cancer patients, the majority still fail to respond adequately, and the available biomarkers cannot reliably predict drug efficacy. The gut microbiota has received widespread attention among the various intrinsic and extrinsic factors contributing to drug resistance. As an essential regulator of physiological function, the impact of gut microbiota on host immunity and response to cancer therapy is increasingly recognized. Several studies have demonstrated significant differences in gut microbiota between responders and nonresponders. The gut microbiota associated with better clinical outcomes is called 'favorable gut microbiota'. Significantly, interventions can alter the gut microbiota. By shifting the gut microbiota to the 'favorable' one through various modifications, preclinical and clinical studies have yielded more pronounced responses and better clinical outcomes when combined with ICIs treatment, providing novel approaches to improve the efficacy of cancer immunotherapy. These findings may be attributed to the effects of gut microbiota and its metabolites on the immune microenvironment and the systemic immune system, but the underlying mechanisms remain to be discovered. In this review, we summarize the clinical evidence that the gut microbiota is strongly associated with the outcomes of ICI treatment and describe the gut microbiota characteristics associated with better clinical outcomes. We then expand on the current prevalent modalities of gut microbiota regulation, provide a comprehensive overview of preclinical and clinical research advances in improving the therapeutic efficacy and prognosis of ICIs by modulating gut microbiota, and suggest fundamental questions we need to address and potential directions for future research expansion.
Collapse
Affiliation(s)
- Jinglong Huang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caifeng Gong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100020, China
| |
Collapse
|
13
|
Wang M, Zhang L, Chang W, Zhang Y. The crosstalk between the gut microbiota and tumor immunity: Implications for cancer progression and treatment outcomes. Front Immunol 2023; 13:1096551. [PMID: 36726985 PMCID: PMC9885097 DOI: 10.3389/fimmu.2022.1096551] [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: 11/12/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
The gastrointestinal tract is inhabited by trillions of commensal microorganisms that constitute the gut microbiota. As a main metabolic organ, the gut microbiota has co-evolved in a symbiotic relationship with its host, contributing to physiological homeostasis. Recent advances have provided mechanistic insights into the dual role of the gut microbiota in cancer pathogenesis. Particularly, compelling evidence indicates that the gut microbiota exerts regulatory effects on the host immune system to fight against cancer development. Some microbiota-derived metabolites have been suggested as potential activators of antitumor immunity. On the contrary, the disequilibrium of intestinal microbial communities, a condition termed dysbiosis, can induce cancer development. The altered gut microbiota reprograms the hostile tumor microenvironment (TME), thus allowing cancer cells to avoid immunosurvelliance. Furthermore, the gut microbiota has been associated with the effects and complications of cancer therapy given its prominent immunoregulatory properties. Therapeutic measures that aim to manipulate the interplay between the gut microbiota and tumor immunity may bring new breakthroughs in cancer treatment. Herein, we provide a comprehensive update on the evidence for the implication of the gut microbiota in immune-oncology and discuss the fundamental mechanisms underlying the influence of intestinal microbial communities on systemic cancer therapy, in order to provide important clues toward improving treatment outcomes in cancer patients.
Collapse
|
14
|
Conti G, D’Amico F, Fabbrini M, Brigidi P, Barone M, Turroni S. Pharmacomicrobiomics in Anticancer Therapies: Why the Gut Microbiota Should Be Pointed Out. Genes (Basel) 2022; 14:55. [PMID: 36672796 PMCID: PMC9859289 DOI: 10.3390/genes14010055] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/09/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Anticancer treatments have shown a variable therapeutic outcome that may be partly attributable to the activity of the gut microbiota on the pathology and/or therapies. In recent years, microbiota-drug interactions have been extensively investigated, but most of the underlying molecular mechanisms still remain unclear. In this review, we discuss the relationship between the gut microbiota and some of the most commonly used drugs in oncological diseases. Different strategies for manipulating the gut microbiota layout (i.e., prebiotics, probiotics, antibiotics, and fecal microbiota transplantation) are then explored in order to optimize clinical outcomes in cancer patients. Anticancer technologies that exploit tumor-associated bacteria to target tumors and biotransform drugs are also briefly discussed. In the field of pharmacomicrobiomics, multi-omics strategies coupled with machine and deep learning are urgently needed to bring to light the interaction among gut microbiota, drugs, and host for the development of truly personalized precision therapies.
Collapse
Affiliation(s)
- Gabriele Conti
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Federica D’Amico
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Marco Fabbrini
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Monica Barone
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|