1
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Tadepalli S, Clements DR, Raquer-McKay HM, Lüdtke A, Saravanan S, Seong D, Vitek L, Richards CM, Carette JE, Mack M, Gottfried-Blackmore A, Graves EE, Idoyaga J. CD301b+ monocyte-derived dendritic cells mediate resistance to radiotherapy. J Exp Med 2025; 222:e20231717. [PMID: 40146036 PMCID: PMC11949126 DOI: 10.1084/jem.20231717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/14/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Monocytes infiltrating tumors acquire various states that distinctly impact cancer treatment. Here, we show that resistance of tumors to radiotherapy (RT) is controlled by the accumulation of monocyte-derived dendritic cells (moDCs). These moDCs are characterized by the expression of CD301b and have a superior capacity to generate regulatory T cells (Tregs). Accordingly, moDC depletion limits Treg generation and improves the therapeutic outcome of RT. Mechanistically, we demonstrate that granulocyte-macrophage colony-stimulating factor (GM-CSF) derived from radioresistant tumor cells following RT is necessary for the accumulation of moDCs. Our results unravel the immunosuppressive function of moDCs and identify GM-CSF as an immunotherapeutic target during RT.
Collapse
Affiliation(s)
- Sirimuvva Tadepalli
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek R. Clements
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Hayley M. Raquer-McKay
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Anja Lüdtke
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Sanjana Saravanan
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - David Seong
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Lorraine Vitek
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher M. Richards
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jan E. Carette
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Andres Gottfried-Blackmore
- Department of Pharmacology, University of California San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, Division of Gastroenterology, University of California San Diego School of Medicine, San Diego, CA, USA
- Gastroenterology Section, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Edward E. Graves
- Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pharmacology, University of California San Diego School of Medicine, San Diego, CA, USA
- Department of Molecular Biology, University of California San Diego School of Biological Sciences, San Diego, CA, USA
| |
Collapse
|
3
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 PMCID: PMC12067981 DOI: 10.1016/j.addr.2025.115579] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
4
|
Zhang X, Fam KT, Dai T, Hang HC. Microbiota mechanisms in cancer progression and therapy. Cell Chem Biol 2025; 32:653-677. [PMID: 40334660 DOI: 10.1016/j.chembiol.2025.04.005] [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: 11/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
The composition of the microbiota in patients has been shown to correlate with cancer progression and response to therapy, highlighting unique opportunities to improve patient outcomes. In this review, we discuss the challenges and advancements in understanding the chemical mechanisms of specific microbiota species, pathways, and molecules involved in cancer progression and treatment. We also describe the modulation of cancer and immunotherapy by the microbiota, along with approaches for investigating microbiota enzymes and metabolites. Elucidating these specific microbiota mechanisms and molecules should offer new opportunities for developing enhanced diagnostics and therapeutics to improve outcomes for cancer patients. Nonetheless, many microbiota mechanisms remain to be determined and require innovative chemical genetic approaches.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Kyong Tkhe Fam
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Tingting Dai
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA; Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
5
|
Fernandez E, Wargo JA, Helmink BA. The Microbiome and Cancer: A Translational Science Review. JAMA 2025:2833859. [PMID: 40354071 DOI: 10.1001/jama.2025.2191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Importance Growing evidence suggests that microbes located within the gastrointestinal tract and other anatomical locations influence the development and progression of diseases such as cancer. Observations Clinical and preclinical evidence suggests that microbes in the gastrointestinal tract and other anatomical locations, such as the respiratory tract, may affect carcinogenesis, development of metastases, cancer treatment response, and cancer treatment-related adverse effects. Within tumors of patients with cancer, microbes may affect response to treatment, and therapies that reduce or eliminate these microbes may improve outcomes in patients with cancer. Modulating gastrointestinal tract (gut) microbes through fecal microbiota transplant and other strategies such as dietary intervention (eg, high-fiber diet intervention) has improved outcomes in small studies of patients treated with cancer immunotherapy. In contrast, disruption of the gut microbiota by receipt of broad-spectrum antibiotics prior to treatment with cancer immunotherapy has been associated with poorer overall survival and higher rates of adverse effects in patients treated with immune checkpoint blockade for solid tumors and also with chimeric antigen receptor T-cell therapy for hematologic malignancies. Conclusions and Relevance Microbes in the gut and other locations in the body may influence the development and progression of cancer and may affect the response to adverse effects from cancer therapy. Future therapies targeting microbes in the gut and other locations in the body could potentially improve outcomes in patients with cancer.
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Beth A Helmink
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
6
|
Zareie P, Weiss ES, Kaplan DH, Mackay LK. Cutaneous T cell immunity. Nat Immunol 2025:10.1038/s41590-025-02145-3. [PMID: 40335684 DOI: 10.1038/s41590-025-02145-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/13/2025] [Indexed: 05/09/2025]
Abstract
The skin is the primary barrier against environmental insults, safeguarding the body from mechanical, chemical and pathogenic threats. The frequent exposure of the skin to environmental challenges requires an immune response that incorporates a sophisticated combination of defenses. Tissue-resident lymphocytes are pivotal for skin immunity, working in tandem with commensal bacteria to maintain immune surveillance and homeostasis, as well as participating in the pathogenesis of several skin diseases. Indeed, it has been estimated that the human skin harbors nearly twice as many T cells as found in the circulation. Effective treatment of skin diseases and new therapy development require a thorough understanding of the complex interactions among skin tissue, immune cells and the microbiota, which together regulate the skin's immune balance. This Review explores the latest developments and understanding of this critical barrier organ, with a specific focus on the role of skin-resident T cells.
Collapse
Affiliation(s)
- Pirooz Zareie
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Eric S Weiss
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel H Kaplan
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
7
|
Wiull K, Kjos M, Eijsink VGH, Mathiesen G. An inverse relationship between fitness and secretion efficiency in a gram-positive bacterium. PNAS NEXUS 2025; 4:pgaf131. [PMID: 40321417 PMCID: PMC12046399 DOI: 10.1093/pnasnexus/pgaf131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Lactiplantibacillus plantarum and other lactic acid bacteria are emerging as promising candidates for mucosal delivery of surface-displayed antigens. However, producing secreted heterologous proteins and anchoring these using LPxTG anchors can significantly reduce bacterial fitness. To understand the underlying mechanisms and limiting factors, we analyzed 11 recombinant L. plantarum strains expressing the HaloTag reporter protein with the same LPxTG anchor but varying signal peptides. By labeling the reporter protein with fluorescent ligands, this approach allowed simultaneous detection of correctly folded intracellular and extracellular protein, revealing signal peptide-dependent variation in the relative amounts of intra- and extracellularly folded HaloTag. Furthermore, this analysis uncovered an unexpected correlation between secretion efficiency and bacterial fitness. Strains with better growth showed more premature intracellular folding and reduced protein translocation and surface display. Conversely, strains with a higher fraction of surface-displayed protein, i.e. strains with greater secretion efficiency, exhibited impaired growth, likely due to increased interactions between the signal peptide and the secretion machinery, leading to secretion overload. Correlation analyses and confirmation of observed correlations by mutational studies of the signal peptides showed that signal peptide hydrophobicity is positively correlated with higher secretion efficiency but is accompanied by a trade-off in fitness. These findings underscore the critical role of signal peptides in balancing protein secretion and bacterial viability, offering valuable insights for optimizing protein secretion and anchoring in gram-positive bacteria.
Collapse
Affiliation(s)
- Kamilla Wiull
- Faculty of Chemistry, Biotechnology and Food Science, NMBU—Norwegian University of Life Sciences, Ås 1433, Norway
| | - Morten Kjos
- Faculty of Chemistry, Biotechnology and Food Science, NMBU—Norwegian University of Life Sciences, Ås 1433, Norway
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, NMBU—Norwegian University of Life Sciences, Ås 1433, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, NMBU—Norwegian University of Life Sciences, Ås 1433, Norway
| |
Collapse
|
8
|
Liu M, Blattman SB, Takahashi M, Mandayam N, Jiang W, Oikonomou P, Tavazoie SF, Tavazoie S. Conserved genetic basis for microbial colonization of the gut. Cell 2025; 188:2505-2520.e22. [PMID: 40187346 PMCID: PMC12048274 DOI: 10.1016/j.cell.2025.03.010] [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: 08/26/2024] [Revised: 12/02/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
Despite the fundamental importance of gut microbes, the genetic basis of their colonization remains largely unexplored. Here, by applying cross-species genotype-habitat association at the tree-of-life scale, we identify conserved microbial gene modules associated with gut colonization. Across thousands of species, we discovered 79 taxonomically diverse putative colonization factors organized into operonic and non-operonic modules. They include previously characterized colonization pathways such as autoinducer-2 biosynthesis and novel processes including tRNA modification and translation. In vivo functional validation revealed YigZ (IMPACT family) and tRNA hydroxylation protein-P (TrhP) are required for E. coli intestinal colonization. Overexpressing YigZ alone is sufficient to enhance colonization of the poorly colonizing MG1655 E. coli by >100-fold. Moreover, natural allelic variations in YigZ impact inter-strain colonization efficiency. Our findings highlight the power of large-scale comparative genomics in revealing the genetic basis of microbial adaptations. These broadly conserved colonization factors may prove critical for understanding gastrointestinal (GI) dysbiosis and developing therapeutics.
Collapse
Affiliation(s)
- Menghan Liu
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Sydney B Blattman
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Mai Takahashi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Nandan Mandayam
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Wenyan Jiang
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Panos Oikonomou
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Saeed Tavazoie
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
9
|
Patra D, Dev G, Hand TW, Overacre-Delgoffe A. Friends close, enemies closer: the complex role of the microbiome in antitumor immunity. Curr Opin Immunol 2025; 93:102537. [PMID: 40015179 DOI: 10.1016/j.coi.2025.102537] [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: 08/27/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Immunotherapy has achieved remarkable advances in cancer treatment by harnessing the immune system to combat tumors, yet its effectiveness remains inconsistent across patients and tumor types. The microbiota, a diverse assemblage of microorganisms residing at host barrier surfaces, is pivotal in shaping immune responses. This review explores the direct and indirect mechanisms via which the microbiota modulates antitumor immune responses both locally within the tumor microenvironment and systemically by affecting distant tumors. We discuss recent findings linking microbiota-derived metabolites and microbiota-derived antigens with antitumor immunity and immunotherapy response. Additionally, we discuss recent advances in microbiome-based therapies, including fecal microbiota transplantation. We propose the use and development of new analytical techniques to further characterize the complex functions and interactions between the microbiome and immune system. To conclude, we outline recommendations for future research and therapeutic approaches to leverage the microbiome to improve current immunotherapies.
Collapse
Affiliation(s)
- Dipyaman Patra
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, USA
| | - Gagan Dev
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Timothy W Hand
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA.
| | - Abigail Overacre-Delgoffe
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, USA.
| |
Collapse
|
10
|
Pérez Escriva P, Correia Tavares Bernardino C, Letellier E. De-coding the complex role of microbial metabolites in cancer. Cell Rep 2025; 44:115358. [PMID: 40023841 DOI: 10.1016/j.celrep.2025.115358] [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: 08/09/2024] [Revised: 12/11/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
The human microbiome, an intricate ecosystem of trillions of microbes residing across various body sites, significantly influences cancer, a leading cause of morbidity and mortality worldwide. Recent studies have illuminated the microbiome's pivotal role in cancer development, either through direct cellular interactions or by secreting bioactive compounds such as metabolites. Microbial metabolites contribute to cancer initiation through mechanisms such as DNA damage, epithelial barrier dysfunction, and chronic inflammation. Furthermore, microbial metabolites exert dual roles on cancer progression and response to therapy by modulating cellular metabolism, gene expression, and signaling pathways. Understanding these complex interactions is vital for devising new therapeutic strategies. This review highlights microbial metabolites as promising targets for cancer prevention and treatment, emphasizing their impact on therapy responses and underscoring the need for further research into their roles in metastasis and therapy resistance.
Collapse
Affiliation(s)
- Pau Pérez Escriva
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Catarina Correia Tavares Bernardino
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
11
|
Wang L, Zhao H, Wu F, Chen J, Xu H, Gong W, Wen S, Yang M, Xia J, Chen Y, Chen D. Bile-Liver phenotype: Exploring the microbiota landscape in bile and intratumor of cholangiocarcinoma. Comput Struct Biotechnol J 2025; 27:1173-1186. [PMID: 40206347 PMCID: PMC11981758 DOI: 10.1016/j.csbj.2025.03.030] [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: 12/11/2024] [Revised: 03/02/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Cholangiocarcinoma (CCA) arises within the peritumoral bile microenvironment, yet microbial translocation from bile to intracholangiocarcinoma (IntraCCA) tissues remains poorly understood. Previous studies on bile microbiota alterations from biliary benign disease (BBD) to CCA have yielded inconsistent results, highlighting the need for cross-study analysis. We presented a comprehensive analysis of five cohorts (N = 266), including our newly established 16S rRNA gene profiling (n = 42), to elucidate these microbiota transitions. The concordance of bacteria between CCA bile and intraCCA tissue, represented by Enterococcus and Staphylococcus, suggested microbiota migration from bile to intratumoral tissues. A computational random forest machine learning model effectively distinguished intraCCA tissue from CCA bile, identifying Rhodococcus and Ralstonia as diagnostically significant. The model also excelled in differentiating CCA bile from BBD bile, achieving an AUC value of 0.931 in external validation. Using unsupervised hierarchical clustering, we established Biletypes based on microbial signatures in our cohort. A combination of 17 genera effectively stratified patients into Biletype A and Biletype B. Biletype B robustly discerned CCA from BBD, with Sub-Biletype B1 correlating with advanced TNM stage and poorer prognosis. Among the 17 genera, bacterial Cluster 1, composed of Sphingomonas, Staphylococcus, Massilia, Paenibacillus, Porphyrobacter, Lawsonella, and Aerococcus, was enriched in Biletype B1 and predicted CCA with an AUC of 0.96. Staphylococcus emerged as a promising single-genus predictor for CCA diagnosis and staging. In conclusion, this study delineates a potential microbiota transition pathway from the gut through CCA bile to intra-CCA tissue, proposing Biletypes and Staphylococcus as biomarkers for CCA prognosis.
Collapse
Affiliation(s)
- Lei Wang
- Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
- Department of Hepatopancreatobiliary Surgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Hui Zhao
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Fan Wu
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Jiale Chen
- Department of Hospital Infection Management,Wujin Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Changzhou 213161, China
| | - Hanjie Xu
- Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, China
| | - Wanwan Gong
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Sijia Wen
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Mengmeng Yang
- Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Jiazeng Xia
- Department of General Surgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China
- Department of General Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Yu Chen
- Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, China
| | - Daozhen Chen
- Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
- Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, China
| |
Collapse
|
12
|
Wiull K, Holmgren E, Svensson S, Eijsink VGH, Grönlund H, Mathiesen G. Surface display of two neoantigens on Lactiplantibacillus plantarum. Sci Rep 2025; 15:7769. [PMID: 40044906 PMCID: PMC11882890 DOI: 10.1038/s41598-025-91180-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
Lactic acid bacteria, such as Lactiplantibacillus plantarum, are becoming increasingly popular hosts for combining production and delivery of therapeutic proteins to immune cells. Soluble antigens are susceptible to rapid proteolysis, hence anchoring of antigens to bacterial cells, which likely protects the antigen, is a preferred delivery strategy that may increase immune responses. In cancer research, personalized immunotherapy has high potential and, in this respect, the so-called neoantigens that accumulate in tumor cells are promising tumor specific targets. Here, we demonstrate that, when using the inducible pSIP expression system, L. plantarum can produce and surface display two neoantigens, NAG1 and ETV6. The antigens could be targeted to both the cell membrane and the cell wall, utilizing four different anchoring methods. The production level and the degree of surface exposure of the antigens varied among the anchors. Flow cytometry analysis showed that antigens anchored to the cell wall were more exposed than those anchored to the cell membrane. To our knowledge these are the first reports of neoantigens being produced and surface-displayed in Lactobacillales.
Collapse
Affiliation(s)
- Kamilla Wiull
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway.
| | - Erik Holmgren
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
| | - Hans Grönlund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- NEOGAP Therapeutics AB, Stockholm, Sweden
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway.
| |
Collapse
|
13
|
Martin Fuentes A. The role of the microbiome in skin cancer development and treatment. Curr Opin Oncol 2025; 37:121-125. [PMID: 39869016 DOI: 10.1097/cco.0000000000001120] [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: 01/28/2025]
Abstract
PURPOSE OF REVIEW Recent research underscores the significant influence of the skin and gut microbiota on melanoma and nonmelanoma skin cancer (NMSC) development and treatment outcomes. This review aims to synthesize current findings on how microbiota modulates immune responses, particularly enhancing the efficacy of immunotherapies such as immune checkpoint inhibitors (ICIs). RECENT FINDINGS The microbiota's impact on skin cancer is multifaceted, involving immune modulation, inflammation, and metabolic interactions. Beneficial strains like Bifidobacterium and Lactobacillus have shown potential in supporting anti-PD-1 and anti-CTLA-4 therapies by promoting T-cell activation and immune surveillance. Evidence from preclinical and clinical studies, including fecal microbiota transplantation (FMT), highlights improved response rates in patients with microbiota-rich profiles. Notably, certain bacterial metabolites, such as inosine, contribute to enhanced antitumor activity by stimulating IFN-γ in CD8 + T cells. SUMMARY Understanding the interplay between microbiota and skin cancer treatment opens promising avenues for adjunctive therapies. Probiotic and prebiotic interventions, FMT, and microbiota modulation are emerging as complementary strategies to improve immunotherapy outcomes and address treatment resistance in melanoma and NMSC.
Collapse
|
14
|
Yang M, Zhong P, Wei P. Living Bacteria: A New Vehicle for Vaccine Delivery in Cancer Immunotherapy. Int J Mol Sci 2025; 26:2056. [PMID: 40076679 PMCID: PMC11900161 DOI: 10.3390/ijms26052056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer vaccines, aimed at evolving the human immune system to eliminate tumor cells, have long been explored as a method of cancer treatment with significant clinical potential. Traditional delivery systems face significant challenges in directly targeting tumor cells and delivering adequate amounts of antigen due to the hostile tumor microenvironment. Emerging evidence suggests that certain bacteria naturally home in on tumors and modulate antitumor immunity, making bacterial vectors a promising vehicle for precision cancer vaccines. Live bacterial vehicles offer several advantages, including tumor colonization, precise drug delivery, and immune stimulation, making them a compelling option for cancer immunotherapy. In this review, we explore the mechanisms of action behind living bacteria-based vaccines, recent progress in popular bacterial chassis, and strategies for specific payload delivery and biocontainment to ensure safety. These approaches will lay the foundation for developing an affordable, widely applicable cancer vaccine delivery system. This review also discusses the challenges and future opportunities in harnessing bacterial-based vaccines for enhanced therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
| | | | - Pengcheng Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (M.Y.); (P.Z.)
| |
Collapse
|
15
|
Nevot G, Santos-Moreno J, Campamà-Sanz N, Toloza L, Parra-Cid C, Jansen PAM, Barbier I, Ledesma-Amaro R, van den Bogaard EH, Güell M. Synthetically programmed antioxidant delivery by a domesticated skin commensal. Cell Syst 2025; 16:101169. [PMID: 39919749 DOI: 10.1016/j.cels.2025.101169] [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: 04/28/2024] [Revised: 09/03/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025]
Abstract
Bacteria represent a promising dynamic delivery system for the treatment of disease. In the skin, the relevant location of Cutibacterium acnes within the hair follicle makes this bacterium an attractive chassis for dermal biotechnological applications. Here, we provide a genetic toolbox for the engineering of this traditionally intractable bacterium, including basic gene expression tools, biocontainment strategies, markerless genetic engineering, and dynamic transcriptional regulation. As a proof of concept, we develop an antioxidant-secreting strain capable of reducing oxidative stress in a UV stress model.
Collapse
Affiliation(s)
- Guillermo Nevot
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Cataluña, Spain
| | - Javier Santos-Moreno
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Cataluña, Spain.
| | - Nil Campamà-Sanz
- Department of Cell and Molecular Biology, Karolinska Institutet, Södermanland and Uppland, 17165 Stockholm, Sweden
| | - Lorena Toloza
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Cataluña, Spain
| | - Cristóbal Parra-Cid
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Cataluña, Spain
| | - Patrick A M Jansen
- Department of Dermatology, Radboud University Medical Center, Nijmegen 6525GA, Güeldres, the Netherlands
| | - Içvara Barbier
- Department of Bioengineering and Centre for Synthetic Biology, Imperial College London, Greater London, London SW72AZ, UK
| | - Rodrigo Ledesma-Amaro
- Department of Bioengineering and Centre for Synthetic Biology, Imperial College London, Greater London, London SW72AZ, UK
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud University Medical Center, Nijmegen 6525GA, Güeldres, the Netherlands
| | - Marc Güell
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Cataluña, Spain; ICREA, Institució Catalana de Recerca i Estudis Avançats, 08003 Barcelona, Cataluña, Spain.
| |
Collapse
|
16
|
Chen YE. A genetic toolbox for engineering C. acnes. Cell Syst 2025; 16:101199. [PMID: 39978310 DOI: 10.1016/j.cels.2025.101199] [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: 01/24/2025] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/22/2025]
Abstract
Cutibacterium acnes is a highly prevalent and abundant skin bacterium that lives deep in the hair follicle, a unique site for host access. Thus, it is a prime target to engineer. This study introduces a genetic toolbox for C. acnes, which will enable basic science and therapeutic bioengineering.
Collapse
Affiliation(s)
- Y Erin Chen
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
17
|
Xia W, Feng Z, Wang Y, Lei R, Zhou Y, Zhuo Y, Xie R, Dong H, Zhao X, Guan X, Wu J. Orthogonally Engineered Bacteria Capture Metabolically Labeled Tumor Antigens to Improve the Systemic Immune Response in Irradiated Tumors. ACS NANO 2025; 19:5376-5391. [PMID: 39889238 DOI: 10.1021/acsnano.4c13320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Abstract
In situ vaccination is considered a promising cancer immunotherapy strategy to elicit a tumor-specific T cell response. Live bacteria effectively enhanced the immune response in irradiated tumors as it can activate multiple immune cells. However, the adaptive immune response remains low since bacteria lack the efficient delivery of antigen to dendritic cells (DCs). Here, we show that tumor antigens can be metabolically labeled with azido groups in situ, allowing for their specific capture by orthogonally engineered Salmonella via bioorthogonal chemistry. Subsequently, these antigens are efficiently delivered to DCs through the active movement of the bacteria. Intratumorally injected engineered bacteria captured the labeled antigens and improved their presentation by DCs. This increased the proportion of antigen-specific CD8+ T cells in tumors, further resulting in systemic antitumor effects in the bilateral melanoma mouse model. The antitumor effects were abrogated in Batf3-/- mice or after CD8+ T cell depletion, indicating that systemic antitumor effects were dependent on adaptive immune responses. Overall, our work presents a strategy combining bacterial engineering and antigen labeling, which may guide the development of in situ vaccines in the future.
Collapse
Affiliation(s)
- Wen Xia
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing 210093, China
- Department of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing 210008, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
- Institute of Drug Research and Development & Jiangsu Engineering Center of Biointelligent Materials, Nanjing University, Nanjing 210093, China
- Wuxi Xishan NJU Institute of Applied Biotechnology, Anzhen Street, Xishan District, Wuxi 214101, China
| | - Zhuo Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing 210093, China
| | - Yuchen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing 210093, China
| | - Ruiqi Lei
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing 210093, China
| | - Yao Zhou
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Yujia Zhuo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing 210093, China
| | - Ran Xie
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing 210093, China
| | - Xiaozhi Zhao
- Department of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing 210008, China
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing 210093, China
- Department of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing 210008, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
- Institute of Drug Research and Development & Jiangsu Engineering Center of Biointelligent Materials, Nanjing University, Nanjing 210093, China
- Wuxi Xishan NJU Institute of Applied Biotechnology, Anzhen Street, Xishan District, Wuxi 214101, China
| |
Collapse
|
18
|
Scharschmidt TC, Segre JA. Skin microbiome and dermatologic disorders. J Clin Invest 2025; 135:e184315. [PMID: 39895627 PMCID: PMC11785926 DOI: 10.1172/jci184315] [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: 02/04/2025] Open
Abstract
Human skin acts as a physical barrier to prevent the entry of pathogenic microbes while simultaneously providing a home for commensal bacteria and fungi. Microbiome sequencing studies have demonstrated the unappreciated diversity and selectivity of these microbes. Functional studies have demonstrated the impact of specific strains to tune the immune system, sculpt the microbial community, provide colonization resistance, and promote epidermal barrier integrity. Recent studies have integrated the microbiome, immunity, and tissue integrity to understand their interplay in common disorders such as atopic dermatitis. In this Review, we explore microbiome shifts associated with cutaneous disorders with an eye toward how the microbiome can be mined to identify new therapeutic opportunities.
Collapse
Affiliation(s)
- Tiffany C. Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, California, USA
| | - Julia A. Segre
- Microbial Genomics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Chemla Y, Sweeney CJ, Wozniak CA, Voigt CA. Design and regulation of engineered bacteria for environmental release. Nat Microbiol 2025; 10:281-300. [PMID: 39905169 DOI: 10.1038/s41564-024-01918-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/04/2024] [Indexed: 02/06/2025]
Abstract
Emerging products of biotechnology involve the release of living genetically modified microbes (GMMs) into the environment. However, regulatory challenges limit their use. So far, GMMs have mainly been tested in agriculture and environmental cleanup, with few approved for commercial purposes. Current government regulations do not sufficiently address modern genetic engineering and limit the potential of new applications, including living therapeutics, engineered living materials, self-healing infrastructure, anticorrosion coatings and consumer products. Here, based on 47 global studies on soil-released GMMs and laboratory microcosm experiments, we discuss the environmental behaviour of released bacteria and offer engineering strategies to help improve performance, control persistence and reduce risk. Furthermore, advanced technologies that improve GMM function and control, but lead to increases in regulatory scrutiny, are reviewed. Finally, we propose a new regulatory framework informed by recent data to maximize the benefits of GMMs and address risks.
Collapse
Affiliation(s)
- Yonatan Chemla
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Connor J Sweeney
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Christopher A Voigt
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
20
|
Bousbaine D, Bauman KD, Chen YE, Lalgudi PV, Nguyen TTD, Swenson JM, Yu VK, Tsang E, Conlan S, Li DB, Jbara A, Zhao A, Naziripour A, Veinbachs A, Lee YE, Phung JL, Dimas A, Jain S, Meng X, Pham TPT, McLaughlin MI, Barkal LJ, Gribonika I, Van Rompay KKA, Kong HH, Segre JA, Belkaid Y, Barnes CO, Fischbach MA. Discovery and engineering of the antibody response to a prominent skin commensal. Nature 2025; 638:1054-1064. [PMID: 39662508 PMCID: PMC12045117 DOI: 10.1038/s41586-024-08489-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
The ubiquitous skin colonist Staphylococcus epidermidis elicits a CD8+ T cell response pre-emptively, in the absence of an infection1. However, the scope and purpose of this anticommensal immune programme are not well defined, limiting our ability to harness it therapeutically. Here, we show that this colonist also induces a potent, durable and specific antibody response that is conserved in humans and non-human primates. A series of S. epidermidis cell-wall mutants revealed that the cell surface protein Aap is a predominant target. By colonizing mice with a strain of S. epidermidis in which the parallel β-helix domain of Aap is replaced by tetanus toxin fragment C, we elicit a potent neutralizing antibody response that protects mice against a lethal challenge. A similar strain of S. epidermidis expressing an Aap-SpyCatcher chimera can be conjugated with recombinant immunogens; the resulting labelled commensal elicits high antibody titres under conditions of physiologic colonization, including a robust IgA response in the nasal and pulmonary mucosa. Thus, immunity to a common skin colonist involves a coordinated T and B cell response, the latter of which can be redirected against pathogens as a new form of topical vaccination.
Collapse
Affiliation(s)
- Djenet Bousbaine
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Katherine D Bauman
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Y Erin Chen
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Pranav V Lalgudi
- ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tam T D Nguyen
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Joyce M Swenson
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Victor K Yu
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Eunice Tsang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Sean Conlan
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - David B Li
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Amina Jbara
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Aishan Zhao
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Arash Naziripour
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Alessandra Veinbachs
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Yu E Lee
- ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jennie L Phung
- ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alex Dimas
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Sunit Jain
- Chan Zuckerberg Biohub, Stanford, CA, USA
| | - Xiandong Meng
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Thi Phuong Thao Pham
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Martin I McLaughlin
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Layla J Barkal
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Inta Gribonika
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, CA, USA
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, CA, USA
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julia A Segre
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Christopher O Barnes
- ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, Stanford, CA, USA
| | - Michael A Fischbach
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- ChEM-H Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, Stanford, CA, USA.
| |
Collapse
|
21
|
Zhu B, Bai Y, Yeo YY, Lu X, Rovira-Clavé X, Chen H, Yeung J, Nkosi D, Glickman J, Delgado-Gonzalez A, Gerber GK, Angelo M, Shalek AK, Nolan GP, Jiang S. A multi-omics spatial framework for host-microbiome dissection within the intestinal tissue microenvironment. Nat Commun 2025; 16:1230. [PMID: 39890778 PMCID: PMC11785740 DOI: 10.1038/s41467-025-56237-7] [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: 04/26/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025] Open
Abstract
The intricate interactions between the host immune system and its microbiome constituents undergo dynamic shifts in response to perturbations to the intestinal tissue environment. Our ability to study these events on the systems level is significantly limited by in situ approaches capable of generating simultaneous insights from both host and microbial communities. Here, we introduce Microbiome Cartography (MicroCart), a framework for simultaneous in situ probing of host and microbiome across multiple spatial modalities. We demonstrate MicroCart by investigating gut host and microbiome changes in a murine colitis model, using spatial proteomics, transcriptomics, and glycomics. Our findings reveal a global but systematic transformation in tissue immune responses, encompassing tissue-level remodeling in response to host immune and epithelial cell state perturbations, bacterial population shifts, localized inflammatory responses, and metabolic process alterations during colitis. MicroCart enables a deep investigation of the intricate interplay between the host tissue and its microbiome with spatial multi-omics.
Collapse
Affiliation(s)
- Bokai Zhu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Yunhao Bai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Yao Yu Yeo
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xiaowei Lu
- Mass Spectrometry Core Facility, Stanford University, Stanford, CA, USA
| | - Xavier Rovira-Clavé
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Han Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Biological and Medical Informatics Program, UCSF, San Francisco, CA, USA
| | - Jason Yeung
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Dingani Nkosi
- Department of Pathology, Massachusetts General Brigham, Boston, MA, USA
| | - Jonathan Glickman
- Department of Pathology, Massachusetts General Brigham, Boston, MA, USA
| | | | - Georg K Gerber
- Division of Computational Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Health Sciences and Technology, Harvard University and MIT, Cambridge, MA, USA
| | - Mike Angelo
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA.
| | - Sizun Jiang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Division of Computational Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Yang Y, Wang J, Zhong Y, Tian M, Zhang H. Advances in Radionuclide-Labeled Biological Carriers for Tumor Imaging and Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4316-4336. [PMID: 39792777 DOI: 10.1021/acsami.4c19059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Biological carriers have emerged as significant tools to deliver radionuclides in nuclear medicine, providing a meaningful perspective for tumor imaging and treatment. Various radionuclide-labeled biological carriers have been developed to meet the needs of biomedical applications. This review introduces the principles of radionuclide-mediated imaging and therapy and the selected criteria of them, as well as a comprehensive description of the characteristics and functions of representative biological carriers including bacteria, cells, viruses, and their biological derivatives, emphasizing the labeled strategies of biological carriers combined with radionuclides. Subsequently, we in-depth introduce the application of radionuclide-labeled biological carriers in tumor imaging and treatment, including the imaging of the behaviors of biological carriers in vivo and tumor metastasis and the tumor treatment by radionuclide therapy, plus other strategies and radiation-induced photodynamic therapy. Finally, the challenges and prospects of radionuclide-labeled biological carriers are discussed to improve the shortcomings of this innovative platform and promote clinical transformation in the field of medical imaging.
Collapse
Affiliation(s)
- Yaozhang Yang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang 310014, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
23
|
Liu W, Chen W, Cai Y, Tang Y, Chen T. Advances in engineered bacteria vaccines for enhancing anti-cancer immunity. MICROBIOME RESEARCH REPORTS 2025; 4:14. [PMID: 40207284 PMCID: PMC11977376 DOI: 10.20517/mrr.2024.75] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/25/2024] [Accepted: 01/09/2025] [Indexed: 04/11/2025]
Abstract
Advances in synthetic biology have enabled the development of tumor-targeted live bacterial therapeutics. In a recent study published in Nature, Redenti et al. engineered Escherichia coli Nissle 1917(EcNcΔlon/ΔompT/LLO+ nAg), which exploits the advantages of living medicines to deliver arrays of tumor-specific neoantigenic epitope in optimal environments, thereby providing a novel strategy for developing effective and durable cancer immunotherapies.
Collapse
Affiliation(s)
- Wu Liu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China
| | - Wenjing Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China
| | - Yujie Cai
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China
| | - Yanhua Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China
- Authors contributed equally
| | - Tingtao Chen
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China
- Jiangxi Province Key Laboratory of Bioengineering Drugs, The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- Authors contributed equally
| |
Collapse
|
24
|
Manole S, Nguyen DH, Min JJ, Zhou S, Forbes N. Setting "cold" tumors on fire: Cancer therapy with live tumor-targeting bacteria. MED 2025; 6:100549. [PMID: 39689707 DOI: 10.1016/j.medj.2024.11.002] [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: 12/05/2023] [Revised: 09/18/2024] [Accepted: 11/01/2024] [Indexed: 12/19/2024]
Abstract
Immunotherapy with checkpoint blockade has shown remarkable efficacy in many patients with a variety of different types of cancer. However, the majority of patients with cancer have yet to benefit from this revolutionary therapy. Studies have shown that checkpoint blockade works best against immune-inflamed tumors characterized by the presence of tumor-infiltrating lymphocytes (TILs). In this review, we summarize studies using live tumor-targeting bacteria to treat cancer and describe various strategies to engineer the tumor-targeting bacteria for maximized immunoregulatory effects. We propose that tumor-localized infections by such engineered bacteria can create an immune microenvironment in favor of a more effective antitumor immunity with or without other therapies, such as immune checkpoint blockade (ICB). Finally, we will briefly outline some exemplary oncology clinical trials involving ICB plus live therapeutic bacteria, with a focus on their ability to modulate antitumor immune responses.
Collapse
Affiliation(s)
- Simin Manole
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Dinh-Huy Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea; Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeonnam 58128, South Korea.
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Neil Forbes
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA; Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA; Department of Microbiology, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
25
|
Singer ZS, Pabón J, Huang H, Sun W, Luo H, Grant KR, Obi I, Coker C, Rice CM, Danino T. Engineered bacteria launch and control an oncolytic virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.09.28.559873. [PMID: 37808855 PMCID: PMC10557668 DOI: 10.1101/2023.09.28.559873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
The ability of bacteria and viruses to selectively replicate in tumors has led to synthetic engineering of new microbial therapies. Here we design a cooperative strategy whereby S. typhimurium bacteria transcribe and deliver the Senecavirus A RNA genome inside host cells, launching a potent oncolytic viral infection. "Encapsidated" by bacteria, the viral genome can further bypass circulating antiviral antibodies to reach the tumor and initiate replication and spread within immune mice. Finally, we engineer the virus to require a bacterially delivered protease to achieve virion maturation, demonstrating bacterial control over the virus. This work extends bacterially delivered therapeutics to viral genomes, and shows how a consortium of microbes can achieve a cooperative aim.
Collapse
|
26
|
Zhang H, Chen L, Chen Q, Chen Q, Zhou J. Genetically Engineered Bacteria as A Living Bioreactor for Monitoring and Elevating Hypoxia-Activated Prodrug Tumor Therapy. Adv Healthc Mater 2025; 14:e2402272. [PMID: 39543798 DOI: 10.1002/adhm.202402272] [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: 06/21/2024] [Revised: 10/11/2024] [Indexed: 11/17/2024]
Abstract
Tirapazamine (TPZ), an antitumor prodrug, can be activated in hypoxic environment. It specifically targets the hypoxic microenvironment of tumors and produces toxic free radicals. However, due to the tumor is not completely hypoxic, TPZ often fails to effectively treat the entire tumor tissue, resulting in suboptimal therapeutic outcomes. Herein, a low pathogenic Escherichia coli TOP10 is utilized to selectively colonize tumor tissues, disrupt blood vessels, and induce thrombus formation, leading to the expansion of hypoxic region and improving the therapeutic effect of TPZ. Additionally, a thermosensitive hydrogel is constructed by Pluronic F-127 (F127), which undergoes gelation in situ at the tumor site, resulting in sustained release of TPZ. To monitor the therapeutic process, it is genetically modified TOP10 by integrating the bioluminescent system luxCDABE (TOP10-Lux). The bioluminescent signal is associated with tumor hypoxia enhancement and thrombus formation, which is beneficial for therapeutic monitoring with bioluminescence imaging. In the murine colon cancer model, the TOP10-Lux combined with TPZ-loaded F127 hydrogel effectively suppressed tumor growth, and the treatment process is efficiently monitored. Together, this work employs genetically modified TOP10-Lux to enhance the therapeutic efficacy of TPZ and monitor the treatment process, providing an effective strategy for bacteria-based tumor-targeted chemotherapy and treatment monitoring.
Collapse
Affiliation(s)
- Hongwei Zhang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Linfu Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Qiufang Chen
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Jun Zhou
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| |
Collapse
|
27
|
Silva CAC, Fidelle M, Almonte AA, Derosa L, Zitvogel L. Gut Microbiota-Related Biomarkers in Immuno-Oncology. Annu Rev Pharmacol Toxicol 2025; 65:333-354. [PMID: 39259979 DOI: 10.1146/annurev-pharmtox-061124-102218] [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] [Indexed: 09/13/2024]
Abstract
Carcinogenesis is associated with the emergence of protracted intestinal dysbiosis and metabolic changes. Increasing evidence shows that gut microbiota-related biomarkers and microbiota-centered interventions are promising strategies to overcome resistance to immunotherapy. However, current standard methods for evaluating gut microbiota composition are cost- and time-consuming. The development of routine diagnostic tools for intestinal barrier alterations and dysbiosis constitutes a critical unmet medical need that can guide routine treatment and microbiota-centered intervention decisions in patients with cancer. In this review, we explore the influence of gut microbiota on cancer immunotherapy and highlight gut-associated biomarkers that have the potential to be transformed into simple diagnostic tools, thus guiding standard treatment decisions in the field of immuno-oncology. Mechanistic insights toward leveraging the complex relationship between cancer immunosurveillance, gut microbiota, and metabolism open exciting opportunities for developing novel biomarkers in immuno-oncology.
Collapse
Affiliation(s)
- Carolina Alves Costa Silva
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Marine Fidelle
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Andrew A Almonte
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Lisa Derosa
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Laurence Zitvogel
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| |
Collapse
|
28
|
Cao Y, Yan W, Yi W, Yin Q, Li Y. Bioengineered therapeutic systems for improving antitumor immunity. Natl Sci Rev 2025; 12:nwae404. [PMID: 40114728 PMCID: PMC11925021 DOI: 10.1093/nsr/nwae404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/29/2024] [Accepted: 11/08/2024] [Indexed: 03/22/2025] Open
Abstract
Immunotherapy, a monumental advancement in antitumor therapy, still yields limited clinical benefits owing to its unguaranteed efficacy and safety. Therapeutic systems derived from cellular, bacterial and viral sources possess inherent properties that are conducive to antitumor immunotherapy. However, crude biomimetic systems have restricted functionality and may produce undesired toxicity. With advances in biotechnology, various toolkits are available to add or subtract certain properties of living organisms to create flexible therapeutic platforms. This review elaborates on the creation of bioengineered systems, via gene editing, synthetic biology and surface engineering, to enhance immunotherapy. The modifying strategies of the systems are discussed, including equipment for navigation and recognition systems to improve therapeutic precision, the introduction of controllable components to control the duration and intensity of treatment, the addition of immunomodulatory components to amplify immune activation, and the removal of toxicity factors to ensure biosafety. Finally, we summarize the advantages of bioengineered immunotherapeutic systems and possible directions for their clinical translation.
Collapse
Affiliation(s)
- Ying Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenlu Yan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenzhe Yi
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
29
|
Jin K, Huang Y, Che H, Wu Y. Engineered Bacteria for Disease Diagnosis and Treatment Using Synthetic Biology. Microb Biotechnol 2025; 18:e70080. [PMID: 39801378 PMCID: PMC11725985 DOI: 10.1111/1751-7915.70080] [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: 07/04/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025] Open
Abstract
Using synthetic biology techniques, bacteria have been engineered to serve as microrobots for diagnosing diseases and delivering treatments. These engineered bacteria can be used individually or in combination as microbial consortia. The components within these consortia complement each other, enhancing diagnostic accuracy and providing synergistic effects that improve treatment efficacy. The application of microbial therapies in cancer, intestinal diseases, and metabolic disorders underscores their significant potential. The impact of these therapies on the host's native microbiota is crucial, as engineered microbes can modulate and interact with the host's microbial environment, influencing treatment outcomes and overall health. Despite numerous advancements, challenges remain. These include ensuring the long-term survival and safety of bacteria, developing new chassis microbes and gene editing techniques for non-model strains, minimising potential toxicity, and understanding bacterial interactions with the host microbiota. This mini-review examines the current state of engineered bacteria and microbial consortia in disease diagnosis and treatment, highlighting advancements, challenges, and future directions in this promising field.
Collapse
Affiliation(s)
- Kai Jin
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Yi Huang
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Hailong Che
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Yihan Wu
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| |
Collapse
|
30
|
Wang H, Xu F, Yao C, Dai H, Xu J, Wu B, Tian B, Shi X, Wang C. Engineering bacteria for cancer immunotherapy by inhibiting IDO activity and reprogramming CD8+ T cell response. Proc Natl Acad Sci U S A 2024; 121:e2412070121. [PMID: 39693352 DOI: 10.1073/pnas.2412070121] [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: 06/16/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Inhibiting indoleamine 2,3 dioxygenase (IDO) for anticancer therapy has garnered significant attention in recent years. However, current IDO inhibitors face significant challenges which limit their clinical application. Here, we genetically engineered a high tryptophan-expressing Clostridium butyricum (L-Trp CB) strain that can colonize tumors strictly following systemic administration. We revealed that butyrate produced by L-Trp CB can inhibit IDO activity, preventing tryptophan catabolism and kynurenine accumulation in tumors. In addition, the large released tryptophan by L-Trp CB can provide discrete signals that support CD8+ T cell activation and energy metabolism within the tumor microenvironment. We observed that L-Trp CB significantly restored the proportion and function of CD8+ T cells, leading to significantly delayed tumor growth in both mouse and rabbit multiple tumor models with limited side effects. We here provide a synthetic biology treatment strategy for enhanced tumor immunotherapy by inhibiting IDO activity and reprogramming CD8+ T cell response in tumors.
Collapse
Affiliation(s)
- Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chenlu Yao
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Huaxing Dai
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jialu Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bingbing Wu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bo Tian
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Xiaolin Shi
- Medical College of Soochow University, Suzhou 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
31
|
Zhang Z, Wu W, Lin J, Li H. Unveiling the hidden causal links: skin flora and cutaneous melanoma. Front Oncol 2024; 14:1451175. [PMID: 39723372 PMCID: PMC11668787 DOI: 10.3389/fonc.2024.1451175] [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: 06/18/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Objective The presence of skin flora (SF) has been identified as a significant factor in the onset and progression of cutaneous melanoma (CM). However, the vast diversity and abundance of SF present challenges to fully understanding the causal relationship between SF and CM. Methods A Two Sample Mendelian Randomization (TSMR) analysis was conducted to investigating the causal relationship between SF and CM. The Inverse-Variance Weighted (IVW) method was utilized as the primary approach to assess the causal relationship under investigation. Furthermore, an independent external cohort was employed to validate the initial findings, followed by a meta-analysis of the consolidated results. To address potential confounding factors related to the influence of SF on CM, a Multivariate Mendelian Randomization (MVMR) analysis was also conducted. Finally, a Reverse Mendelian Randomization (RMR) was conducted to further validate the causal association. Results TSMR results showed that 9 SF have a causal relationship with CM in the training cohort. Although these 9 SF weren't confirmed in the testing cohort, 4 SF remained significant in the meta-analysis after integrating results from both cohorts. MVMR analysis indicated that 3 SF were still significantly associated with CM after accounting for the interactions between different SF in the training cohort. No reverse causal relationship was identified in RMR analysis. Conclusion A total of 9 SF were identified as having a potential causal relationship with CM; however, a large randomized controlled trial is needed to verify these results.
Collapse
Affiliation(s)
- Zexin Zhang
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenfeng Wu
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiajia Lin
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongyi Li
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
32
|
Shi Y, Zhao W, Ding Y, Ge X, Ju M. Research on the influence of radiotherapy-related genes on immune infiltration, immunotherapy response and prognosis in melanoma based on multi-omics. Front Immunol 2024; 15:1467098. [PMID: 39687627 PMCID: PMC11647020 DOI: 10.3389/fimmu.2024.1467098] [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: 07/19/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Background Skin cutaneous melanoma (SKCM) is a significant oncological challenge due to its aggressive nature and poor treatment outcomes. This study explores the comprehensive effects of radiotherapy (RT) in SKCM, focusing on cell signaling pathways, immune infiltration, immune gene correlations, immunotherapy response, and prognosis. Methods Using the Cancer Genome Atlas (TCGA) database, differentially expressed genes (DEGs) in SKCM patients undergoing RT were identified. A risk score model based on these DEGs was developed to assess the effects of RT-related genes on drug sensitivity, immune cell infiltration, immunotherapy response, and prognosis through multi-omics analysis. Human melanoma cells UACC62 and UACC257 were irradiated with 8 Gy gamma ray to establish an in vitro model, verifying the impact of radiotherapy on gene expression. Results The risk score demonstrated significant prognostic value and emerged as an independent prognostic factor. miRNA-mRNA and transcription factor regulatory networks underscored its clinical significance. Four key genes were identified: DUSP1, CXCL13, SLAMF7, and EVI2B. Analysis of single-cell and immunotherapy datasets indicated that these genes enhance immune response and immunotherapy efficacy in melanoma patients. PCR results confirmed that gamma rays increased the expression of these genes in human melanoma cells UACC62 and UACC257. Conclusion Using a multi-omics approach, we analyzed and validated the impact of RT on the immune landscape of melanoma patients. Our findings highlight the critical role of RT-related genes in predicting SKCM prognosis and guiding personalized therapy strategies, particularly in the context of immunotherapy. These contribute to understanding the role of radiotherapy combined with immunotherapy in melanoma.
Collapse
Affiliation(s)
- Yujing Shi
- Department of Oncology, Affiliated Jurong Hospital of Jiangsu University, Zhenjiang, ;China
| | - Wantong Zhao
- Department of Radiation Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, ;China
| | - Yuanjian Ding
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Osaka, ;Japan
| | - Xiaolin Ge
- Department of Radiation Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, ;China
| | - Mengyang Ju
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Osaka, ;Japan
| |
Collapse
|
33
|
Madaan T, Doan K, Hartman A, Gherardini D, Ventrola A, Zhang Y, Kotagiri N. Advances in Microbiome-Based Therapeutics for Dermatological Disorders: Current Insights and Future Directions. Exp Dermatol 2024; 33:e70019. [PMID: 39641544 PMCID: PMC11663288 DOI: 10.1111/exd.70019] [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: 06/04/2024] [Revised: 10/30/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
The human skin hosts an estimated 1000 bacterial species that are essential for maintaining skin health. Extensive clinical and preclinical studies have established the significant role of the skin microbiome in dermatological disorders such as atopic dermatitis, psoriasis, diabetic foot ulcers, hidradenitis suppurativa and skin cancers. In these conditions, the skin microbiome is not only altered but, in some cases, implicated in disease pathophysiology. Microbiome-based therapies (MBTs) represent an emerging category of live biotherapeutic products with tremendous potential as a novel intervention platform for skin diseases. Beyond using established wild-type strains native to the skin, these therapies can be enhanced to express targeted therapeutic molecules, offering more tailored treatment approaches. This review explores the role of the skin microbiome in various common skin disorders, with a particular focus on the development and therapeutic potential of MBTs for treating these conditions.
Collapse
Affiliation(s)
- Tushar Madaan
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Kyla Doan
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Alexandra Hartman
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Dominick Gherardini
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Alec Ventrola
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Yuhang Zhang
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Nalinikanth Kotagiri
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| |
Collapse
|
34
|
Yang X, Hang HC. Chemical genetic approaches to dissect microbiota mechanisms in health and disease. Science 2024; 386:eado8548. [PMID: 39541443 DOI: 10.1126/science.ado8548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Advances in genomics, proteomics, and metabolomics have revealed associations between specific microbiota species in health and disease. However, the precise mechanism(s) of action for many microbiota species and molecules have not been fully elucidated, limiting the development of microbiota-based diagnostics and therapeutics. In this Review, we highlight innovative chemical and genetic approaches that are enabling the dissection of microbiota mechanisms and providing causation in health and disease. Although specific microbiota molecules and mechanisms have begun to emerge, new approaches are still needed to go beyond phenotypic associations and translate microbiota discoveries into actionable targets and therapeutic leads to prevent and treat diseases.
Collapse
Affiliation(s)
- Xinglin Yang
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
- Department of Chemistry, Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
35
|
Macandog ADG, Catozzi C, Capone M, Nabinejad A, Nanaware PP, Liu S, Vinjamuri S, Stunnenberg JA, Galiè S, Jodice MG, Montani F, Armanini F, Cassano E, Madonna G, Mallardo D, Mazzi B, Pece S, Tagliamonte M, Vanella V, Barberis M, Ferrucci PF, Blank CU, Bouvier M, Andrews MC, Xu X, Santambrogio L, Segata N, Buonaguro L, Cocorocchio E, Ascierto PA, Manzo T, Nezi L. Longitudinal analysis of the gut microbiota during anti-PD-1 therapy reveals stable microbial features of response in melanoma patients. Cell Host Microbe 2024; 32:2004-2018.e9. [PMID: 39481388 PMCID: PMC11629153 DOI: 10.1016/j.chom.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024]
Abstract
Immune checkpoint inhibitors (ICIs) improve outcomes in advanced melanoma, but many patients are refractory or experience relapse. The gut microbiota modulates antitumor responses. However, inconsistent baseline predictors point to heterogeneity in responses and inadequacy of cross-sectional data. We followed patients with unresectable melanoma from baseline and during anti-PD-1 therapy, collecting fecal and blood samples that were surveyed for changes in the gut microbiota and immune markers. Varying patient responses were linked to different gut microbiota dynamics during ICI treatment. We select complete responders by their stable microbiota functions and validate them using multiple external cohorts and experimentally. We identify major histocompatibility complex class I (MHC class I)-restricted peptides derived from flagellin-related genes of Lachnospiraceae (FLach) as structural homologs of tumor-associated antigens, detect FLach-reactive CD8+ T cells in complete responders before ICI therapy, and demonstrate that FLach peptides improve antitumor immunity. These findings highlight the prognostic value of microbial functions and therapeutic potential of tumor-mimicking microbial peptides.
Collapse
Affiliation(s)
- Angeli D G Macandog
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Carlotta Catozzi
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Mariaelena Capone
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Amir Nabinejad
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Padma P Nanaware
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Smita Vinjamuri
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Johanna A Stunnenberg
- Netherlands Cancer Institute (NKI)-AVL, North Holland, Amsterdam 1066 CX, the Netherlands
| | - Serena Galiè
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Maria Giovanna Jodice
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Francesca Montani
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Federica Armanini
- Department of CIBIO, University of Trento, Trento, Povo 38123, Italy
| | - Ester Cassano
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Gabriele Madonna
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Domenico Mallardo
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | | | - Salvatore Pece
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Maria Tagliamonte
- Innovative Immunological Models, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Vito Vanella
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Massimo Barberis
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | | | - Christian U Blank
- Netherlands Cancer Institute (NKI)-AVL, North Holland, Amsterdam 1066 CX, the Netherlands
| | - Marlene Bouvier
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Miles C Andrews
- Department of Medicine, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nicola Segata
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy; Department of CIBIO, University of Trento, Trento, Povo 38123, Italy
| | - Luigi Buonaguro
- Innovative Immunological Models, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Emilia Cocorocchio
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Teresa Manzo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin 10126, Italy
| | - Luigi Nezi
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy.
| |
Collapse
|
36
|
Khan M, Dong Y, Ullah R, Li M, Huang Q, Hu Y, Yang L, Luo Z. Recent Advances in Bacterium-Based Therapeutic Modalities for Melanoma Treatment. Adv Healthc Mater 2024; 13:e2401076. [PMID: 39375965 DOI: 10.1002/adhm.202401076] [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: 03/21/2024] [Revised: 07/16/2024] [Indexed: 10/09/2024]
Abstract
Melanoma is one of the most severe skin cancer indications with rapid progression and a high risk of metastasis. However, despite the accumulated advances in melanoma treatment including adjuvant radiation, chemotherapy, and immunotherapy, the overall melanoma treatment efficacy in the clinics is still not satisfactory. Interestingly, bacterial therapeutics have demonstrated unique properties for tumor-related therapeutic applications, such as tumor-targeted motility, tailorable cytotoxicity, and immunomodulatory capacity of the tumor microenvironment, which have emerged as a promising platform for melanoma therapy. Indeed, the recent advances in genetic engineering and nanotechnologies have boosted the application potential of bacterium-based therapeutics for treating melanoma by further enhancing their tumor-homing, cell-killing, drug delivery, and immunostimulatory capacities. This review provides a comprehensive summary of the state-of-the-art bacterium-based anti-melanoma modalities, which are categorized according to their unique functional merits, including tumor-specific cytotoxins, tumor-targeted drug delivery platforms, and immune-stimulatory agents. Furthermore, a perspective is provided discussing the potential challenges and breakthroughs in this area. The insights in this review may facilitate the development of more advanced bacterium-based therapeutic modalities for improved melanoma treatment efficacy.
Collapse
Affiliation(s)
- Mubassir Khan
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yilong Dong
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325016, P. R. China
| | - Razi Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Lab for Vascular Implants College of Bioengineering Chongqing University, Chongqing, 400030, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Qiping Huang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| |
Collapse
|
37
|
Knödlseder N, Fábrega MJ, Santos-Moreno J, Manils J, Toloza L, Marín Vilar M, Fernández C, Broadbent K, Maruotti J, Lemenager H, Carolis C, Zouboulis CC, Soler C, Lood R, Brüggemann H, Güell M. Delivery of a sebum modulator by an engineered skin microbe in mice. Nat Biotechnol 2024; 42:1661-1666. [PMID: 38195987 DOI: 10.1038/s41587-023-02072-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 11/17/2023] [Indexed: 01/11/2024]
Abstract
Microorganisms can be equipped with synthetic genetic programs for the production of targeted therapeutic molecules. Cutibacterium acnes is the most abundant commensal of the human skin, making it an attractive chassis to create skin-delivered therapeutics. Here, we report the engineering of this bacterium to produce and secrete the therapeutic molecule neutrophil gelatinase-associated lipocalin, in vivo, for the modulation of cutaneous sebum production.
Collapse
Affiliation(s)
- Nastassia Knödlseder
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - María-José Fábrega
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Javier Santos-Moreno
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Joan Manils
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
- Serra Húnter Programme, Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Lorena Toloza
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Maria Marín Vilar
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Cristina Fernández
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Katrina Broadbent
- Protein Technologies Facility, Center of Genomic Regulation, Barcelona, Spain
| | | | | | - Carlo Carolis
- Protein Technologies Facility, Center of Genomic Regulation, Barcelona, Spain
| | - Christos C Zouboulis
- Hochschulklinik für Dermatologie, Venerologie und Allergologie, Immunologisches Zentrum; Städtisches Klinikum Dessau; and Medizinische Hochschule Brandenburg Theodor Fontane und Fakultät für Gesundheitswissenschaften Brandenburg, Dessau-Roßlau, Germany
| | - Concepció Soler
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Rolf Lood
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, Lund, Sweden
| | | | - Marc Güell
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
- ICREA, Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
| |
Collapse
|
38
|
Wang L, Ji P, Yu J, Qiu S, An B, Huo M, Shi J. Hybridized and engineered microbe for catalytic generation of peroxynitrite and cancer immunotherapy under sonopiezo initiation. SCIENCE ADVANCES 2024; 10:eadp7540. [PMID: 39475601 PMCID: PMC11524182 DOI: 10.1126/sciadv.adp7540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/24/2024] [Indexed: 11/02/2024]
Abstract
Living therapeutics is an emerging antitumor modality by living microorganisms capable of selective tropism and effective therapeutics. Nevertheless, primitive microbes could only present limited therapeutic functionalities against tumors. Hybridization of the microbes with multifunctional nanocatalysts is of great significance to achieve enhanced tumor catalytic therapy. In the present work, nitric oxide synthase (NOS)-engineered Escherichia coli strain MG1655 (NOBac) was used to hybridize with the sonopiezocatalytic BaTiO3 nanoparticles (BTO NPs) for efficient tumor-targeted accumulation and antitumor therapy. Under ultrasound irradiation, superoxide anions created by the piezocatalytic reaction of BTO NPs could immediately react with nitric oxide (NO) generated from NOBac to produce highly oxidative peroxynitrite ONOO- species in cascade, resulting in robust tumor piezocatalytic therapeutic efficacy, prompting prominent and sustained antitumoral immunoactivation simultaneously. The present work presents a promising cancer immunotherapy based on the engineered and hybridized microbes for highly selective and sonopiezo-controllable tumor catalytic therapy.
Collapse
Affiliation(s)
- Liping Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medicine Science, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Penghao Ji
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Jiadie Yu
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Shuwen Qiu
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Bolin An
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- CAS Key Laboratory of Quantitative Engineering Biology, Materials Synthetic Biology Center, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Minfeng Huo
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medicine Science, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
39
|
Redenti A, Im J, Redenti B, Li F, Rouanne M, Sheng Z, Sun W, Gurbatri CR, Huang S, Komaranchath M, Jang Y, Hahn J, Ballister ER, Vincent RL, Vardoshivilli A, Danino T, Arpaia N. Probiotic neoantigen delivery vectors for precision cancer immunotherapy. Nature 2024; 635:453-461. [PMID: 39415001 PMCID: PMC11560847 DOI: 10.1038/s41586-024-08033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 09/06/2024] [Indexed: 10/18/2024]
Abstract
Microbial systems have been synthetically engineered to deploy therapeutic payloads in vivo1,2. With emerging evidence that bacteria naturally home in on tumours3,4 and modulate antitumour immunity5,6, one promising application is the development of bacterial vectors as precision cancer vaccines2,7. Here we engineered probiotic Escherichia coli Nissle 1917 as an antitumour vaccination platform optimized for enhanced production and cytosolic delivery of neoepitope-containing peptide arrays, with increased susceptibility to blood clearance and phagocytosis. These features enhance both safety and immunogenicity, achieving a system that drives potent and specific T cell-mediated anticancer immunity that effectively controls or eliminates tumour growth and extends survival in advanced murine primary and metastatic solid tumours. We demonstrate that the elicited antitumour immune response involves recruitment and activation of dendritic cells, extensive priming and activation of neoantigen-specific CD4+ and CD8+ T cells, broader activation of both T and natural killer cells, and a reduction of tumour-infiltrating immunosuppressive myeloid and regulatory T and B cell populations. Taken together, this work leverages the advantages of living medicines to deliver arrays of tumour-specific neoantigen-derived epitopes within the optimal context to induce specific, effective and durable systemic antitumour immunity.
Collapse
Affiliation(s)
- Andrew Redenti
- Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jongwon Im
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Benjamin Redenti
- Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA
| | - Fangda Li
- Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA
| | - Mathieu Rouanne
- Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA
| | - Zeren Sheng
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - William Sun
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Candice R Gurbatri
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Shunyu Huang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Meghna Komaranchath
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - YoungUk Jang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jaeseung Hahn
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Edward R Ballister
- Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Rosa L Vincent
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Ana Vardoshivilli
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
- Data Science Institute, Columbia University, New York, NY, USA.
| | - Nicholas Arpaia
- Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
| |
Collapse
|
40
|
Qin S, He G, Yang J. Nanomaterial combined engineered bacteria for intelligent tumor immunotherapy. J Mater Chem B 2024; 12:9795-9820. [PMID: 39225508 DOI: 10.1039/d4tb00741g] [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: 09/04/2024]
Abstract
Cancer remains the leading cause of human death worldwide. Compared to traditional therapies, tumor immunotherapy has received a lot of attention and research focus due to its potential to activate both innate and adaptive immunity, low toxicity to normal tissue, and long-term immune activity. However, its clinical effectiveness and large-scale application are limited due to the immunosuppression microenvironment, lack of spatiotemporal control, expensive cost, and long manufacturing time. Recently, nanomaterial combined engineered bacteria have emerged as a promising solution to the challenges of tumor immunotherapy, which offers spatiotemporal control, reversal of immunosuppression, and scalable production. Therefore, we summarize the latest research on nanomaterial-assisted engineered bacteria for precise tumor immunotherapies, including the cross-talk of nanomaterials and bacteria as well as their application in different immunotherapies. In addition, we further discuss the advantages and challenges of nanomaterial-engineered bacteria and their future prospects, inspiring more novel and intelligent tumor immunotherapy.
Collapse
Affiliation(s)
- Shurong Qin
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Guanzhong He
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Jingjing Yang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
41
|
Belaid A, Roméo B, Rignol G, Benzaquen J, Audoin T, Vouret-Craviari V, Brest P, Varraso R, von Bergen M, Hugo Marquette C, Leroy S, Mograbi B, Hofman P. Impact of the Lung Microbiota on Development and Progression of Lung Cancer. Cancers (Basel) 2024; 16:3342. [PMID: 39409962 PMCID: PMC11605235 DOI: 10.3390/cancers16193342] [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: 08/30/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 12/01/2024] Open
Abstract
The past several years have provided a more profound understanding of the role of microbial species in the lung. The respiratory tract is a delicate ecosystem of bacteria, fungi, parasites, and viruses. Detecting microbial DNA, pathogen-associated molecular patterns (PAMPs), and metabolites in sputum is poised to revolutionize the early diagnosis of lung cancer. The longitudinal monitoring of the lung microbiome holds the potential to predict treatment response and side effects, enabling more personalized and effective treatment options. However, most studies into the lung microbiota have been observational and have not adequately considered the impact of dietary intake and air pollutants. This gap makes it challenging to establish a direct causal relationship between environmental exposure, changes in the composition of the microbiota, lung carcinogenesis, and tumor progression. A holistic understanding of the lung microbiota that considers both diet and air pollutants may pave the way to improved prevention and management strategies for lung cancer.
Collapse
Affiliation(s)
- Amine Belaid
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Barnabé Roméo
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Guylène Rignol
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Laboratory of Clinical and Experimental Pathology (LPCE), Biobank (BB-0033-00025), Centre Hospitalier Universitaire (CHU) de Nice, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| | - Jonathan Benzaquen
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Centre Hospitalier Universitaire (CHU) de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| | - Tanguy Audoin
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Valérie Vouret-Craviari
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Patrick Brest
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Raphaëlle Varraso
- Université Paris-Saclay, Équipe d’Épidémiologie Respiratoire Intégrative, CESP, INSERM, 94800 Villejuif, France;
| | - Martin von Bergen
- Helmholtz Centre for Environmental Research GmbH—UFZ, Department of Molecular Systems Biology, Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, 04109 Leipzig, Germany;
| | - Charles Hugo Marquette
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Centre Hospitalier Universitaire (CHU) de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| | - Sylvie Leroy
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Centre Hospitalier Universitaire (CHU) de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| | - Baharia Mograbi
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
| | - Paul Hofman
- Université Côte d’Azur, Institute for Research on Ageing and Cancer, Nice (IRCAN), Institut Hospitalo Universitaire (IHU) RespirERA, Fédérations Hospitalo-Universitaires (FHU) OncoAge, Centre Antoine Lacassagne, Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), 06107 Nice, France; (A.B.); (B.R.); (G.R.); (J.B.); (T.A.); (V.V.-C.); (P.B.); (C.H.M.); (S.L.); (P.H.)
- Laboratory of Clinical and Experimental Pathology (LPCE), Biobank (BB-0033-00025), Centre Hospitalier Universitaire (CHU) de Nice, FHU OncoAge, IHU RespirERA, 06000 Nice, France
| |
Collapse
|
42
|
Quinn G, Maggiore G, Li B. Genomic investigation of innate sensing pathways in the tumor microenvironment. BMC Cancer 2024; 24:1157. [PMID: 39289651 PMCID: PMC11409789 DOI: 10.1186/s12885-024-12944-w] [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: 01/25/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system is the first responder to infectious agents, cellular debris, and cancerous growths. This system plays critical roles in the antitumor immune responses by boosting and priming T cell-mediated cytotoxicity but is understudied due to the complexity and redundancy of its various downstream signaling cascades. We utilized a mathematical tool to holistically quantify innate immune signaling cascades and immunophenotype over 8,000 tumors from The Cancer Genome Atlas (TCGA). We found that innate immune activation was predictive of patient mortality in a subset of cancers. Further analysis identified PHF genes as transcripts that were associated with genomic stability and innate activation. Knockdown of PHF gene transcripts in vitro led to an increase in cell death and IFNB1 expression in a cGAS-dependent manner, validating PHF genes as potential anti-tumor targets. We also found an association between innate immune activation and both tumor immunogenicity and intratumor microbes, which highlights the versatility of this model. In conclusion, interrogating activation of innate immune signaling cascades demonstrated the importance of studying innate signaling in cancer and broadened the search for new therapeutic adjuvants.
Collapse
Affiliation(s)
- Gabriella Quinn
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Gianna Maggiore
- Children's Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bo Li
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA.
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
43
|
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
|
44
|
Renteria-Flores FI, García-Chagollán M, Jave-Suárez LF. Bactofection, Bacterial-Mediated Vaccination, and Cancer Therapy: Current Applications and Future Perspectives. Vaccines (Basel) 2024; 12:968. [PMID: 39340000 PMCID: PMC11435753 DOI: 10.3390/vaccines12090968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
From the first report in 1891 by Dr. Coley of the effective treatment of tumors in 1000 patients with Streptococcus and the first successful use of bacterial vectors for transferring therapeutic genes in 1980 by Dr. Schnaffer, bactofection has been shown to be a promising strategy in the fields of vaccination, gene therapy, and cancer therapy. This review describes the general theory of bactofection and its advantages, disadvantages, challenges, and expectations, compiling the most notable advances in 14 vaccination studies, 27 cancer therapy studies, and 13 clinical trials. It also describes the current scope of bactofection and promising results. The extensive knowledge of Salmonella biology, as well as the multiple adequacies of the Ty21a vaccination platform, has allowed notable developments worldwide that have mainly been reflected in therapeutic efforts against cancer. In this regard, we strongly recommend the creation of a recombinant Ty21a model that constitutively expresses the GtgE protease from S. typhimurium, allowing this vector to be used in animal trials, thus enhancing the likelihood of favorable results that could quickly transition to clinical trials. From the current perspective, it is necessary to explore a greater diversity of bacterial vectors and find the best combination of implemented attenuations, generating personalized models that guarantee the maximum effectiveness in cancer therapy and vaccination.
Collapse
Affiliation(s)
- Francisco Israel Renteria-Flores
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Mariel García-Chagollán
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Luis Felipe Jave-Suárez
- Division of Immunology, Biomedical Research Centre of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
45
|
Najar TA, Hao Y, Hao Y, Romero-Meza G, Dolynuk A, Littman DR. Microbiota-induced plastic T cells enhance immune control of antigen-sharing tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607605. [PMID: 39185202 PMCID: PMC11343098 DOI: 10.1101/2024.08.12.607605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Therapies that harness the immune system to target and eliminate tumor cells have revolutionized cancer care. Immune checkpoint blockade (ICB), which boosts the anti-tumor immune response by inhibiting negative regulators of T cell activation1-3, is remarkably successful in a subset of cancer patients, yet a significant proportion do not respond to treatment, emphasizing the need to understand factors influencing the therapeutic efficacy of ICB4-9. The gut microbiota, consisting of trillions of microorganisms residing in the gastrointestinal tract, has emerged as a critical determinant of immune function and response to cancer immunotherapy, with multiple studies demonstrating association of microbiota composition with clinical response10-16. However, a mechanistic understanding of how gut commensal bacteria influence the efficacy of ICB remains elusive. Here we utilized a gut commensal microorganism, segmented filamentous bacteria (SFB), which induces an antigen-specific Th17 cell effector program17, to investigate how colonization with it affects the efficacy of ICB in restraining distal growth of tumors sharing antigen with SFB. We find that anti-PD-1 treatment effectively inhibits the growth of implanted SFB antigen-expressing melanoma only if mice are colonized with SFB. Through T cell receptor clonal lineage tracing, fate mapping, and peptide-MHC tetramer staining, we identify tumor-associated SFB-specific Th1-like cells derived from the homeostatic Th17 cells induced by SFB colonization in the small intestine lamina propria. These gut-educated ex-Th17 cells produce high levels of the pro-inflammatory cytokines IFN-γ and TNF-α, and promote expansion and effector functions of CD8+ tumor-infiltrating cytotoxic lymphocytes, thereby controlling tumor growth. A better understanding of how distinct intestinal commensal microbes can promote T cell plasticity-dependent responses against antigen-sharing tumors may allow for the design of novel cancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Tariq A. Najar
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Yuan Hao
- Applied Bioinformatics Laboratories, Division of Advanced Research Technologies, New York, New York University School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- New York Genome Center, New York, NY 10013, USA
| | - Gabriela Romero-Meza
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Alexandra Dolynuk
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Dan R. Littman
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| |
Collapse
|
46
|
Gan Y, Zhang J, Qi F, Hu Z, Sweren E, Reddy SK, Chen L, Feng X, Grice EA, Garza LA, Wang G. Commensal microbe regulation of skin cells in disease. Cell Host Microbe 2024; 32:1264-1279. [PMID: 39146798 PMCID: PMC11457753 DOI: 10.1016/j.chom.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Human skin is the host to various commensal microbes that constitute a substantial microbial community. The reciprocal communication between these microbial inhabitants and host cells upholds both the morphological and functional attributes of the skin layers, contributing indispensably to microenvironmental and tissue homeostasis. Thus, disruption of the skin barrier or imbalances in the microbial communities can exert profound effects on the behavior of host cells. This influence, mediated by the microbes themselves or their metabolites, manifests in diverse outcomes. In this review, we examine existing knowledge to provide insight into the nuanced behavior exhibited by the microbiota on skin cells in health and disease states. These interactions provide insight into potential cellular targets for future microbiota-based therapies to prevent and treat skin disease.
Collapse
Affiliation(s)
- Yuyang Gan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Jiarui Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Fangfang Qi
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Evan Sweren
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sashank K Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Lu Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Xinyi Feng
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Elizabeth A Grice
- Department of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Luis A Garza
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| | - Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China; Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| |
Collapse
|
47
|
Chen M, Xia L, Wu C, Wang Z, Ding L, Xie Y, Feng W, Chen Y. Microbe-material hybrids for therapeutic applications. Chem Soc Rev 2024; 53:8306-8378. [PMID: 39005165 DOI: 10.1039/d3cs00655g] [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: 07/16/2024]
Abstract
As natural living substances, microorganisms have emerged as useful resources in medicine for creating microbe-material hybrids ranging from nano to macro dimensions. The engineering of microbe-involved nanomedicine capitalizes on the distinctive physiological attributes of microbes, particularly their intrinsic "living" properties such as hypoxia tendency and oxygen production capabilities. Exploiting these remarkable characteristics in combination with other functional materials or molecules enables synergistic enhancements that hold tremendous promise for improved drug delivery, site-specific therapy, and enhanced monitoring of treatment outcomes, presenting substantial opportunities for amplifying the efficacy of disease treatments. This comprehensive review outlines the microorganisms and microbial derivatives used in biomedicine and their specific advantages for therapeutic application. In addition, we delineate the fundamental strategies and mechanisms employed for constructing microbe-material hybrids. The diverse biomedical applications of the constructed microbe-material hybrids, encompassing bioimaging, anti-tumor, anti-bacteria, anti-inflammation and other diseases therapy are exhaustively illustrated. We also discuss the current challenges and prospects associated with the clinical translation of microbe-material hybrid platforms. Therefore, the unique versatility and potential exhibited by microbe-material hybrids position them as promising candidates for the development of next-generation nanomedicine and biomaterials with unique theranostic properties and functionalities.
Collapse
Affiliation(s)
- Meng Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai 200444, P. R. China.
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Li Ding
- Department of Medical Ultrasound, National Clinical Research Center of Interventional Medicine, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Tongji University, Shanghai, 200072, P. R. China.
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai 200444, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
- Shanghai Institute of Materdicine, Shanghai 200051, P. R. China
| |
Collapse
|
48
|
Xu X, Ding Y, Dong Y, Yuan H, Xia P, Qu C, Ma J, Wang H, Zhang X, Zhao L, Li Z, Liang Z, Wang J. Nanobody-Engineered Biohybrid Bacteria Targeting Gastrointestinal Cancers Induce Robust STING-Mediated Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401905. [PMID: 38888519 PMCID: PMC11336900 DOI: 10.1002/advs.202401905] [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: 02/22/2024] [Revised: 05/22/2024] [Indexed: 06/20/2024]
Abstract
Bacteria can be utilized for cancer therapy owing to their preferential colonization at tumor sites. However, unmodified non-pathogenic bacteria carry potential risks due to their non-specific targeting effects, and their anti-tumor activity is limited when used as monotherapy. In this study, a biohybrid-engineered bacterial system comprising non-pathogenic MG1655 bacteria modified with CDH17 nanobodies on their surface and conjugated with photosensitizer croconium (CR) molecules is developed. The resultant biohybrid bacteria can efficiently home to CDH17-positive tumors, including gastric, pancreatic, and colorectal cancers, and significantly suppress tumor growth upon irradiation. More importantly, biohybrid bacteria-mediated photothermal therapy (PTT) induced abundant macrophage infiltration in a syngeneic murine colorectal model. Further, that the STING pathway is activated in tumor macrophages by the released bacterial nucleic acid after PTT is revealed, leading to the production of type I interferons. The addition of CD47 nanobody but not PD-1 antibody to the PTT regimen can eradicate the tumors and extend survival. This results indicate that bacteria endowed with tumor-specific selectivity and coupled with photothermal payloads can serve as an innovative strategy for low-immunogenicity cancers. This strategy can potentially reprogram the tumor microenvironment by inducing macrophage infiltration and enhancing the efficacy of immunotherapy targeting macrophages.
Collapse
Affiliation(s)
- Xiaolong Xu
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Integrated Chinese and Western Medicine Postdoctoral Research StationJinan UniversityGuangzhou510632China
| | - Youbin Ding
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Department of Medical ImagingThe Third Affiliated HospitalSouthern Medical University (Academy of Orthopedics Guangdong Province)Guangzhou510515China
| | - Yafang Dong
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Department of Medical ImagingThe Third Affiliated HospitalSouthern Medical University (Academy of Orthopedics Guangdong Province)Guangzhou510515China
| | - Haitao Yuan
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Peng Xia
- Department of Hepatobiliary & Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanHubei430071China
| | - Chengming Qu
- Department of Hepatobiliary & Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanHubei430071China
| | - Jingbo Ma
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Huifang Wang
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Xiaodong Zhang
- Department of Medical ImagingThe Third Affiliated HospitalSouthern Medical University (Academy of Orthopedics Guangdong Province)Guangzhou510515China
| | - Liang Zhao
- Department of PathologyShunde Hospital, Southern Medical University (The First People's Hospital of Shunde)Foshan528308China
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Zhijie Li
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Zhen Liang
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Jigang Wang
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuan646000China
- Department of Traditional Chinese Medicine and School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, and Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700China
- State Key Laboratory of Antiviral DrugsSchool of PharmacyHenan UniversityKaifeng475004China
| |
Collapse
|
49
|
Kyriazi AA, Karaglani M, Agelaki S, Baritaki S. Intratumoral Microbiome: Foe or Friend in Reshaping the Tumor Microenvironment Landscape? Cells 2024; 13:1279. [PMID: 39120310 PMCID: PMC11312414 DOI: 10.3390/cells13151279] [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: 06/05/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
The role of the microbiome in cancer and its crosstalk with the tumor microenvironment (TME) has been extensively studied and characterized. An emerging field in the cancer microbiome research is the concept of the intratumoral microbiome, which refers to the microbiome residing within the tumor. This microbiome primarily originates from the local microbiome of the tumor-bearing tissue or from translocating microbiome from distant sites, such as the gut. Despite the increasing number of studies on intratumoral microbiome, it remains unclear whether it is a driver or a bystander of oncogenesis and tumor progression. This review aims to elucidate the intricate role of the intratumoral microbiome in tumor development by exploring its effects on reshaping the multileveled ecosystem in which tumors thrive, the TME. To dissect the complexity and the multitude of layers within the TME, we distinguish six specialized tumor microenvironments, namely, the immune, metabolic, hypoxic, acidic, mechanical and innervated microenvironments. Accordingly, we attempt to decipher the effects of the intratumoral microbiome on each specialized microenvironment and ultimately decode its tumor-promoting or tumor-suppressive impact. Additionally, we portray the intratumoral microbiome as an orchestrator in the tumor milieu, fine-tuning the responses in distinct, specialized microenvironments and remodeling the TME in a multileveled and multifaceted manner.
Collapse
Affiliation(s)
- Athina A. Kyriazi
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Makrina Karaglani
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Laboratory of Hygiene and Environmental Protection, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Sofia Agelaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Stavroula Baritaki
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| |
Collapse
|
50
|
Wang X, Zhu Z. A Mendelian randomization analysis reveals the multifaceted role of the skin microbiota in liver cancer. Front Microbiol 2024; 15:1422132. [PMID: 39113845 PMCID: PMC11303314 DOI: 10.3389/fmicb.2024.1422132] [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: 04/23/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC, or hepatic cancer, HC) and cholangiocarcinoma (CCA, or hepatic bile duct cancer, HBDC) are two major types of primary liver cancer (PLC). Previous studies have suggested that microbiota can either act as risk factors or preventive factors in PLC. However, no study has reported the relationship between skin microbiota and PLC. Therefore, we conducted a two-sample Mendelian randomization (MR) study to assess the causality between skin microbiota and PLC. Methods Data from the genome-wide association study (GWAS) on skin microbiota were collected. The GWAS summary data of GCST90018803 (HBDC) and GCST90018858 (HC) were utilized in the discovery and verification phases, respectively. The inverse variance weighted (IVW) method was utilized as the principal method in our MR study. The MR-Egger intercept test, Cochran's Q-test, MR-Pleiotropy RESidual Sum and Outlier (MR-PRESSO), and leave-one-out analysis were conducted to identify the heterogeneity and pleiotropy. Results The results showed that Veillonella (unc.) plays a protective role in HBDC, while the family Neisseriaceae has a positive association with HBDC risk. The class Betaproteobacteria, Veillonella (unc.), and the phylum Bacillota (Firmicutes) play a protective role in HC. Staphylococcus epidermidis, Corynebacterium (unc.), the family Neisseriaceae, and Pasteurellaceae sp. were associated with an increased risk of HC. Conclusion This study provided new evidence regarding the association between skin microbiota and PLC, suggesting that skin microbiota plays a role in PLC progression. Skin microbiota could be a novel and effective way for PLC diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaoxue Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zexin Zhu
- Department of Surgical Oncology, The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|