1
|
Wang H, Xu F, Wang C. Metabolic reprogramming of tumor microenviroment by engineered bacteria. Semin Cancer Biol 2025; 112:58-70. [PMID: 40157514 DOI: 10.1016/j.semcancer.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The tumor microenvironment (TME) is a complex ecosystem that plays a crucial role in tumor progression and response to therapy. The metabolic characteristics of the TME are fundamental to its function, influencing not only cancer cell proliferation and survival but also the behavior of immune cells within the tumor. Metabolic reprogramming-where cancer cells adapt their metabolic pathways to support rapid growth and immune evasion-has emerged as a key factor in cancer immunotherapy. Recently, the potential of engineered bacteria in cancer immunotherapy has gained increasing recognition, offering a novel strategy to modulate TME metabolism and enhance antitumor immunity. This review summarizes the metabolic properties and adaptations of tumor and immune cells within the TME and summarizes the strategies by which engineered bacteria regulate tumor metabolism. We discuss how engineered bacteria can overcome the immunosuppressive TME by reprogramming its metabolism to improve antitumor therapy. Furthermore, we examine the advantages, potential challenges, and future clinical translation of engineered bacteria in reshaping TME metabolism.
Collapse
Affiliation(s)
- Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
2
|
Ma C, Xu A, Zuo L, Li Q, Fan F, Hu Y, Sun C. Methionine Dependency and Restriction in Cancer: Exploring the Pathogenic Function and Therapeutic Potential. Pharmaceuticals (Basel) 2025; 18:640. [PMID: 40430461 PMCID: PMC12114517 DOI: 10.3390/ph18050640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Methionine, an essential amino acid, is obtained by dietary intake to fulfill the requirements of our bodies. Accumulating evidence indicates that methionine plays a pivotal role in various biological processes, including protein synthesis, energy metabolism, redox balance maintenance, and methylation modifications. Numerous advances underscore the heightened dependence of cancer cells on methionine, which is a significant factor in cancer pathogenesis and development. A profound comprehension of the intricate relationship between methionine metabolism and tumorigenesis is imperative for advancing the field of cancer therapeutics. Herein, we delve into the role of methionine in supporting cancer growth, the impact on epigenetic modifications, and the interaction between methionine and the tumor microenvironment. Additionally, we provide insights into the development of various methionine-targeted therapy strategies. This paper summarizes the current state of research and its translational potential, emphasizing the challenges and opportunities associated with harnessing methionine dependence as a target for innovative cancer treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chunyan Sun
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.M.); (A.X.); (L.Z.); (Q.L.); (F.F.); (Y.H.)
| |
Collapse
|
3
|
Liu B, Wang K, Li Q, Xiao Z, Chen Z, Zhang Y, Wu Y, Xu Y, Wu Y, Liu Z. Engineered VNP20009 expressing IL-15&15Rα augments anti-tumor immunity for bladder cancer treatment. Biomaterials 2025; 315:122951. [PMID: 39531748 DOI: 10.1016/j.biomaterials.2024.122951] [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: 05/12/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Surgical resection combined with intravesical instillation of chemotherapeutics or Bacillus Calmette-Guerin (BCG) to remove residual cancer cells is the gold standard for the clinical treatment of patients with bladder cancer. In a recent clinical trial, a new super-agonist complex of IL-15 - N803, has shown promising results when used in combination with BCG to treat patients with bladder cancer who do not respond to BCG. Herein, we used temperature-controlled pBV220 plasmid encoding Interleukin-15 and its receptor alpha subunit (IL-15&15Rα) to transform VNP20009, an attenuated salmonella typhimurium strain, obtaining engineered bacteria named 15&15Rα@VNP. After induction at 42 °C, 15&15Rα@VNP can secrete functional IL-15&15Rα stably. It was found that intravesical instillation of thermally activated 15&15Rα@VNP could inhibit the growth of bladder tumors if used alone. Moreover, the sequential intravesical instillation of epirubicin (EPI), a first-line bladder cancer drug, followed by thermally activated 15&15Rα@VNP, could achieve further improved therapeutic responses, without causing significant side effects. Therefore, this study shows that 15&15Rα@VNP can be effectively used in the treatment of bladder cancer and can be used as a complementary therapy to chemotherapy agents, promising for potential clinical translation in bladder cancer treatment.
Collapse
Affiliation(s)
- Bo Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kaiwei Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qiaofeng Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhisheng Xiao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zihao Chen
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215124, China
| | - Yuting Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yuzhe Wu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yuchun Xu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yumin Wu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China; Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, 999078 Macau SAR, China.
| |
Collapse
|
4
|
Parkhitko AA, Cracan V. Xenotopic synthetic biology: Prospective tools for delaying aging and age-related diseases. SCIENCE ADVANCES 2025; 11:eadu1710. [PMID: 40153513 DOI: 10.1126/sciadv.adu1710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/24/2025] [Indexed: 03/30/2025]
Abstract
Metabolic dysregulation represents one of the major driving forces in aging. Although multiple genetic and pharmacological manipulations are known to extend longevity in model organisms, aging is a complex trait, and targeting one's own genes may be insufficient to prevent age-dependent deterioration. An alternative strategy could be to use enzymes from other species to reverse age-associated metabolic changes. In this review, we discuss a set of enzymes from lower organisms that have been shown to affect various metabolic parameters linked to age-related processes. These enzymes include modulators of steady-state levels of amino acids (METase, ASNase, and ADI), NADPH/NADP+ and/or reduced form of coenzyme Q (CoQH2)/CoQ redox potentials (NDI1, AOX, LbNOX, TPNOX, EcSTH, RquA, LOXCAT, Grubraw, and ScURA), GSH (StGshF), mitochondrial membrane potential (mtON and mito-dR), or reactive oxygen species (DAAO and KillerRed-SOD1). We propose that leveraging non-mammalian enzymes represents an untapped resource that can be used to delay aging and age-related diseases.
Collapse
Affiliation(s)
- Andrey A Parkhitko
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, PA, USA
| | - Valentin Cracan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
5
|
Kubota Y, Zhao M, Han Q, Aoki Y, Masaki N, Obara K, Morinaga S, Mizuta K, Sato M, Bouvet M, Kubota K, Tsunoda T, Hoffman RM. Engineered Methioninase-expressing Tumor-targeting Salmonella typhimurium A1-R Inhibits Syngeneic-Cancer Mouse Models by Depleting Tumor Methionine. Cancer Genomics Proteomics 2025; 22:247-257. [PMID: 39993809 PMCID: PMC11880925 DOI: 10.21873/cgp.20499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND/AIM We previously developed Salmonella typhimurium A1-R, which selectively targets and kills tumors. In the present study, we established recombinant methioninase (rMETase)-producing Salmonella typhimurium A1-R (A1-R-rMETase), by transfer of the Pseudomonas putida methioninase gene, to target methionine addiction of syngeneic-cancer mouse models. MATERIALS AND METHODS A plasmid containing the Pseudomonas putida methioninase gene was extracted from METase-producing recombinant E. coli and inserted into Salmonella typhimurium A1-R using electroporation. Lewis Lung Carcinoma (LLC) cells (106) were injected subcutaneously in male C57BL/6 mice aged 4-6 weeks. We determined that 108 Salmonella typhimurium A1-R-rMETase administered iv was a safe dosage in C57BL/6 mice and was used for efficacy studies on LLC tumors in C57BL/6 mice. Tumor size was measured with calipers three times per week for 3 weeks. On day 22, tumor methionine levels were measured using HPLC in the control mice injected with phosphate-buffered saline (PBS) and the mice injected with Salmonella typhimurium A1-R-rMETase. RESULTS The mean LLC tumor size of each group on day 22 was as follows: PBS control: 741.5 mm3; mice injected with Salmonella typhimurium A1-R: 566.3 mm3 (p=0.370); and mice injected with Salmonella typhimurium A1-R-rMETase: 198.8 mm3 (p=0.0003 vs control and p=0.0117 vs. Salmonella A1-R). The mice injected with Salmonella typhimurium A1-R-rMETase showed a significantly lower mean tumor methionine level than mice injected with PBS (5.9 nM/mg protein vs 11.1 nM/mg protein, p=0.0095). Salmonella typhimurium A1-R-rMETase grew continuously in the tumors but not in the liver or spleen. CONCLUSION Tumor-targeting Salmonella typhimurium A1-R engineered to express the Pseudomonas putida methioninase gene, inhibited LLC tumor growth in a syngeneic mouse model and reduced the methionine level in the tumor. Salmonella typhimurium A1-R-rMETase combines the tumor targeting and killing capability of Salmonella typhimurium A1-R plus rMETase which targets the methionine addiction of cancer.
Collapse
Affiliation(s)
- Yutaro Kubota
- AntiCancer Inc., San Diego, CA, U.S.A
- Department of Surgery, University of California, San Diego, CA, U.S.A
- Division of Internal Medicine, Department of Medical Oncology, Showa University School of Medicine, Tokyo, Japan
| | - Ming Zhao
- AntiCancer Inc., San Diego, CA, U.S.A
| | | | - Yusuke Aoki
- AntiCancer Inc., San Diego, CA, U.S.A
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Noriyuki Masaki
- AntiCancer Inc., San Diego, CA, U.S.A
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Koya Obara
- AntiCancer Inc., San Diego, CA, U.S.A
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Sei Morinaga
- AntiCancer Inc., San Diego, CA, U.S.A
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Kohei Mizuta
- AntiCancer Inc., San Diego, CA, U.S.A
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Motokazu Sato
- AntiCancer Inc., San Diego, CA, U.S.A
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Koichi Kubota
- Department of Microbiology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Takuya Tsunoda
- Division of Internal Medicine, Department of Medical Oncology, Showa University School of Medicine, Tokyo, Japan
| | - Robert M Hoffman
- AntiCancer Inc., San Diego, CA, U.S.A.;
- Department of Surgery, University of California, San Diego, CA, U.S.A
| |
Collapse
|
6
|
Tsapieva AN, Chernov AN, Duplik NV, Morozova AO, Filatenkova TA, Suvorova MA, Egidarova E, Galimova ES, Bogatireva K, Suvorov AN. Studying the Oncolytic Activity of Streptococcus pyogenes Strains Against Hepatoma, Glioma, and Pancreatic Cancer In Vitro and In Vivo. Microorganisms 2025; 13:76. [PMID: 39858844 PMCID: PMC11767589 DOI: 10.3390/microorganisms13010076] [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: 12/12/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Cancer remains a leading cause of mortality globally. Conventional treatment modalities, including radiation and chemotherapy, often fall short of achieving complete remission, highlighting the critical need for novel therapeutic strategies. One promising approach involves the oncolytic potential of Group A Streptococcus (GAS) strains for tumor treatment. This study aimed to investigate the oncolytic efficacy of S. pyogenes GUR and its M protein knockout mutant, S. pyogenes strain GURSA1, which was genetically constructed to minimize overall toxicity, against mouse hepatoma 22A, pancreatic cancer PANC02, and human glioma U251 cells, both in vitro and in vivo, using the C57BL/6 mouse model. METHODS The in vitro oncolytic cytotoxic activity of GAS strains was studied against human glioma U251, pancreatic cancer PANC02, murine hepatoma 22a, and normal skin fibroblast cells using the MTT assay and the real-time xCELLigence system. A syngeneic mouse model of hepatoma and pancreatic cancer was used to evaluate the in vivo oncolytic effect of GAS strains. Statistical analysis was conducted using Student's t-test and Mann-Whitney U-test with GraphPad Prism software. RESULTS The in vitro model showed that the live S. pyogenes GUR strain had a strong cytotoxic effect (67.4 ± 1.9%) against pancreatic cancer PANC02 cells. This strain exhibited moderate (38.0 ± 1.8%) and weak (16.3 ± 5.4%) oncolytic activities against glioma and hepatoma cells, respectively. In contrast, the S. pyogenes GURSA1 strain demonstrated strong (86.5 ± 1.6%) and moderate (36.5 ± 1.8%) oncolytic activities against glioma and hepatoma cells. Additionally, the S. pyogenes GURSA1 strain did not exhibit cytotoxic activity against healthy skin fibroblast cells (cell viability 104.2 ± 1.3%, p = 0.2542). We demonstrated that tumor treatment with S. pyogenes GURSA1 significantly increased the lifespan of C57BL/6 mice with hepatoma (34 days, p = 0.040) and pancreatic cancer (32 days, p = 0.039) relative to the control groups (24 and 28 days, respectively). Increased lifespan was accompanied by a slowdown in tumor progression, as evidenced by a reduction in the growth of hepatoma and pancreatic cancer tumors under treatment with GAS strains in mice. CONCLUSIONS Both S. pyogenes GUR and S. pyogenes GURSA1 strains demonstrated strong oncolytic activity against murine hepatoma 22a, pancreatic cancer PANC02, and human U251 glioma cells in vitro. In contrast, S. pyogenes GUR and GURSA1 did not show toxicity against human normal skin fibroblasts. The overall survival rate and lifespan of mice treated with S. pyogenes GURSA1, a strain lacking the M protein on its surface, were significantly higher compared to the control and S. pyogenes GUR strain groups.
Collapse
Affiliation(s)
- Anna N. Tsapieva
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
| | - Alexander N. Chernov
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
- Department of Biological Chemistry, Federal State Budgetary Educational Institution of Higher Education Saint Petersburg, State Pediatric Medical University of the Ministry of Health of Russia, 194100 Saint Petersburg, Russia
| | - Nadezhda V. Duplik
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
| | - Anastasiya O. Morozova
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
| | - Tatiana A. Filatenkova
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
| | - Mariia A. Suvorova
- Department of Biotechnology and Biomedicine, Disease Systems Immunology, Technical University of Denmark, Søltofts Plads 224, 2800 Kongens Lyngby, Denmark;
| | - Elena Egidarova
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
| | - Elvira S. Galimova
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Thorez av., 44, 194223 Saint Petersburg, Russia
| | - Kseniya Bogatireva
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
| | - Alexander N. Suvorov
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the Center for Personalized Medicine of Federal State Budgetary Scientific Institution, Institute of Experimental Medicine, Acad. Pavlov Street, 12, 197022 Saint Petersburg, Russia; (N.V.D.); (A.O.M.); (T.A.F.); (E.E.); (E.S.G.); (K.B.); (A.N.S.)
| |
Collapse
|
7
|
He X, Guo J, Bai Y, Sun H, Yang J. Salmonella-based therapeutic strategies: improving tumor microenvironment and bringing new hope for cancer immunotherapy. Med Oncol 2024; 42:27. [PMID: 39666238 DOI: 10.1007/s12032-024-02578-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
Immunotherapy has revolutionized cancer treatment, yet its effectiveness is limited by immunosuppressive tumor microenvironment (TME). To overcome this challenge, innovative strategies to effectively modulate the TME are urgently needed. Over the past decades, bacteria-mediated cancer immunotherapy has recaptured increasing attention, driven by advances in synthetic biology, genetic engineering and our knowledge of host-pathogen interactions. Among various bacterial species, Salmonella has emerged as a leading candidate with significant therapeutic potential due to its broad-spectrum anti-tumor activity, tumor-targeting ability, immunomodulatory effects, oncolytic properties, genetic programmability, and engineering flexibility. These characteristics enable Salmonella to reshape the immunosuppressive TME, thereby enhancing anti-tumor efficacy. This review elaborates the regulatory effects of Salmonella on key components of the TME, the versatile engineering strategies for optimizing Salmonella's ability to modulate the TME, and recent advancements in combination cancer therapies. We also summarize current clinical applications and discuss challenges of developing safer and more effective Salmonella-based cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaoe He
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, 730030, Gansu, China
| | - Jiayin Guo
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, 730030, Gansu, China
| | - Yanrui Bai
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, 730030, Gansu, China
| | - Hui Sun
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, 730030, Gansu, China
| | - Jing Yang
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
8
|
Bansal G, Ghanem M, Sears KT, Galen JE, Tennant SM. Genetic engineering of Salmonella spp. for novel vaccine strategies and therapeutics. EcoSal Plus 2024; 12:eesp00042023. [PMID: 39023252 PMCID: PMC11636237 DOI: 10.1128/ecosalplus.esp-0004-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
Salmonella enterica is a diverse species that infects both humans and animals. S. enterica subspecies enterica consists of more than 1,500 serovars. Unlike typhoidal Salmonella serovars which are human host-restricted, non-typhoidal Salmonella (NTS) serovars are associated with foodborne illnesses worldwide and are transmitted via the food chain. Additionally, NTS serovars can cause disease in livestock animals causing significant economic losses. Salmonella is a well-studied model organism that is easy to manipulate and evaluate in animal models of infection. Advances in genetic engineering approaches in recent years have led to the development of Salmonella vaccines for both humans and animals. In this review, we focus on current progress of recombinant live-attenuated Salmonella vaccines, their use as a source of antigens for parenteral vaccines, their use as live-vector vaccines to deliver foreign antigens, and their use as therapeutic cancer vaccines in humans. We also describe development of live-attenuated Salmonella vaccines and live-vector vaccines for use in animals.
Collapse
Affiliation(s)
- Garima Bansal
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mostafa Ghanem
- Department of Veterinary Medicine, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Khandra T. Sears
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - James E. Galen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sharon M. Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
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
|
10
|
Wu L, Li L, Qiao L, Li C, Zhang S, Yin X, Du Z, Sun Y, Qiu J, Chang X, Wang B, Hua Z. Programmable Bacteria with Dynamic Virulence Modulation System for Precision Antitumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404069. [PMID: 39058336 PMCID: PMC11423194 DOI: 10.1002/advs.202404069] [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: 04/17/2024] [Revised: 07/11/2024] [Indexed: 07/28/2024]
Abstract
Engineered bacteria-mediated antitumor approaches have been proposed as promising immunotherapies for cancer. However, the off-target bacterial toxicity narrows the therapeutic window. Living microbes will benefit from their controllable immunogenicity within tumors for safer antitumor applications. In this study, a genetically encoded microbial activation strategy is reported that uses tunable and dynamic expression of surface extracellular polysaccharides to improve bacterial biocompatibility while retaining therapeutic efficacy. Based on screening of genes associated with Salmonella survival in macrophages, a novel attenuated Salmonella chassis strain AIS (htrA gene-deficient) highly enriched in tumors after administration and rapidly cleared from normal organs are reported. Subsequently, an engineered bacterial strain, AISI-H, is constructed based on the AIS strain and an optimized quorum-sensing regulatory system. The AISI-H strain can achieve recovery of dynamic tumor-specific bacterial virulence through a novel HTRA-RCSA axis-based and quorum-sensing synthetic gene circuit-mediated increase in extracellular polysaccharide content. These strains act "off" in normal organs to avoid unwanted immune activation and "on" in tumors for precise tumor suppression in mice. The AISI-H strain shows significant tumor inhibition and potent activation of anticancer immunity in a melanoma mouse model. The AISI-H strain exhibits excellent biocompatibility. This bacterial regulation strategy expands the applications of microbe-based antitumor therapeutics.
Collapse
Affiliation(s)
- Leyang Wu
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
- Nanjing Generecom Biotechnology Co., Ltd., Nanjing, Jiangsu, 210023, P. R. China
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories, Inc., Changzhou, Jiangsu, 213164, P. R. China
| | - Lin Li
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Liyuan Qiao
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Chenyang Li
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Shuhui Zhang
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Xingpeng Yin
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Zengzheng Du
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Ying Sun
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Jiahui Qiu
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Xiaoyao Chang
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Bohao Wang
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
| | - Zichun Hua
- Department of Neurology of Nanjing Drum Tower Hospital and The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu, 21008, P. R. China
- Nanjing Generecom Biotechnology Co., Ltd., Nanjing, Jiangsu, 210023, P. R. China
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories, Inc., Changzhou, Jiangsu, 213164, P. R. China
- Faculty of Pharmaceutical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453002, P. R. China
| |
Collapse
|
11
|
Tieu MV, Pham DT, Cho S. Bacteria-based cancer therapy: Looking forward. Biochim Biophys Acta Rev Cancer 2024; 1879:189112. [PMID: 38761983 DOI: 10.1016/j.bbcan.2024.189112] [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: 11/29/2023] [Revised: 03/25/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
The field of bacteria-based cancer therapy, which focuses on the key role played by the prevalence of bacteria, specifically in tumors, in controlling potential targets for cancer therapy, has grown enormously over the past few decades. In this review, we discuss, for the first time, the global cancer situation and the timeline for using bacteria in cancer therapy. We also explore how interdisciplinary collaboration has contributed to the evolution of bacteria-based cancer therapies. Additionally, we address the challenges that need to be overcome for bacteria-based cancer therapy to be accepted in clinical trials and the latest advancements in the field. The groundbreaking technologies developed through bacteria-based cancer therapy have opened up new therapeutic strategies for a wide range of therapeutics in cancer.
Collapse
Affiliation(s)
- My-Van Tieu
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Duc-Trung Pham
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Sungbo Cho
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea.
| |
Collapse
|
12
|
Song W, He Y, Feng Y, Wang Y, Li X, Wu Y, Zhang S, Zhong L, Yan F, Sun L. Image-Guided Photothermal and Immune Therapy of Tumors via Melanin-Producing Genetically Engineered Bacteria. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305764. [PMID: 38368252 DOI: 10.1002/smll.202305764] [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: 07/10/2023] [Revised: 01/10/2024] [Indexed: 02/19/2024]
Abstract
Photothermal therapy (PTT) is a new treatment modality for tumors. However, the efficient delivery of photothermal agents into tumors remains difficult, especially in hypoxic tumor regions. In this study, an approach to deliver melanin, a natural photothermal agent, into tumors using genetically engineered bacteria for image-guided photothermal and immune therapy is developed. An Escherichia coli MG1655 is transformed with a recombinant plasmid harboring a tyrosinase gene to produce melanin nanoparticles. Melanin-producing genetically engineered bacteria (MG1655-M) are systemically administered to 4T1 tumor-bearing mice. The tumor-targeting properties of MG1655-M in the hypoxic environment integrate the properties of hypoxia targeting, photoacoustic imaging, and photothermal therapeutic agents in an "all-in-one" manner. This eliminates the need for post-modification to achieve image-guided hypoxia-targeted cancer photothermal therapy. Tumor growth is significantly suppressed by irradiating the tumor with an 808 nm laser. Furthermore, strong antitumor immunity is triggered by PTT, thereby producing long-term immune memory effects that effectively inhibit tumor metastasis and recurrence. This work proposes a new photothermal and immune therapy guided by an "all-in-one" melanin-producing genetically engineered bacteria, which can offer broad potential applications in cancer treatment.
Collapse
Affiliation(s)
- Weijian Song
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| | - Yaling He
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Yanan Feng
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, P. R. China
| | - Yuanyuan Wang
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Xiaoying Li
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| | - Yingnan Wu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| | - Shanxin Zhang
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| | - Lin Zhong
- School of Public Health, Nanchang University, Nanchang, Jiangxi, 330019, P. R. China
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| |
Collapse
|
13
|
Zhang X, Zhao Z, Wang X, Zhang S, Zhao Z, Feng W, Xu L, Nie J, Li H, Liu J, Xiao G, Zhang Y, Li H, Lu M, Mai J, Zhou S, Zhao AZ, Li F. Deprivation of methionine inhibits osteosarcoma growth and metastasis via C1orf112-mediated regulation of mitochondrial functions. Cell Death Dis 2024; 15:349. [PMID: 38769167 PMCID: PMC11106329 DOI: 10.1038/s41419-024-06727-1] [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: 01/06/2024] [Revised: 04/26/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Osteosarcoma is a malignant bone tumor that primarily inflicts the youth. It often metastasizes to the lungs after chemotherapy failure, which eventually shortens patients' lives. Thus, there is a dire clinical need to develop a novel therapy to tackle osteosarcoma metastasis. Methionine dependence is a special metabolic characteristic of most malignant tumor cells that may offer a target pathway for such therapy. Herein, we demonstrated that methionine deficiency restricted the growth and metastasis of cultured human osteosarcoma cells. A genetically engineered Salmonella, SGN1, capable of overexpressing an L-methioninase and hydrolyzing methionine led to significant reduction of methionine and S-adenosyl-methionine (SAM) specifically in tumor tissues, drastically restricted the growth and metastasis in subcutaneous xenograft, orthotopic, and tail vein-injected metastatic models, and prolonged the survival of the model animals. SGN1 also sharply suppressed the growth of patient-derived organoid and xenograft. Methionine restriction in the osteosarcoma cells initiated severe mitochondrial dysfunction, as evident in the dysregulated gene expression of respiratory chains, increased mitochondrial ROS generation, reduced ATP production, decreased basal and maximum respiration, and damaged mitochondrial membrane potential. Transcriptomic and molecular analysis revealed the reduction of C1orf112 expression as a primary mechanism underlies methionine deprivation-initiated suppression on the growth and metastasis as well as mitochondrial functions. Collectively, our findings unraveled a molecular linkage between methionine restriction, mitochondrial function, and osteosarcoma growth and metastasis. A pharmacological agent, such as SGN1, that can achieve tumor specific deprivation of methionine may represent a promising modality against the metastasis of osteosarcoma and potentially other types of sarcomas as well.
Collapse
Affiliation(s)
- Xindan Zhang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Zhenggang Zhao
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xuepeng Wang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Shiwei Zhang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Zilong Zhao
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Wenbin Feng
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Lijun Xu
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Junhua Nie
- South China University of Technology School of Medicine, Guangzhou, China
| | - Hong Li
- Biomedical Laboratory, Guangzhou Jingke Life Science Institute, Guangzhou, China
| | - Jia Liu
- South China University of Technology School of Medicine, Guangzhou, China
| | - Gengmiao Xiao
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Yu Zhang
- Department of Orthopedic Oncology, Guangdong Provincial People's Hospital Affiliated to South China University of Technology School of Medicine, Guangzhou, China
| | - Haomiao Li
- Department of Musculoskeletal Oncology, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ming Lu
- Department of Musculoskeletal Oncology, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jialuo Mai
- Guangzhou Sinogen Pharmaceutical Co., Ltd., Guangzhou, Guangdong Province, China
| | - Sujin Zhou
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Allan Z Zhao
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Fanghong Li
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
14
|
Mantooth SM, Abdou Y, Saez-Ibañez AR, Upadhaya S, Zaharoff DA. Intratumoral delivery of immunotherapy to treat breast cancer: current development in clinical and preclinical studies. Front Immunol 2024; 15:1385484. [PMID: 38803496 PMCID: PMC11128577 DOI: 10.3389/fimmu.2024.1385484] [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: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Breast cancer poses one of the largest threats to women's health. Treatment continues to improve for all the subtypes of breast cancer, but some subtypes, such as triple negative breast cancer, still present a significant treatment challenge. Additionally, metastasis and local recurrence are two prevalent problems in breast cancer treatment. A newer type of therapy, immunotherapy, may offer alternatives to traditional treatments for difficult-to-treat subtypes. Immunotherapy engages the host's immune system to eradicate disease, with the potential to induce long-lasting, durable responses. However, systemic immunotherapy is only approved in a limited number of indications, and it benefits only a minority of patients. Furthermore, immune related toxicities following systemic administration of potent immunomodulators limit dosing and, consequently, efficacy. To address these safety considerations and improve treatment efficacy, interest in local delivery at the site of the tumor has increased. Numerous intratumorally delivered immunotherapeutics have been and are being explored clinically and preclinically, including monoclonal antibodies, cellular therapies, viruses, nucleic acids, cytokines, innate immune agonists, and bacteria. This review summarizes the current and past intratumoral immunotherapy clinical landscape in breast cancer as well as current progress that has been made in preclinical studies, with a focus on delivery parameters and considerations.
Collapse
Affiliation(s)
- Siena M. Mantooth
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
| | - Yara Abdou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | - David A. Zaharoff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
15
|
Zhou S, Zhang S, Zheng K, Li Z, Hu E, Mu Y, Mai J, Zhao A, Zhao Z, Li F. Salmonella-mediated methionine deprivation drives immune activation and enhances immune checkpoint blockade therapy in melanoma. J Immunother Cancer 2024; 12:e008238. [PMID: 38302417 PMCID: PMC10836381 DOI: 10.1136/jitc-2023-008238] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Although immune checkpoint inhibitor (ICI)-based therapy is advantageous for patients with advanced melanoma, resistance and relapse are frequent. Thus, it is crucial to identify effective drug combinations and develop new therapies for the treatment of melanoma. SGN1, a genetically modified Salmonella typhimurium species that causes the targeted deprivation of methionine in tumor tissues, is currently under investigation in clinical trials. However, the inhibitory effect of SGN1 on melanoma and the benefits of SGN1 in combination with ICIs remain largely unexplored. Therefore, this study aims to investigate the antitumor potential of SGN1, and its ability to enhance the efficacy of antibody-based programmed cell death-ligand 1 (PD-L1) inhibitors in the treatment of murine melanoma. METHODS The antitumor activity of SGN1 and the effect of SGN1 on the efficacy of PD-L1 inhibitors was studied through murine melanoma models. Further, The Cancer Genome Atlas-melanoma cohort was clustered using ConsensusClusterPlus based on the methionine deprivation-related genes, and immune characterization was performed using xCell, Microenvironment Cell Populations-counter, Estimation of Stromal and Immune cells in MAlignant Tumor tissues using Expression data, and immunophenoscore (IPS) analyses. The messenger RNA data on programmed death-1 (PD-1) immunotherapy response were obtained from the Gene Expression Omnibus database. Gene Set Enrichment Analysis of methionine deprivation-up gene set was performed to determine the differences between pretreatment responders and non-responders. RESULTS This study showed that both, the intratumoral and the intravenous administration of SGN1 in subcutaneous B16-F10 melanomas, suppress tumor growth, which was associated with an activated CD8+T-cell response in the tumor microenvironment. Combination therapy of SGN1 with systemic anti-PD-L1 therapy resulted in better antitumor activity than the individual monotherapies, respectively, and the high therapeutic efficacy of the combination was associated with an increase in the systemic level of tumor-specific CD8+ T cells. Two clusters consisting of methionine deprivation-related genes were identified. Patients in cluster 2 had higher expression of methionine_deprivation_up genes, better clinical outcomes, and higher immune infiltration levels compared with patients in cluster 1. Western blot, IPS analysis, and immunotherapy cohort study revealed that methionine deficiency may show a better response to ICI therapy CONCLUSIONS:: This study reports Salmonella-based SGN1 as a potent anticancer agent against melanoma, and lays the groundwork for the potential synergistic effect of ICIs and SGN1 brought about by improving the immune microenvironment in melanomas.
Collapse
Affiliation(s)
- Sujin Zhou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Shiwei Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Kexin Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Zixuan Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Enyu Hu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Yunping Mu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Jialuo Mai
- Guangzhou Sinogen Pharmaceutical Co Ltd, Guangzhou, Guangdong, China
| | - Allan Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Zhenggang Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Fanghong Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Pérez Jorge G, Gontijo MTP, Brocchi M. Salmonella enterica and outer membrane vesicles are current and future options for cancer treatment. Front Cell Infect Microbiol 2023; 13:1293351. [PMID: 38116133 PMCID: PMC10728604 DOI: 10.3389/fcimb.2023.1293351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
Conventional cancer therapies have many limitations. In the last decade, it has been suggested that bacteria-mediated immunotherapy may circumvent the restrictions of traditional treatments. For example, Salmonella enterica is the most promising bacteria for treating cancer due to its intrinsic abilities, such as killing tumor cells, targeting, penetrating, and proliferating into the tumor. S. enterica has been genetically modified to ensure safety and increase its intrinsic antitumor efficacy. This bacterium has been used as a vector for delivering anticancer agents and as a combination therapy with chemotherapy, radiotherapy, or photothermic. Recent studies have reported the antitumor efficacy of outer membrane vesicles (OMVs) derived from S. enterica. OMVs are considered safer than attenuated bacteria and can stimulate the immune system as they comprise most of the immunogens found on the surface of their parent bacteria. Furthermore, OMVs can also be used as nanocarriers for antitumor agents. This review describes the advances in S. enterica as immunotherapy against cancer and the mechanisms by which Salmonella fights cancer. We also highlight the use of OMVs as immunotherapy and nanocarriers of anticancer agents. OMVs derived from S. enterica are innovative and promising strategies requiring further investigation.
Collapse
Affiliation(s)
- Genesy Pérez Jorge
- Universidade Estadual de Campinas (UNICAMP), Departamento de Genética, Evolução, Microbiologia e Imunologia, Laboratório de Doenças Tropicais, Instituto de Biologia, Campinas, Brazil
| | - Marco Túlio Pardini Gontijo
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Marcelo Brocchi
- Universidade Estadual de Campinas (UNICAMP), Departamento de Genética, Evolução, Microbiologia e Imunologia, Laboratório de Doenças Tropicais, Instituto de Biologia, Campinas, Brazil
| |
Collapse
|