The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer DOI
Gauri Mirji,

Alison Worth,

Sajad A. Bhat

и другие.

Science Immunology, Год журнала: 2022, Номер 7(75)

Опубликована: Сен. 9, 2022

The composition of the gut microbiome can control innate and adaptive immunity has emerged as a key regulator tumor growth, especially in context immune checkpoint blockade (ICB) therapy. However, underlying mechanisms for how affects growth remain unclear. Pancreatic ductal adenocarcinoma (PDAC) tends to be refractory therapy, including ICB. Using nontargeted, liquid chromatography–tandem mass spectrometry–based metabolomic screen, we identified microbe–derived metabolite trimethylamine N -oxide (TMAO), which enhanced antitumor PDAC. Delivery TMAO intraperitoneally or via dietary choline supplement orthotopic PDAC-bearing mice reduced associated with an immunostimulatory tumor-associated macrophage (TAM) phenotype, activated effector T cell response microenvironment. Mechanistically, potentiated type I interferon (IFN) pathway conferred effects IFN–dependent manner. Delivering TMAO-primed macrophages intravenously produced similar effects. Combining ICB (anti-PD1 and/or anti-Tim3) mouse model PDAC significantly burden improved survival beyond alone. Last, levels bacteria containing CutC (an enzyme that generates trimethylamine, precursor) correlated long-term patients anti-PD1 melanoma. Together, our study identifies microbial driver lays groundwork potential therapeutic strategies targeting TMAO.

Язык: Английский

Cancer pharmacomicrobiomics: targeting microbiota to optimise cancer therapy outcomes DOI Creative Commons

Nick Lung-Ngai Ting,

Harry Cheuk-Hay Lau, Jun Yu

и другие.

Gut, Год журнала: 2022, Номер 71(7), С. 1412 - 1425

Опубликована: Март 11, 2022

Despite the promising advances in novel cancer therapy such as immune checkpoint inhibitors (ICIs), limitations including therapeutic resistance and toxicity remain. In recent years, relationship between gut microbiota has been extensively studied. Accumulating evidence reveals role of defining efficacy toxicity. Unlike host genetics, can be easily modified via multiple strategies, faecal transplantation (FMT), probiotics antibiotics. Preclinical studies have identified mechanisms on how microbes influence treatment outcomes. Clinical trials also demonstrated potential modulation treatments. Herein, we review mechanistic insights microbial interactions with chemotherapy ICIs, particularly focusing interplay bacteria pharmacokinetics (eg, metabolism, enzymatic degradation) or pharmacodynamics immunomodulation) treatment. The translational basic findings clinical settings is then explored, using predictive biomarkers by antibiotics, probiotics, prebiotics, dietary modulations FMT. We further discuss current patients suggest essential directions for future study. era personalised medicine, it crucial to understand its cancer. Manipulating augment responses provide new into

Язык: Английский

Процитировано

182

Chemically and Biologically Engineered Bacteria‐Based Delivery Systems for Emerging Diagnosis and Advanced Therapy DOI
Zhaoting Li, Yixin Wang, Jun Liu

и другие.

Advanced Materials, Год журнала: 2021, Номер 33(38)

Опубликована: Авг. 4, 2021

Abstract Bacteria are one of the main groups organisms, which dynamically and closely participate in human health disease development. With integration chemical biotechnology, bacteria have been utilized as an emerging delivery system for various biomedical applications. Given unique features such their intrinsic biocompatibility motility, bacteria‐based systems drawn wide interest diagnosis treatment diseases, including cancer, infectious kidney failure, hyperammonemia. Notably, at interface biotechnology bacteria, many research opportunities initiated, opening a promising frontier application. Herein, current synergy design principles systems, microbial modulation, clinical translation reviewed, with special focus on advances therapy.

Язык: Английский

Процитировано

181

CD8+ T cells in the cancer-immunity cycle DOI Creative Commons
Josephine R. Giles, Anna-Maria Globig,

Susan M. Kaech

и другие.

Immunity, Год журнала: 2023, Номер 56(10), С. 2231 - 2253

Опубликована: Окт. 1, 2023

Язык: Английский

Процитировано

176

Role of the gut microbiota in anticancer therapy: from molecular mechanisms to clinical applications DOI Creative Commons
Lin-Yong Zhao,

Jia-Xin Mei,

Gang Yu

и другие.

Signal Transduction and Targeted Therapy, Год журнала: 2023, Номер 8(1)

Опубликована: Май 13, 2023

Abstract In the past period, due to rapid development of next-generation sequencing technology, accumulating evidence has clarified complex role human microbiota in cancer and therapeutic response. More importantly, available seems indicate that modulating composition gut improve efficacy anti-cancer drugs may be feasible. However, intricate complexities exist, a deep comprehensive understanding how interacts with is critical realize its full potential treatment. The purpose this review summarize initial clues on molecular mechanisms regarding mutual effects between development, highlight relationship microbes immunotherapy, chemotherapy, radiation therapy surgery, which provide insights into formulation individualized strategies for management. addition, current emerging microbial interventions as well their clinical applications are summarized. Although many challenges remain now, great importance cannot overstated strategies, it necessary explore holistic approach incorporates modulation cancer.

Язык: Английский

Процитировано

175

The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer DOI
Gauri Mirji,

Alison Worth,

Sajad A. Bhat

и другие.

Science Immunology, Год журнала: 2022, Номер 7(75)

Опубликована: Сен. 9, 2022

The composition of the gut microbiome can control innate and adaptive immunity has emerged as a key regulator tumor growth, especially in context immune checkpoint blockade (ICB) therapy. However, underlying mechanisms for how affects growth remain unclear. Pancreatic ductal adenocarcinoma (PDAC) tends to be refractory therapy, including ICB. Using nontargeted, liquid chromatography–tandem mass spectrometry–based metabolomic screen, we identified microbe–derived metabolite trimethylamine N -oxide (TMAO), which enhanced antitumor PDAC. Delivery TMAO intraperitoneally or via dietary choline supplement orthotopic PDAC-bearing mice reduced associated with an immunostimulatory tumor-associated macrophage (TAM) phenotype, activated effector T cell response microenvironment. Mechanistically, potentiated type I interferon (IFN) pathway conferred effects IFN–dependent manner. Delivering TMAO-primed macrophages intravenously produced similar effects. Combining ICB (anti-PD1 and/or anti-Tim3) mouse model PDAC significantly burden improved survival beyond alone. Last, levels bacteria containing CutC (an enzyme that generates trimethylamine, precursor) correlated long-term patients anti-PD1 melanoma. Together, our study identifies microbial driver lays groundwork potential therapeutic strategies targeting TMAO.

Язык: Английский

Процитировано

174