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
|
Khormi MA, Al-maaqar SM, Al Johni AR, Al-Tayyar NA, Alhamad JA, Ghyathuddin AA, Alblawi Z, Behairy SM, Alghamdi MA, Alsubhi WA, Teklemariam MD. Oncolytic bacteria: A revolutionary approach to cancer therapy. Open Life Sci 2025; 20:20251076. [PMID: 40519767 PMCID: PMC12163578 DOI: 10.1515/biol-2025-1076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/18/2025] [Accepted: 02/11/2025] [Indexed: 06/18/2025] Open
Abstract
Cancer is one of the most devastating diseases all over the globe, and it is the second worldwide cause of death, exceeded only by cardiovascular diseases. The therapeutic approach to human cancer has evolved significantly and has varied depending on the type and stage of cancer, as well as the general health status of the patient. Despite the advancements in cancer treatment, various challenges persist in the treatment of cancer, including side effects, drug resistance, and incomplete eradication of tumors. The use of oncolytic bacteria (cancer targeting and destroying bacteria) has been identified to have several advantages over the traditional methods of cancer treatment. Several bacterial species have been identified to be used in the treatment of different types of cancers. Oncolytic therapy can be achieved through the use of a naturally occurring and/or genetically modified bacterial species, including Clostridium, Salmonella, Escherichia coli, and Listeria spp. with their toxins, enzymes, biofilms, and secondary metabolites as well as their spores that leads to direct or indirect killing of cancer cells. This review provides some highlights about the biology and therapeutic potential of oncolytic bacteria individually or in combination with other therapeutic approaches against different types of cancers. Besides, the current challenges and future perspectives will be explored.
Collapse
Affiliation(s)
- Mohsen A. Khormi
- Department of Biology, College of Science, Jazan University, P.O. Box 114, Jazan, 45142, Saudi Arabia
| | - Saleh M. Al-maaqar
- Department of Biology, Faculty of Education, Albaydha University, Al-Baydha, Yemen
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | | | | | - Jafar Abdullah Alhamad
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Abdullah A. Ghyathuddin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Zakia Alblawi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Sabreen M. Behairy
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Mohammed A. Alghamdi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Wael A. Alsubhi
- Department of Pharmacy Practice, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Mikiyas D. Teklemariam
- School of Nursing, Department of Midwifery, Addis Ababa University, College of Health Sciences, Addis Ababa, Ethiopia
| |
Collapse
|
3
|
Aktay-Cetin Ö, Pullamsetti SS, Herold S, Savai R. Lung tumor immunity: redirecting macrophages through infection-induced inflammation. Trends Immunol 2025; 46:471-484. [PMID: 40382244 DOI: 10.1016/j.it.2025.04.004] [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: 01/05/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
Macrophages play a central role in maintaining tissue homeostasis and in surveillance against pathogens and disease. In the lung, they can adopt either proinflammatory or anti-inflammatory states depending on the nature of the stimulus. As the predominant immune cells in both the lung tumor microenvironment and in sites of lung infection, the functional plasticity of macrophages makes them key players in determining disease outcome. Accurately defining their inflammatory profiles offers an opportunity to reprogram infection-associated macrophages towards enhanced tumor-killing phenotypes. This review explores how acute inflammation can drive macrophage-mediated antitumor immunity and highlights key molecules and signaling pathways that may be leveraged to therapeutically modulate macrophage function.
Collapse
Affiliation(s)
- Öznur Aktay-Cetin
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Soni Savai Pullamsetti
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany; Department of Internal Medicine II, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Susanne Herold
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Department of Internal Medicine V, German Center for Lung Research (DZL), German Center for Infection Research (DZIF), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany; Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany; Department of Internal Medicine II, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.
| |
Collapse
|
4
|
Blair AB, Zheng L, Soares KC. The Landmark Series: Therapeutic Cancer Vaccine Strategies for Cold Tumors. Ann Surg Oncol 2025:10.1245/s10434-025-17281-1. [PMID: 40325301 DOI: 10.1245/s10434-025-17281-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 05/07/2025]
Abstract
Immunologically cold tumors present a significant challenge in cancer treatment due to their limited baseline immune infiltration and resistance to immunotherapy. Cancer vaccines offer a promising strategy to overcome this barrier by introducing high-quality, tumor-relevant antigens that can stimulate an effective anti-tumor immune response. Therapeutic cancer vaccines are being explored in the neoadjuvant, adjuvant, and minimal residual disease contexts to enhance immune activation and promote immune cell infiltration and function, with the goal to eradicate malignant cells and improve patient survival. Critical hurdles remain in optimizing antigen selection, determining the most effective vaccine formulations, and defining the ideal clinical setting for vaccine use. Moreover, rational combinations of cancer vaccines with other immune modulators (e.g., adjuvants, immune checkpoint inhibitors, and cytokines) may hold the key to enhancing vaccine efficacy and expanding therapeutic options for difficult-to-treat malignancies. This review examines current advancements in cancer vaccines and their utilization for immunologically cold tumors in the perioperative setting, highlighting ongoing challenges and future directions in this evolving field.
Collapse
Affiliation(s)
- Alex B Blair
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lei Zheng
- Mays Cancer Center at the University of Texas Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
- Department of Oncology and Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin C Soares
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
5
|
Laine RA, Lopez HW, Takematsu H. Tumor Hemorrhage-inducing polysaccharides secreted by streptococci and Serratia proposed as the active principal ingredients (API's) of Coley's toxin: on PS1, the Serratia marcescens API. Glycobiology 2025; 35:cwaf021. [PMID: 40226871 DOI: 10.1093/glycob/cwaf021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Coley's Toxin comprised a mixture of cell-free, heat-treated culture media from Streptococcus pyogenes (originally Streptococus erysipelatos) and Serratia marcescens (originally Bacillus prodigiosus). A 250 kDa tumor hemorrhage-inducing polysaccharide "PS1" is reported here secreted into culture medium by S. marcescens. Four h after PS1 is injected at 32 μg/kg (10pM) into the tail vein of Balb/C mice bearing C26 subcutaneous colon-derived tumors, tumor-specific capillary hemorrhage is exhibited in 90% of tumors. As a positive control, CM101, a similar tumor hemorrhagic polysaccharide from Streptococcus agalactica caused tumor hemorrhage in 75% of tumors in the Balb/C-C26 model at 7.5 μg/kg(2.5pM). CM101 has previously been safety tested in a Phase I clinical trial. These two polysaccharides have merit to be identified as the active principal ingredients (API's) of Coley'sToxin. Additional approaches to cancer therapy are a global need. No matter the level of wealth of victims, some cancers are still incurable. Recall in recent years the tragic early cancer deaths of Steve Jobs and Paul Allen among other luminaries. Streptococcal and Serratia bacterial extracts have unique tumor specific capillary destructive activity, with observations originating with sarcomas cured by nosocomial erysipelas infections in the 1860's. The active pharmaceutical ingredients (API's) in these extracts and Coley's Toxins are proposed to be polysaccharides.
Collapse
Affiliation(s)
- Roger A Laine
- Departments of Biological Sciences, and Chemistry, Louisiana State University and A&M College, College of Science, 201 Life Sciences Bldg. Baton Rouge, LA 70803, USA
| | - Henry W Lopez
- Murigenics, Inc., 941 Railroad Ave. Vallejo, CA 94592, USA
| | - Hiromu Takematsu
- Graduate School of Health Sciences, Fujita Health University, Building 3 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
6
|
Fu Z, Lin S, Chen H, Guo H, Li J, Chen Y, Lu Y, Liu J, Huang W, Pang Y. Generating Self-Adjuvated Nanofiber Vaccines by Coating Bacterial Flagella with Antigens. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415887. [PMID: 39981905 DOI: 10.1002/adma.202415887] [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: 10/17/2024] [Revised: 01/28/2025] [Indexed: 02/22/2025]
Abstract
Bacteria-based vaccines have received increasing attention given the ability to induce strong systemic immune responses. However, the application of bacteria as therapeutic agents inevitably suffers from infection-associated side effects due to the living characteristics. Here, the use of bacteria-derived flagella is described to construct self-adjuvated nanofiber vaccines. With the help of charge-reversal mediated by decoration with cationic polymers, the flagella can be coated with negatively charged antigens through electrostatic interaction. By virtue of the large aspect ratio, the resulting nanofiber vaccines show prolonged retention at the injection site and increased uptake by dendritic cells and macrophages. Thanks to the innate immunogenicity, self-adjuvated flagella robustly promote dendritic cell maturation and macrophage polarization, resulting in the elicitation of antigen-specific T-cell and B-cell immune responses. In ovalbumin-overexpressing melanoma-bearing mice, immunization with ovalbumin-carried vaccines not only exhibits a favorable tolerance, but also displays superior inhibition efficacies on tumor growth and metastasis separately under the therapeutic and prophylactic settings. The flexibility of this approach is further demonstrated for vaccine fabrication by coating with the SARS-CoV-2 Spike protein S1 subunit. Bacterial flagella-based self-adjuvated nanofiber platform proposes a versatile strategy to develop various vaccines for disease prevention and treatment.
Collapse
Affiliation(s)
- Zhenzhen Fu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huan Chen
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haiyan Guo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Juanjuan Li
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanmei Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yue Lu
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Xiangfu Laboratory, Jiaxing, Zhejiang, 314102, China
| | - Yan Pang
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
7
|
Choi H, Choi B, Kim DH. Anaerobic bacterial metabolism responsive microspheres for bacterial embolization cancer therapy. Biomaterials 2025; 314:122902. [PMID: 39454505 DOI: 10.1016/j.biomaterials.2024.122902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/09/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
Anaerobic bacteriolytic cancer therapy, whether delivered locally or systemically, frequently encounters challenges related to limited colonization within hypoxic pockets of central tumors and activation of innate immunity. Herein we have developed trans-arterial bacteria embolization therapy using bacterial embolic microspheres. C. novyi-NT spores loaded calcium alginate embolic microspheres demonstrated C. novyi-NT metabolites-mediated microsphere degradation, releasing vegetative C. novyi-NT bacterial in hypoxic condition. Transcatheter directed bacterial microsphere embolization therapy occludes tumor feeding vessels with infused bacterial embolic microspheres and enhances tumoral hypoxia. Notably, anaerobic bacterial metabolism responsive microsphere-bacterial embolization therapy achieved a complete tumor response with enhanced tumor-specific bacterial delivery and colonization, resulting in cancer cell killing across the entire tumor. In vivo tumor response and immunological profiling revealed that bacterial embolization uniquely enhances anti-cancer response, effectively engaging direct anaerobic bacterial oncolysis and adaptive and innate immune responses in a cooperative manner.
Collapse
Affiliation(s)
- Hyunjun Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Bongseo Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA; Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, 60208, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
8
|
Kirtane AR, Traverso G. Improving the Efficacy of Cancer mRNA Vaccines. Cancer J 2025; 31:e0764. [PMID: 40126883 DOI: 10.1097/ppo.0000000000000764] [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: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 03/26/2025]
Abstract
mRNA vaccines consist of antigen-encoding mRNA, which produces the antigenic protein upon translation. Coupling antigen production with innate immune activation can generate a potent, antigen-specific T-cell response. Clinical reports have demonstrated the ability of mRNA vaccines to elicit an anticancer immune response against various tumor types. Here, we discuss strategies to enhance the potency of mRNA vaccines. We provide an overview of existing knowledge regarding the activation and trafficking mechanisms of mRNA vaccines and share optimization strategies to boost mRNA-mediated antigen production. In addition, we address methods to target mRNA vaccines to dendritic cells and lymph nodes, key initiators of the immune response. Finally, we review strategies for enhancing immune activation using adjuvants compatible with mRNA vaccines. mRNA vaccines offer unique advantages that can be utilized for oncology applications. However, significant work is needed to understand their underlying mechanisms and develop technologies to improve their effectiveness.
Collapse
Affiliation(s)
- Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology
- Broad Institute, Massachusetts Institute of Technology, Cambridge, MA
- Department of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
9
|
Zhang X, Zang Z, Liang Z, Xu X, Zheng J, Liang N, Shabiti S, Wang Z, Yu S, Wang Y, Liu C, Li W, Cai L. Nanobiohybrid Oncolytic Bacteria with Optimized Intratumoral Distribution for Combined Sono-Photodynamic/Immunotherapy. ACS NANO 2025; 19:6437-6453. [PMID: 39902865 DOI: 10.1021/acsnano.4c16740] [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: 02/06/2025]
Abstract
"Living therapeutic carriers" present a promising avenue for cancer research, but it is still challenging to achieve uniform and durable distribution of payloads throughout the solid tumor owing to the tumor microenvironment heterogeneity. Herein, a living drug sprinkle biohybrid (YB1-HCNs) was constructed by hitching acid/enzyme-triggered detachable nanoparticles (HCNs) backpack on the surface of metabolic oligosaccharide-engineered oncolytic bacteria YB1. Along with the process of tumor penetration by bacterial hypoxia navigation, YB1-HCNs responsively and continuously release HCNs, achieving a uniform distribution of loaded agents throughout the tumor. Upon successive irradiation of laser and ultrasound (US), the HCNs can separately generate type II and type I ROS for superior sono-photodynamic therapy (SPDT), which enables HCNs to synergize with YB1 for a satisfactory therapeutic effect in both superficial normoxic and deep hypoxic regions of the tumor. After a single dose, this efficient combination realized 98.3% primary tumor inhibition rate and prolonged survival of mice for 90 days with no recurrence, further inducing a powerful immunological memory effect to completely suppress tumor rechallenge in cured mice. Such a bacterial hybridization vector enables optimization of the spatial distribution of YB1 and HCNs, providing an innovative strategy to maximize therapeutic outcomes and evoke durable antitumor immunity.
Collapse
Affiliation(s)
- Xu Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zhongsheng Zang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhenguo Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Xiaoyu Xu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Jinling Zheng
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Na Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Shayibai Shabiti
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zixi Wang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Shiwen Yu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Yujue Wang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Chenli Liu
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenjun Li
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Sino-Euro Center of Biomedicine and Health, Luohu, Shenzhen, 518024, P. R. China
| |
Collapse
|
10
|
Hu Y, Yang R, Ni S, Song Z. Bibliometric analysis of targeted immunotherapy for osteosarcoma-current knowledge, hotspots and future perspectives. Front Immunol 2025; 15:1485053. [PMID: 39995821 PMCID: PMC11847827 DOI: 10.3389/fimmu.2024.1485053] [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: 08/23/2024] [Accepted: 12/24/2024] [Indexed: 02/26/2025] Open
Abstract
Objective The objective of this study is to conduct a bibliometric analysis on examining the current condition, areas of interest, and rising trends of immunotherapy for osteosarcoma (ITFOS), as well as its importance in associated research domains. Methods An extensive collection of academic papers on the use of ITFOS was obtained from the Web of Science between January 1, 2000 and October 20, 2023. Then, using a variety of tools like HisCite, VOSviewer, CiteSpace, and the bibliometrix package, a bibliometric study was carried out. This study included the collection of information on country, institution, author, journal, and keywords. Results A comprehensive analysis was undertaken on a total of 616 publications obtained from 247 journals, encompassing the contributions of 3725 authors affiliated with 831 institutes spanning across 43 countries/regions. Notably, China exhibited the highest quantity of published 277 (44.99%) articles on ITFOS. The most productive institution was Zhejiang University, with 26 (4.22%) publications. The author with the highest publication output was Tsukahara, Tomohide from Japan with 15 (2.44%) publications. The article with the most citation was "DOI: 10.1200/JCO.2014.58.0225". Frontiers in Immunology demonstrated the highest level of productivity, having published a total of 31 (5.03%) articles. The most frequently used were "osteosarcoma," "immunotherapy," and "cancer,". Meanwhile, "sequencing", "prognostic signature" and "immune microenvironment" have been identified as the research frontiers for the forthcoming years. Conclusion This paper provides a thorough evaluation of current research trends and advancements in ITFOS. It includes relevant research findings and emphasizes collaborative efforts among authors, institutions, and countries.
Collapse
Affiliation(s)
- Yunxiang Hu
- Department of Orthopaedic Trauma, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, China
| | - Rui Yang
- School of Graduates, Dalian Medical University, Dalian, Liaoning, China
| | - Shuai Ni
- Department of Orthopaedic Trauma, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, China
| | - Zefeng Song
- School of Graduates, Dalian University of Technology, Dalian, Liaoning, China
| |
Collapse
|
11
|
Adewuyi E, Chorya H, Muili A, Moradeyo A, Kayode A, Naik A, Odedele T, Opabode M. Chemotherapy, immunotherapy, and targeted therapy for osteosarcoma: Recent advancements. Crit Rev Oncol Hematol 2025; 206:104575. [PMID: 39581243 DOI: 10.1016/j.critrevonc.2024.104575] [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: 05/30/2024] [Revised: 10/22/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
Recent advancements in the treatment of osteosarcoma, a rare and aggressive form of bone cancer, have seen significant progress with chemotherapy, immunotherapy, and targeted therapy. Chemotherapy, the conventional approach, has witnessed refined drug regimens and novel agents tailored to enhance efficacy while minimizing adverse effects. This evolution aims to strike a balance between eradicating cancer cells and preserving patients' overall well-being. Immunotherapy has emerged as a promising avenue, leveraging the body's immune system to recognize and combat cancer cells. Innovative immunotherapeutic strategies, including immune checkpoint inhibitors, adoptive T cell therapy, and chimeric antigen receptor (CAR)-T cell therapy, exhibit the potential to enhance immune responses against osteosarcoma. Moreover, targeted therapy, designed to disrupt specific molecular pathways crucial for cancer growth, has gained traction in the treatment of osteosarcoma. Precision medicine approaches, such as identifying biomarkers and employing targeted agents, aim to tailor therapies to individual patients, maximizing effectiveness while minimizing collateral damage to healthy tissues. This article analyzes the current state of these three treatment modalities while comparing the efficacies of current chemotherapy, immunotherapy and targeted therapy agents.
Collapse
Affiliation(s)
- Esther Adewuyi
- Department of Medicine and Surgery, Ladoke Akintola University, Ogbomoso, Nigeria; Ladoke Akintola University Medical Journal Club, Ogbomoso, Nigeria.
| | - Harshal Chorya
- Department of Medicine and Surgery, Baroda Medical College, India
| | - Abdulbasit Muili
- Department of Medicine and Surgery, Ladoke Akintola University, Ogbomoso, Nigeria; Ladoke Akintola University Medical Journal Club, Ogbomoso, Nigeria
| | - Abdulrahmon Moradeyo
- Department of Medicine and Surgery, Ladoke Akintola University, Ogbomoso, Nigeria; Ladoke Akintola University Medical Journal Club, Ogbomoso, Nigeria
| | - Ayomide Kayode
- Department of Medicine and Surgery, Ladoke Akintola University, Ogbomoso, Nigeria; Ladoke Akintola University Medical Journal Club, Ogbomoso, Nigeria
| | - Aastha Naik
- Department of Medicine and Surgery, Parul Institute of Medical Sciences and Research, Parul University, India
| | - Temitayo Odedele
- Department of Medicine and Surgery, Ladoke Akintola University, Ogbomoso, Nigeria; Ladoke Akintola University Medical Journal Club, Ogbomoso, Nigeria
| | - Muntaqim Opabode
- Department of Medicine and Surgery, Ladoke Akintola University, Ogbomoso, Nigeria; Ladoke Akintola University Medical Journal Club, Ogbomoso, Nigeria
| |
Collapse
|
12
|
van den Berg J, Läubli H, Khanna N, Jeker LT, Holbro A. Basic Concepts and Indications of CAR T Cells. Hamostaseologie 2025; 45:14-23. [PMID: 39970899 DOI: 10.1055/a-2491-3652] [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: 02/21/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized cancer immunotherapy, particularly for hematological malignancies. This personalized approach is based on genetically engineering T cells derived from the patient to target antigens expressed-among others-on malignant cells. Nowadays they offer new hope where conventional therapies, such as chemotherapy and radiation, have often failed. Since the first FDA approval in 2017, CAR T cell therapy has rapidly expanded, proving highly effective against previously refractory diseases with otherwise a dismal outcome. Despite its promise, CAR T cell therapy continues to face significant challenges, including complex manufacturing, the management of toxicities, resistance mechanisms that impact long-term efficacy, and limited access as well as high costs, which continue to shape ongoing research and clinical applications. This review aims to provide an overview of CAR T cell therapy, including its fundamental concepts, clinical applications, current challenges, and future directions in hematological malignancies.
Collapse
Affiliation(s)
- Jana van den Berg
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Nina Khanna
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Lukas T Jeker
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Andreas Holbro
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| |
Collapse
|
13
|
Oyama R, Nabeshima A, Endo M, Novikov A, Fujiwara T, Phelip C, Yokoyama N, Oda Y, Caroff M, Matsumoto Y, Kerzerho J, Nakashima Y. A detoxified TLR4 agonist inhibits tumour growth and lung metastasis of osteosarcoma by promoting CD8+ cytotoxic lymphocyte infiltration. BJC REPORTS 2025; 3:5. [PMID: 39870886 PMCID: PMC11772650 DOI: 10.1038/s44276-024-00120-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND Osteosarcoma is the most common malignant bone tumour with limited treatment options and poor outcomes in advanced metastatic cases. Current immunotherapies show limited efficacy, highlighting the need for novel therapeutic approaches. Systemic immune activation by Toll-like receptor 4 (TLR4) immunostimulants has shown great promise; however, current TLR4 agonists' toxicity hinders this systemic approach in patients with osteosarcoma. METHODS We compared the antitumour effect of lipopolysaccharides (LPS) with that of an innovative chemically detoxified TLR4 agonist (Lipo-MP-LPS) in a syngeneic metastatic osteosarcoma mouse model. Lipo-MP-LPS exhibited an optimal safety and solubility profile for systemic administration at an effective dose. We evaluated tumour growth, lung metastases, and immune cell infiltration in wild-type and TLR4-mutant mice and performed selective immunodepletion. RESULTS Lipo-MP-LPS exhibited antitumour effects against localised osteosarcoma tumours and lung metastases, like those of natural LPS. Lipo-MP-LPS promoted CD8+ T cells and M1 macrophages infiltration in primary tumours and CD8+ T cells in metastases, with an M1-phenotype macrophage shift. The Lipo-MP-LPS antitumour effects were found to depend on TLR4 and CD8+ T cells, but not on macrophages. CONCLUSION Lipo-MP-LPS inhibited tumour growth and lung metastasis of osteosarcoma by promoting CD8 + T cell infiltration, indicating its therapeutic potential for advanced osteosarcoma.
Collapse
Affiliation(s)
- Ryunosuke Oyama
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Nabeshima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Makoto Endo
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Alexey Novikov
- HEPHAISTOS-Pharma, Université Paris-Saclay, Orsay, France
| | - Toshifumi Fujiwara
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Nobuhiko Yokoyama
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Martine Caroff
- HEPHAISTOS-Pharma, Université Paris-Saclay, Orsay, France
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | | | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
14
|
Skalickova M, Hadrava Vanova K, Uher O, Leischner Fialova J, Petrlakova K, Masarik M, Kejík Z, Martasek P, Pacak K, Jakubek M. Injecting hope: the potential of intratumoral immunotherapy for locally advanced and metastatic cancer. Front Immunol 2025; 15:1479483. [PMID: 39850897 PMCID: PMC11754201 DOI: 10.3389/fimmu.2024.1479483] [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: 08/12/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Despite enormous progress, advanced cancers are still one of the most serious medical problems in current society. Although various agents and therapeutic strategies with anticancer activity are known and used, they often fail to achieve satisfactory long-term patient outcomes and survival. Recently, immunotherapy has shown success in patients by harnessing important interactions between the immune system and cancer. However, many of these therapies lead to frequent side effects when administered systemically, prompting treatment modifications or discontinuation or, in severe cases, fatalities. New therapeutic approaches like intratumoral immunotherapy, characterized by reduced side effects, cost, and systemic toxicity, offer promising prospects for future applications in clinical oncology. In the context of locally advanced or metastatic cancer, combining diverse immunotherapeutic and other treatment strategies targeting multiple cancer hallmarks appears crucial. Such combination therapies hold promise for improving patient outcomes and survival and for promoting a sustained systemic response. This review aims to provide a current overview of immunotherapeutic approaches, specifically focusing on the intratumoral administration of drugs in patients with locally advanced and metastatic cancers. It also explores the integration of intratumoral administration with other modalities to maximize therapeutic response. Additionally, the review summarizes recent advances in intratumoral immunotherapy and discusses novel therapeutic approaches, outlining future directions in the field.
Collapse
Affiliation(s)
- Marketa Skalickova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Katerina Hadrava Vanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Jindriska Leischner Fialova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Katerina Petrlakova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Michal Masarik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Pavel Martasek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
15
|
Gallego RA, Cho-Schultz S, Del Bel M, Dechert-Schmitt AM, Donaldson JS, He M, Jalaie M, Kania R, Matthews J, McTigue M, Tuttle JB, Risley H, Zhou D, Zhou R, Ahmad OK, Bernier L, Berritt S, Braganza J, Chen Z, Cianfrogna JA, Collins M, Costa Jones C, Cronin CN, Davis C, Dress K, Edwards M, Farrell W, France SP, Grable N, Johnson E, Johnson TW, Jones R, Knauber T, Lafontaine J, Loach RP, Maestre M, Miller N, Moen M, Monfette S, Morse P, Nager AR, Niosi M, Richardson P, Rohner AK, Sach NW, Timofeevski S, Tucker JW, Vetelino B, Zhang L, Nair SK. Discovery of PF-07265028, A Selective Small Molecule Inhibitor of Hematopoietic Progenitor Kinase 1 (HPK1) for the Treatment of Cancer. J Med Chem 2024; 67:22002-22038. [PMID: 39651809 DOI: 10.1021/acs.jmedchem.4c01930] [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: 12/11/2024]
Abstract
Hematopoietic progenitor kinase 1 (HPK1/MAP4K1) represents a high interest target for the treatment of cancer through an immune-mediated mechanism. Herein we present highlights of the drug discovery campaign within the lactam/azalactam series of inhibitors that yielded a small molecule (21, PF-07265028), which was advanced to a phase 1 clinical trial (NCT05233436). Key components of the discovery effort included optimization of potency through mitigation of ligand strain as guided by the use of cocrystal structures, mitigation of ADME liabilities (plasma instability and fraction metabolism by CYP2D6), and optimization of kinase selectivity, particularly over immune-modulating kinases with high homology to HPK1. Structure-based drug design via leveraging cocrystal structures and lipophilic efficiency analysis proved to be valuable tools that ultimately enabled the delivery of a clinical-quality small molecule inhibitor of HPK1.
Collapse
Affiliation(s)
- Rebecca A Gallego
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Sujin Cho-Schultz
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Matthew Del Bel
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | | | - Joyann S Donaldson
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Mingying He
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Mehran Jalaie
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Rob Kania
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Jean Matthews
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Michele McTigue
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Jamison B Tuttle
- Worldwide Research and Development, Pfizer, Inc., Cambridge, Massachusetts 02139, United States
| | - Hud Risley
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Dahui Zhou
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Ru Zhou
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Omar K Ahmad
- Worldwide Research and Development, Pfizer, Inc., Cambridge, Massachusetts 02139, United States
| | - Louise Bernier
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Simon Berritt
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - John Braganza
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Zecheng Chen
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Julie A Cianfrogna
- Pharmacokinetics, Dynamics and Metabolism Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Michael Collins
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Cinthia Costa Jones
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Ciaran N Cronin
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Carl Davis
- Pharmacokinetics, Dynamics and Metabolism Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Klaus Dress
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Martin Edwards
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - William Farrell
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Scott P France
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Nicole Grable
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Eric Johnson
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Ted W Johnson
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Rhys Jones
- Pharmacokinetics, Dynamics and Metabolism Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Thomas Knauber
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Jennifer Lafontaine
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Richard P Loach
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Michael Maestre
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Nichol Miller
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Mark Moen
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Sebastien Monfette
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Peter Morse
- Pharmacokinetics, Dynamics and Metabolism Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Andrew Ross Nager
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Mark Niosi
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Paul Richardson
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Allison K Rohner
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Neal W Sach
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Sergei Timofeevski
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Joseph W Tucker
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Beth Vetelino
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Lei Zhang
- Worldwide Research and Development, Pfizer, Inc., Cambridge, Massachusetts 02139, United States
| | - Sajiv K Nair
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| |
Collapse
|
16
|
Levi J, Das M, Vasanawala MS, Behl D, Pomper M, Forde PM, Nakajima E, Sayre J, Shen B, Cabrera H, Del Mar N, Gullen M, Pierini M, Cox L, Lokre O, Perk T, Chae HD. [ 18F]F-AraG Uptake in Vertebral Bone Marrow May Predict Survival in Patients with Non-Small Cell Lung Cancer Treated with Anti-PD-(L)1 Immunotherapy. J Nucl Med 2024; 65:1869-1875. [PMID: 39448270 PMCID: PMC11619592 DOI: 10.2967/jnumed.124.268253] [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: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Despite the systemic impact of both cancer and the associated immune response, immuno-PET is predominantly centered on assessment of the immune milieu within the tumor microenvironment. The aim of this study was to assess the value of [18F]F-AraG PET imaging as a noninvasive method for evaluation of system-wide immune status of patients with non-small cell lung cancer before starting immunotherapy. Methods: Eleven patients with advanced non-small cell lung cancer were imaged with [18F]F-AraG before starting immunotherapy. Diagnostic [18F]FDG PET/CT scans were analyzed to assess differences in the extent of disease among patients. SUVmax, SUVmean, and total SUV (SUVtotal) from all tumor lesions, active lymph nodes, spleen, vertebral bone marrow, liver, thyroid, heart, and bowel were extracted from the baseline [18F]F-AraG scans, and discriminant and Kaplan-Meier analyses were performed to test their ability to predict patient response and overall survival. Results: The extent of the disease was variable in the patient cohort, but none of the [18F]FDG biomarkers associated with tumor burden (SUVmax, total metabolic tumor volume, and total lesion glycolysis) was predictive of patient survival. The differences in the [18F]F-AraG and [18F]FDG distribution were observed both within and between lesions, confirming that they capture distinct aspects of the tumor microenvironment. Of the 3 SUV parameters studied, [18F]F-AraG SUVtotal provided a dynamic range suitable for stratifying tumors or patients according to their immune activity. [18F]F-AraG SUVtotal measured in the lumbar and sacral vertebrae differentiated between patients who progressed on therapy and those who did not with 90.9% and 81.8% accuracy, respectively. The Kaplan-Meier analysis revealed that patients with high [18F]F-AraG SUVtotal in the lumbar bone marrow had significantly lower probability of survival than those with a low signal (P = 0.0003). Conclusion: This study highlights the significance of assessing systemic immunity and indicates the potential of the [18F]F-AraG bone marrow signal as a predictive imaging biomarker for patient stratification and treatment guidance.
Collapse
Affiliation(s)
- Jelena Levi
- CellSight Technologies Inc., San Francisco, California;
| | - Millie Das
- Department of Medicine, Stanford University, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Minal S Vasanawala
- Department of Medicine, Stanford University, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Deepti Behl
- Sutter Medical Center, Sacramento, California
| | - Martin Pomper
- Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland
| | | | | | - James Sayre
- Department of Radiology, David Geffen School of Medicine at UCLA, UCLA Center for the Health Sciences, Los Angeles, California
| | - Bin Shen
- Department of Radiology, Stanford University, Palo Alto, California; and
| | - Hilda Cabrera
- CellSight Technologies Inc., San Francisco, California
| | - Niko Del Mar
- Department of Medicine, Stanford University, Palo Alto, California
| | | | | | - Laura Cox
- Sutter Medical Center, Sacramento, California
| | | | | | - Hee-Don Chae
- CellSight Technologies Inc., San Francisco, California
| |
Collapse
|
17
|
Devaraja K, Singh M, Sharan K, Aggarwal S. Coley's Toxin to First Approved Therapeutic Vaccine-A Brief Historical Account in the Progression of Immunobiology-Based Cancer Treatment. Biomedicines 2024; 12:2746. [PMID: 39767654 PMCID: PMC11726767 DOI: 10.3390/biomedicines12122746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Cancer immunobiology is one of the hot topics of discussion amongst researchers today, and immunotherapeutic modalities are among the selected few emerging approaches to cancer treatment that have exhibited a promising outlook. However, immunotherapy is not a new kid on the block; it has been around for centuries. The origin of cancer immunotherapy in modern medicine can be traced back to the initial reports of spontaneous regression of malignant tumors in some patients following an acute febrile infection, at the turn of the twentieth century. This review briefly revisits the historical accounts of immunotherapy, highlighting some of the significant developments in the field of cancer immunobiology, that have been instrumental in bringing back the immunotherapeutic approaches to the forefront of cancer research. Some of the topics covered are: Coley's toxin-the first immunotherapeutic; the genesis of the theory of immune surveillance; the discovery of T lymphocytes and dendritic cells and their roles; the role of tumor antigens; relevance of tumor microenvironment; the anti-tumor (therapeutic) ability of Bacillus Calmette- Guérin; Melacine-the first therapeutic vaccine engineered; theories of immunoediting and immunophenotyping of cancer; and Provenge-the first FDA-approved therapeutic vaccine. In this review, head and neck cancer has been taken as the reference tumor for narrating the progression of cancer immunobiology, particularly for highlighting the advent of immunotherapeutic agents.
Collapse
Affiliation(s)
- K. Devaraja
- Department of Head and Neck Surgery, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Manisha Singh
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Krishna Sharan
- Department of Radiation Oncology, K S Hegde Medical College, Nitte University, Mangalore 574110, India;
| | - Sadhna Aggarwal
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
18
|
Diop MP, van der Stegen SJC. The Pluripotent Path to Immunotherapy. Exp Hematol 2024; 139:104648. [PMID: 39251182 DOI: 10.1016/j.exphem.2024.104648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/14/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Adoptive cell therapy (ACT) enhances the patient's own immune cells' ability to identify and eliminate cancer cells. Several immune cell types are currently being applied in autologous ACT, including T cells, natural killer (NK) cells, and macrophages. The cells' inherent antitumor capacity can be used, or they can be targeted toward tumor-associated antigen through expression of a chimeric antigen receptor (CAR). Although CAR-based ACT has achieved great results in hematologic malignancies, the accessibility of ACT is limited by the autologous nature of the therapy. Induced pluripotent stem cells (iPSCs) hold the potential to address this challenge, because they can provide an unlimited source for the in vitro generation of immune cells. Various immune subsets have been generated from iPSC for application in ACT, including several T-cell subsets (αβT cells, mucosal-associated invariant T cells, invariant NKT [iNKT] cells, and γδT cells), as well as NK cells, macrophages, and neutrophils. iPSC-derived αβT, NK, and iNKT cells are currently being tested in phase I clinical trials. The ability to perform (multiplexed) gene editing at the iPSC level and subsequent differentiation into effector populations not only expands the arsenal of ACT but allows for development of ACT utilizing cell types which cannot be efficiently obtained from peripheral blood or engineered and expanded in vitro.
Collapse
Affiliation(s)
- Mame P Diop
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
19
|
Chirullo B, Fruscione F, Del Zotto G, Dell'Anno F, Tarantino M, Porcellato I, Petrucci P, De Ciucis CG, Bufalari A, Guardone L, Cappelli K, Moretti G, Mecocci S, Monti E, De Paolis L, Razzuoli E. Evaluation of attenuated Salmonella Typhimurium (STMΔznuABC) anticancer activity on canine mammary cancer-associated fibroblasts. Res Vet Sci 2024; 180:105438. [PMID: 39447298 DOI: 10.1016/j.rvsc.2024.105438] [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: 06/10/2024] [Revised: 09/23/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
Bacteria-mediated treatments gained increasing attention as alternative therapies against tumors. An attenuated mutant strain of Salmonella enterica serovar Typhimurium (STMΔznuABC) has recently been considered as a potential new anti-cancer strategy. However, it is unclear whether this activity is tumor-induced or species-specific, and no data are available regarding STMΔznuABC on canine mammary tumors (CMTs). This study aimed to investigate the ability of STMΔznuABC in modulating the response of CMTs, focusing on cancer-associated fibroblasts. Four CMT cell lines (CF33, TM51, TM52 TM53) were treated with STMΔznuABC. Then, antiproliferative activity (MTT assay), bacterial invasion, and CMT cell lines gene expression analysis (RT-qPCR) of genes involved in immune response and cancer aggressiveness were evaluated. STMΔznuABC penetrated in TM51, TM52, TM53, and CF33 cell lines, causing a significant reduction of cell viability. Moreover, the expression of several genes was significantly modulated in all CMT cell lines: STMΔznuABC infection determined a significant up-regulation of CXCL8, IL18, IL10, TLR4 and RAD51, while CD14, IL6, CXCR4, P53, PTEN, STAT5, TLR5 and TGFB1 were downregulated in TM53. In CF33, CXCL8 and P53 were upregulated, while MYD88, MD2, IL18, TLR4,5, TGFB1 were downregulated. In TM52, CXCL8, CD44 and MD2 were upregulated and PTEN was downregulated, while in TM51 CXCL8, CD44 and ErbB2 were downregulated. We demonstrated the anti-proliferative and immuno-modulatory activity of STMΔznuABC in CMTs, paving the way for potential new anti-cancer treatments.
Collapse
Affiliation(s)
- Barbara Chirullo
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Floriana Fruscione
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| | | | - Filippo Dell'Anno
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| | - Michela Tarantino
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Ilaria Porcellato
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Paola Petrucci
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy.
| | - Antonello Bufalari
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Lisa Guardone
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy.
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Giulia Moretti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Eleonora Monti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Livia De Paolis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| |
Collapse
|
20
|
Omole AO, Zhao Z, Chang-Liao S, de Oliveira JFA, Boone CE, Sutorus L, Sack M, Varner J, Fiering SN, Steinmetz NF. Virus nanotechnology for intratumoural immunotherapy. NATURE REVIEWS BIOENGINEERING 2024; 2:916-929. [PMID: 39698315 PMCID: PMC11655125 DOI: 10.1038/s44222-024-00231-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 12/20/2024]
Abstract
Viruses can be designed to be tools and carrier vehicles for intratumoural immunotherapy. Their nanometre-scale size and shape allow for functionalization with or encapsulation of medical cargoes and tissue-specific ligands. Importantly, immunotherapies may particularly benefit from the inherent immunomodulatory properties of viruses. For example, mammalian viruses have already been tested for oncolytic virotherapy, and bacteriophages and plant viruses can be engineered for immunotherapeutic treatment approaches. In this Review, we discuss how viruses - including oncolytic viruses, immunomodulatory plant viruses and bacteriophages - and virus-like particles can be designed for intratumoural immunotherapy to elicit anti-tumour immunity and induce systemic anti-tumour responses at distant non-injected sites. We further highlight the engineering of viruses and virus-like particles as drug-delivery systems, and outline key translational challenges and clinical opportunities.
Collapse
Affiliation(s)
- Anthony O. Omole
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Zhongchao Zhao
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Sabrina Chang-Liao
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jessica Fernanda Affonso de Oliveira
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Christine E. Boone
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Lucas Sutorus
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Judith Varner
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Center for Engineering in Cancer, Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Steven N. Fiering
- Department of Microbiology and Immunology, Dartmouth Cancer Center, Dartmouth Geisel School of Medicine and Dartmouth-Hitchock Health, Lebanon, NH, USA
| | - Nicole F. Steinmetz
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
- Shu and K. C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Center for Engineering in Cancer, Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
21
|
Huang J, Xiong L, Tang S, Zhao J, Zuo L. Balancing Tumor Immunotherapy and Immune-Related Adverse Events: Unveiling the Key Regulators. Int J Mol Sci 2024; 25:10919. [PMID: 39456702 PMCID: PMC11507008 DOI: 10.3390/ijms252010919] [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: 08/28/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Tumor immunotherapy has emerged as a promising approach in cancer treatment in recent years, offering vast potential. This method primarily involves targeting and inhibiting the suppressive checkpoints present in different immune cells to enhance their activation, ultimately leading to tumor regression. However, tumor cells exploit the surrounding immune cells and tissues to establish a tumor microenvironment (TME) that supports their survival and growth. Within the TME, the efficacy of effector immune cells is compromised, as tumor cells exploit inhibitory immune cells to suppress their function. Furthermore, certain immune cells can be co-opted by tumor cells to facilitate tumor growth. While significantly enhancing the body's tumor immunity can lead to tumor regression, it can also result in severe toxic side effects and an inflammatory factor storm. As a consequence, patients often discontinue treatment due to immune-related adverse events (irAEs) or, in extreme cases, succumb to toxic side effects before experiencing tumor regression. In this analysis, we examined several remission regimens for irAEs, each with its own drawbacks, including toxic side effects or suppression of tumor immunotherapy, which is undesirable. A recent research study, specifically aimed at downregulating intestinal epithelial barrier permeability, has shown promising results in reducing the severity of inflammatory bowel disease (IBD) while preserving immune function. This approach effectively reduces the severity of IBD without compromising the levels of TNF-α and IFN-γ, which are crucial for maintaining the efficacy of tumor immunotherapy. Based on the substantial similarities between IBD and ICI colitis (combo immune checkpoint inhibitors-induced colitis), this review proposes that targeting epithelial cells represents a crucial research direction for mitigating irAEs in the future.
Collapse
Affiliation(s)
- Jianshang Huang
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Lei Xiong
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Sainan Tang
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Junhao Zhao
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Li Zuo
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| |
Collapse
|
22
|
Iwai Y, Baldwin XL, Feeney T, Agala CB, Yanagihara TK, Stein JN, Kim HJ, Spanheimer PM. Trends in the use of immunotherapy to treat soft tissue sarcoma. Am J Surg 2024; 236:115794. [PMID: 38879356 PMCID: PMC11392640 DOI: 10.1016/j.amjsurg.2024.115794] [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: 04/29/2024] [Accepted: 06/04/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND The role of immune-oncology (IO) therapy in soft tissue sarcoma (STS) is underexplored. This study characterized IO use in STS. METHODS This is a retrospective analysis of patients with a soft tissue mass in the National Cancer Database, 2011-2021. Patients were categorized by IO receipt status. Groupwise testing and proportional trend tests were performed with Chi-squared tests. Multivariate logistic regression was performed to assess factors associated with IO receipt. RESULTS Of the 103,092 patients with STS, 1935 (1.9 %) received or were recommended IO therapy. IO use increased 10-fold (0.24 %-2.5 % from 2011 to 2021; p < 0.0001). Patients had higher odds of receiving IO when having higher grade tumors and metastatic disease, and when treated at an academic research center (all p < 0.001). CONCLUSIONS IO use in STS is low but increasing and primarily used in the metastatic setting. Future studies should identify biomarkers of IO response and facilitators for treatment receipt.
Collapse
Affiliation(s)
- Yoshiko Iwai
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xavier L Baldwin
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Timothy Feeney
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chris B Agala
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ted K Yanagihara
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jacob N Stein
- Department of Medical Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hong Jin Kim
- Division of Surgical Oncology, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Philip M Spanheimer
- Division of Surgical Oncology, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
23
|
Spalato-Ceruso M, Ghazzi NE, Italiano A. New strategies in soft tissue sarcoma treatment. J Hematol Oncol 2024; 17:76. [PMID: 39218932 PMCID: PMC11368005 DOI: 10.1186/s13045-024-01580-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Soft tissue sarcomas (STS) have long been a formidable challenge in oncology, partly because of their rarity and diversity, which complicates large-scale studies and slows the advent of new treatments. Traditionally anchored by anthracycline-based chemotherapy, the landscape of STS treatment hasn't shifted dramatically in the past twenty years. However, recent strides in research are starting to paint a more hopeful picture. Leveraging advanced molecular profiling, researchers are now tailoring treatments to the unique genetic makeup of tumors, with targeted therapies showing promise. Innovations such as NTRK inhibitors for NTRK-rearranged sarcomas and gamma-secretase inhibitors for desmoid tumors are changing clinical practices. The rise of immunotherapy, including novel agents like LAG-3 inhibitors and bifunctional proteins that target both TGF-β and PD-L1, offers new avenues for treatment, particularly when combined with traditional therapies like chemotherapy. Meanwhile, the approval of epigenetic treatments for specific sarcoma subtypes heralds a new wave of strategy based on histological specificity, which could lead to more personalized and effective care. While challenges remain, the field of STS treatment is evolving, driven by a deeper understanding of the disease's biological underpinnings and a commitment to innovative research approaches.
Collapse
Affiliation(s)
| | - Nathan El Ghazzi
- Sarcoma Unit, Institut Bergonié, Bordeaux, France
- INSERM U1312, Bordeaux, France
| | - Antoine Italiano
- Sarcoma Unit, Institut Bergonié, Bordeaux, France.
- INSERM U1312, Bordeaux, France.
- Université de Bordeaux, Bordeaux, France.
| |
Collapse
|
24
|
Munir M, Cheema AY, Ogedegbe OJ, Aslam MF, Kim S. William Coley: The Pioneer and the Father of Immunotherapy. Cureus 2024; 16:e69113. [PMID: 39391466 PMCID: PMC11466495 DOI: 10.7759/cureus.69113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
William Coley was an unacclaimed hero of early cancer treatment. His work is often overshadowed by more recent advancements in immunotherapy. Coley's innovative work in the 1910s and 1930s laid the groundwork for what would become a major field in oncology. His experiments with bacterial vaccines by making use of the immune system to combat cancer preceded contemporary immunotherapy for several decades. This review provides a comprehensive exploration of Coley's life, his groundbreaking research, the socio-scientific challenges he faced, and his lasting impact on cancer treatment. Even though he faced lots of initial resistance and challenges, Coley's work has influenced modern immunotherapy practices.
Collapse
Affiliation(s)
- Mishaal Munir
- Medicine, Ghurki Trust and Teaching Hospital, Lahore, PAK
- Internal Medicine, Lahore Medical & Dental College, Lahore, PAK
| | - Asfand Yar Cheema
- Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, USA
- Medicine, Services Hospital, Lahore, PAK
| | | | - Muhammad Faisal Aslam
- Internal Medicine, Icahn School of Medicine at Mount Sinai, Queens Hospital Center, New York, USA
| | - San Kim
- Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, USA
| |
Collapse
|
25
|
Kohli AS, Sanyal S, Kaushal RS, Dwivedi M. An Insight into Immunological Therapeutic Approach against Cancer: Potential Anti-cancer Vaccines. Curr Genomics 2024; 26:175-190. [PMID: 40433416 PMCID: PMC12105320 DOI: 10.2174/0113892029319505240821063238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 05/29/2025] Open
Abstract
The development of a cancer vaccine comes with its complications and designing and developing a vaccine against foreign invaders such as bacterial and viral particles is not as complex and multi-faceted as the preparation of immunotherapy for host-infected cells which resemble our own body cells. The entire research and development framework of designing a vaccine for cancerous cells lies entirely on the remarkable aspect of notifying specific interactions and acclimatising the immune system. This review aims to compile the several fronts research-based methodology applies to in terms of developing a therapeutic, preventive or personalised vaccine for cancer. The approach lays focus on the identification and selection of targets for vaccine development which have come to light as immune biomarkers. Furthemore, significant aspects of personalised and precision vaccines and the fine line that runs between these approaches have also been discussed.
Collapse
Affiliation(s)
- Arjun Singh Kohli
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomtinagar Ext., Lucknow, 226028, India
| | - Somali Sanyal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomtinagar Ext., Lucknow, 226028, India
| | | | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomtinagar Ext., Lucknow, 226028, India
- Research Cell, Amity University Uttar Pradesh, Lucknow Campus, India
| |
Collapse
|
26
|
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
|
27
|
Radford GA, Vrbanac L, de Nys RT, Worthley DL, Wright JA, Hasty J, Woods SL. Towards Understanding Tumour Colonisation by Probiotic Bacterium E. coli Nissle 1917. Cancers (Basel) 2024; 16:2971. [PMID: 39272829 PMCID: PMC11394440 DOI: 10.3390/cancers16172971] [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: 08/06/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The last decade has seen a rapid increase in studies utilising a genetically modified probiotic, Escherichia coli Nissle 1917 (EcN), as a chassis for cancer treatment and detection. This approach relies on the ability of EcN to home to and selectively colonise tumours over normal tissue, a characteristic common to some bacteria that is thought to result from the low-oxygen, nutrient-rich and immune-privileged niche the tumour provides. Pre-clinical studies have used genetically modified EcN to deliver therapeutic payloads that show efficacy in reducing tumour burden as a result of high-tumour and low off-target colonisation. Most recently, the EcN chassis has been expanded into an effective tumour-detection tool. These advances provide strong justification for the movement of genetically modified EcN into clinical oncology trials. What is currently unknown in the field is a deep mechanistic understanding of how EcN distributes to and localises within tumours. This review summarises the existing EcN literature, with the inclusion of research undertaken with other tumour-homing and pathogenic bacteria, to provide insights into possible mechanisms of EcN tumour homing for future validation. Understanding exactly how and why EcN colonises neoplastic tissue will inform the design and testing of the next generation of EcN chassis strains to address biosafety and containment concerns and optimise the detection and treatment of cancer.
Collapse
Affiliation(s)
| | - Laura Vrbanac
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Rebekah T. de Nys
- Precision Cancer Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | | | - Josephine A. Wright
- Precision Cancer Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | - Jeff Hasty
- Synthetic Biology Institute, University of California, San Diego, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, CA 92093, USA
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, CA 92093, USA
- Center for Microbiome Innovation, University of California, San Diego, CA 92093, USA
| | - Susan L. Woods
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| |
Collapse
|
28
|
Xiong Y, Lo Y, Song H, Lu J. Development of a Self-Luminescent Living Bioreactor for Enhancing Photodynamic Therapy in Breast Cancer. Bioconjug Chem 2024; 35:1269-1282. [PMID: 39120495 DOI: 10.1021/acs.bioconjchem.4c00334] [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: 08/10/2024]
Abstract
The penetration ability of visible light (<2 mm) and near-infrared (NIR) light (∼1 cm) remarkably impairs the therapeutic efficacy and clinical applications of photodynamic therapy (PDT). To address the limitation of light penetration depth, a novel self-luminescent bacterium, teLuc.FP-EcN, has been engineered through transfection of a fusion expression plasmid containing the luciferase gene teLuc and bright red fluorescent protein mScarlet-I into Escherichia coli Nissle 1917 (EcN). The engineered teLuc.FP-EcN can specifically target and colonize tumors without significant toxicity to the host. Acting as a continuous internal light source, teLuc.FP-EcN can activate the photosensitizer chlorin e6 (Ce6) to generate reactive oxygen species (ROS) and then effectively destroy tumor tissue from the inside. As a result, a significant reduction in tumor proliferation and extension of the overall survival in mouse tumor models has been observed. Furthermore, teLuc.FP-EcN-boosted PDT amplified its therapeutic effect by activating antitumor immune response, including the conversion of M2 macrophages into pro-inflammatory M1 macrophages, as well as an increase in the proportion of CD3+ T cells and a decrease in T-cell exhaustion. In conclusion, teLuc.FP-EcN can be used as an implantable light source for tumor phototherapy, which simultaneously possesses ROS generation and immune regulation.
Collapse
Affiliation(s)
- Yanian Xiong
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yingtung Lo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Huizhu Song
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianzhong Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
29
|
Mundhara N, Sadhukhan P. Cracking the Codes behind Cancer Cells' Immune Evasion. Int J Mol Sci 2024; 25:8899. [PMID: 39201585 PMCID: PMC11354234 DOI: 10.3390/ijms25168899] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Immune evasion is a key phenomenon in understanding tumor recurrence, metastasis, and other critical steps in tumor progression. The tumor microenvironment (TME) is in constant flux due to the tumor's ability to release signals that affect it, while immune cells within it can impact cancer cell behavior. Cancer cells undergo several changes, which can change the enrichment of different immune cells and modulate the activity of existing immune cells in the tumor microenvironment. Cancer cells can evade immune surveillance by downregulating antigen presentation or expressing immune checkpoint molecules. High levels of tumor-infiltrating lymphocytes (TILs) correlate with better outcomes, and robust immune responses can control tumor growth. On the contrary, increased enrichment of Tregs, myeloid-derived suppressor cells, and M2-like anti-inflammatory macrophages can hinder effective immune surveillance and predict poor prognosis. Overall, understanding these immune evasion mechanisms guides therapeutic strategies. Researchers aim to modulate the TME to enhance immune surveillance and improve patient outcomes. In this review article, we strive to summarize the composition of the tumor immune microenvironment, factors affecting the tumor immune microenvironment (TIME), and different therapeutic modalities targeting the immune cells. This review is a first-hand reference to understand the basics of immune surveillance and immune evasion.
Collapse
Affiliation(s)
| | - Pritam Sadhukhan
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
30
|
Sui C, Wu H, Li X, Wang Y, Wei J, Yu J, Wu X. Cancer immunotherapy and its facilitation by nanomedicine. Biomark Res 2024; 12:77. [PMID: 39097732 PMCID: PMC11297660 DOI: 10.1186/s40364-024-00625-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024] Open
Abstract
Cancer immunotherapy has sparked a wave of cancer research, driven by recent successful proof-of-concept clinical trials. However, barriers are emerging during its rapid development, including broad adverse effects, a lack of reliable biomarkers, tumor relapses, and drug resistance. Integration of nanomedicine may ameliorate current cancer immunotherapy. Ultra-large surface-to-volume ratio, extremely small size, and easy modification surface of nanoparticles enable them to selectively detect cells and kill cancer cells in vivo. Exciting synergistic applications of the two approaches have emerged in treating various cancers at the intersection of cancer immunotherapy and cancer nanomedicine, indicating the potential that the combination of these two therapeutic modalities can lead to new paradigms in the treatment of cancer. This review discusses the status of current immunotherapy and explores the possible opportunities that the nanomedicine platform can make cancer immunotherapy more powerful and precise by synergizing the two approaches.
Collapse
Affiliation(s)
- Chao Sui
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 East Duarte, Los Angeles, CA, 91010, USA
| | - Heqing Wu
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xinxin Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an Shaanxi, 710072, China
| | - Yuhang Wang
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jiaqi Wei
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 East Duarte, Los Angeles, CA, 91010, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
| | - Xiaojin Wu
- The First Affiliated Hospital of Soochow University, Suzhou, China.
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
31
|
Martin J, Hans S, Ferris RL, Baudouin R. Over 132 Years Ago, the Inaugural Immunotherapy Treated Its First Head and Neck Cancer Patient. Otolaryngol Head Neck Surg 2024; 171:621-624. [PMID: 38587009 DOI: 10.1002/ohn.745] [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: 12/21/2023] [Accepted: 03/03/2024] [Indexed: 04/09/2024]
Abstract
Immunotherapy in oncology has a more extensive history than is commonly perceived. Rooted in the observations and experiences of multiple physicians in the late 19th century, immunological interventions are currently integral to the oncological therapeutic repertoire. This article seeks to delineate the evolution of cancer immunotherapy, tracing its inception in 1891 with the pioneering work of an American surgeon, William B. Coley, who achieved the first documented cure of a cancer case involving a malignant head and neck tumor. The narrative extends to encompass successive historical breakthroughs and prospective developments in this dynamic field.
Collapse
Affiliation(s)
- Joey Martin
- Department of Head and Neck Surgery, Institut Curie, Paris/Saint-Cloud, France
| | - Stéphane Hans
- Department of Otolaryngology-Head and Neck Surgery, Foch Hospital, Suresnes, France
- UFR of Health Sciences (UVSQ/Paris-Saclay University), Montigny-Le-Bretonneux, France
| | - Robert L Ferris
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robin Baudouin
- Department of Otolaryngology-Head and Neck Surgery, Foch Hospital, Suresnes, France
- UFR of Health Sciences (UVSQ/Paris-Saclay University), Montigny-Le-Bretonneux, France
- Laboratory Anthropology, Archaeology, Biology (LAAB), UFR of Health Sciences (UVSQ/Paris-Saclay University), Montigny-Le-Bretonneux, France
| |
Collapse
|
32
|
Kiaheyrati N, Babaei A, Ranji R, Bahadoran E, Taheri S, Farokhpour Z. Cancer therapy with the viral and bacterial pathogens: The past enemies can be considered the present allies. Life Sci 2024; 349:122734. [PMID: 38788973 DOI: 10.1016/j.lfs.2024.122734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024]
Abstract
Cancer continues to be one of the leading causes of mortality worldwide despite significant advancements in cancer treatment. Many difficulties have arisen as a result of the detrimental consequences of chemotherapy and radiotherapy as a common cancer therapy, such as drug inability to penetrate deep tumor tissue, and also the drug resistance in tumor cells continues to be a major concern. These obstacles have increased the need for the development of new techniques that are more selective and effective against cancer cells. Bacterial-based therapies and the use of oncolytic viruses can suppress cancer in comparison to other cancer medications. The tumor microenvironment is susceptible to bacterial accumulation and proliferation, which can trigger immune responses against the tumor. Oncolytic viruses (OVs) have also gained considerable attention in recent years because of their potential capability to selectively target and induce apoptosis in cancer cells. This review aims to provide a comprehensive summary of the latest literature on the role of bacteria and viruses in cancer treatment, discusses the limitations and challenges, outlines various strategies, summarizes recent preclinical and clinical trials, and emphasizes the importance of optimizing current strategies for better clinical outcomes.
Collapse
Affiliation(s)
- Niloofar Kiaheyrati
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Microbiology and Immunology, School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Abouzar Babaei
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Microbiology and Immunology, School of Medicine, Qazvin University of Medical Science, Qazvin, Iran.
| | - Reza Ranji
- Department of Genetics, Faculty of Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ensiyeh Bahadoran
- School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Shiva Taheri
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Zahra Farokhpour
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
33
|
Sanaei MJ, Pourbagheri-Sigaroodi A, Rezvani A, Zaboli E, Salari S, Masjedi MR, Bashash D. Lung cancer vaccination from concept to reality: A critical review of clinical trials and latest advances. Life Sci 2024; 346:122652. [PMID: 38641048 DOI: 10.1016/j.lfs.2024.122652] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Lung cancer is a highly lethal malignancy that poses a significant burden on public health worldwide. There have been numerous therapeutic approaches, among which cancer vaccines have emerged as a promising approach to harnessing the patient's immune system to induce long-lasting anti-tumor immunity. The current study aims to provide an overview of cancer vaccination in the context of lung cancer to establish a clearer landscape for lung cancer treatment. To provide a comprehensive review, we not only gathered the published studies of lung cancer vaccination and discussed their effectiveness and safety profile but also analyzed all the relevant clinical trials registered on www.clinicaltrials.gov until March 2024. We demonstrated all utilized vaccine platforms along with having a glance at novel technologies such as mRNA vaccines. The present review discussed the challenges and shortcomings of lung cancer vaccination, as well as the way they could be managed to pave the way for reaching the most optimized vaccine formulation.
Collapse
Affiliation(s)
- Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Rezvani
- Department of Internal Medicine, Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ehsan Zaboli
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sina Salari
- Department of Medical Oncology-Hematology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Masjedi
- Cancer Control Research Center, Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran; Department of Pulmonary Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Recent Insights into the Molecular Mechanisms of the Toll-like Receptor Response to Influenza Virus Infection. Int J Mol Sci 2024; 25:5909. [PMID: 38892096 PMCID: PMC11172706 DOI: 10.3390/ijms25115909] [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: 04/21/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Influenza A viruses (IAVs) pose a significant global threat to human health. A tightly controlled host immune response is critical to avoid any detrimental effects of IAV infection. It is critical to investigate the association between the response of Toll-like receptors (TLRs) and influenza virus. Because TLRs may act as a double-edged sword, a balanced TLR response is critical for the overall benefit of the host. Consequently, a thorough understanding of the TLR response is essential for targeting TLRs as a novel therapeutic and prophylactic intervention. To date, a limited number of studies have assessed TLR and IAV interactions. Therefore, further research on TLR interactions in IAV infection should be conducted to determine their role in host-virus interactions in disease causation or clearance of the virus. Although influenza virus vaccines are available, they have limited efficacy, which should be enhanced to improve their efficacy. In this study, we discuss the current status of our understanding of the TLR response in IAV infection and the strategies adopted by IAVs to avoid TLR-mediated immune surveillance, which may help in devising new therapeutic or preventive strategies. Furthermore, recent advances in the use of TLR agonists as vaccine adjuvants to enhance influenza vaccine efficacy are discussed.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
35
|
Fang X, Tong W, Wu S, Zhu Z, Zhu J. The role of intratumoral microorganisms in the progression and immunotherapeutic efficacy of head and neck cancer. ONCOLOGIE 2024; 26:349-360. [DOI: 10.1515/oncologie-2023-0511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Abstract
The effectiveness of cancer immunization is largely dependent on the tumor’s microenvironment, especially the tumor immune microenvironment. Emerging studies say microbes exist in tumor cells and immune cells, suggesting that these microbes can affect the state of the immune microenvironment of the tumor. Our comprehensive review navigates the intricate nexus between intratumoral microorganisms and their role in tumor biology and immune modulation. Beginning with an exploration of the historical acknowledgment of microorganisms within tumors, the article underscores the evolution of the tumor microenvironment (TME) and its subsequent implications. Using findings from recent studies, we delve into the unique bacterial compositions across different tumor types and their influence on tumor growth, DNA damage, and immune regulation. Furthermore, we illuminate the potential therapeutic implications of targeting these intratumoral microorganisms, emphasizing their multifaceted roles from drug delivery agents to immunotherapy enhancers. As advancements in next-generation sequencing (NGS) technology redefine our understanding of the tumor microbiome, the article underscores the importance of discerning their precise role in tumor progression and tailoring therapeutic interventions. The review culminates by emphasizing ongoing challenges and the pressing need for further research to harness the potential of intratumoral microorganisms in cancer care.
Collapse
Affiliation(s)
- Xuzhe Fang
- The Fourth School of Clinical Medicine , Zhejiang Chinese Medical University , Hangzhou , China
| | - Weihong Tong
- The Fourth School of Clinical Medicine , Zhejiang Chinese Medical University , Hangzhou , China
| | - Sheng Wu
- The Fourth School of Clinical Medicine , Zhejiang Chinese Medical University , Hangzhou , China
| | - Zhengyong Zhu
- The Fourth School of Clinical Medicine , Zhejiang Chinese Medical University , Hangzhou , China
| | - Jin Zhu
- Department of Otorhinolaryngology and Head Neck Surgery, Affiliated Hangzhou First People’s Hospital , Zhejiang University School of Medicine , Hangzhou , China
| |
Collapse
|
36
|
Wu LY, Park SH, Jakobsson H, Shackleton M, Möller A. Immune Regulation and Immune Therapy in Melanoma: Review with Emphasis on CD155 Signalling. Cancers (Basel) 2024; 16:1950. [PMID: 38893071 PMCID: PMC11171058 DOI: 10.3390/cancers16111950] [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: 03/28/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma is commonly diagnosed in a younger population than most other solid malignancies and, in Australia and most of the world, is the leading cause of skin-cancer-related death. Melanoma is a cancer type with high immunogenicity; thus, immunotherapies are used as first-line treatment for advanced melanoma patients. Although immunotherapies are working well, not all the patients are benefitting from them. A lack of a comprehensive understanding of immune regulation in the melanoma tumour microenvironment is a major challenge of patient stratification. Overexpression of CD155 has been reported as a key factor in melanoma immune regulation for the development of therapy resistance. A more thorough understanding of the actions of current immunotherapy strategies, their effects on immune cell subsets, and the roles that CD155 plays are essential for a rational design of novel targets of anti-cancer immunotherapies. In this review, we comprehensively discuss current anti-melanoma immunotherapy strategies and the immune response contribution of different cell lineages, including tumour endothelial cells, myeloid-derived suppressor cells, cytotoxic T cells, cancer-associated fibroblast, and nature killer cells. Finally, we explore the impact of CD155 and its receptors DNAM-1, TIGIT, and CD96 on immune cells, especially in the context of the melanoma tumour microenvironment and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Li-Ying Wu
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Su-Ho Park
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haakan Jakobsson
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
| | - Mark Shackleton
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
- School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Andreas Möller
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
37
|
Wang Y, Yan F. Sono-activatable engineered bacteria for antitumor therapy. Cell Rep Med 2024; 5:101562. [PMID: 38776871 PMCID: PMC11148798 DOI: 10.1016/j.xcrm.2024.101562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
Bacteria show promising potential in tumor treatment, but safety concerns limit their application. In this issue, Gao et al.1 develop ultrasound-controlled engineered bacteria through integrating sono-activatable gene circuits, achieving local production and release of therapeutic cargos in tumor.
Collapse
Affiliation(s)
- Yuanyuan Wang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
38
|
Xia X, Zhang JW, Zhao B, Zhang M, Chen ZR, Zhang BF, Ji YL, Wang X, Xiong WM, Li JW, Lv QL. Progress of engineered bacteria for tumour therapy. Int Immunopharmacol 2024; 132:111935. [PMID: 38599096 DOI: 10.1016/j.intimp.2024.111935] [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: 12/09/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/12/2024]
Abstract
Finding novel therapeutic modalities, improving drug delivery efficiency and targeting, and reducing the immune escape of tumor cells are currently hot topics in the field of tumor therapy. Bacterial therapeutics have proven highly effective in preventing tumor spread and recurrence, used alone or in combination with traditional therapies. In recent years, a growing number of researchers have significantly improved the targeting and penetration of bacteria by using genetic engineering technology, which has received widespread attention in the field of tumor therapy. In this paper, we provide an overview and assessment of the advancements made in the field of tumor therapy using genetically engineered bacteria. We cover three major aspects: the development of engineered bacteria, their integration with other therapeutic techniques, and the current state of clinical trials. Lastly, we discuss the limitations and challenges that are currently being faced in the utilization of engineered bacteria for tumor therapy.
Collapse
Affiliation(s)
- Xue Xia
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China; College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China
| | - Jing-Wen Zhang
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China; College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China
| | - Bing Zhao
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China; College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China
| | - Min Zhang
- Nanchang Inspection and Testing Center, Nanchang Key Laboratory for Quality and Safety Risk Assessment of Health Food and its Contact Materials, Nanchang 330012, PR China
| | - Zhang-Ren Chen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330000, PR China
| | - Bing-Feng Zhang
- College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China
| | - Yu-Long Ji
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China
| | - Xia Wang
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China
| | - Wen-Min Xiong
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China
| | - Jia-Wei Li
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, Jiangxi, PR China.
| | - Qiao-Li Lv
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China; College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China.
| |
Collapse
|
39
|
Sun A, Xing Z, Lv R, Niu P, Zhao B, Ma S, Li H. Research progress of immunotherapy for advanced head and neck cancer. Med Oncol 2024; 41:133. [PMID: 38703250 DOI: 10.1007/s12032-024-02375-9] [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: 02/01/2024] [Accepted: 03/28/2024] [Indexed: 05/06/2024]
Abstract
Head and neck cancer accounts for about one-fifth of all malignant tumors, and the incidence is increasing year by year. The overall mortality rate was high and the 5-year survival rate was low. At present, the combination of surgery, radiotherapy, and chemotherapy is the main treatment in clinical practice, but the treatment of recurrent or metastatic advanced head and neck cancer is still a challenge. With the rise of immunotherapy, more and more studies on immune checkpoint inhibitors have been conducted. This review summarizes the mechanism, clinical application and safety of immunotherapy for advanced head and neck cancer.
Collapse
Affiliation(s)
- Anchi Sun
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, 2600 Donghai Avenue, Bengbu, 233030, Anhui, China
| | - Zhiwei Xing
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, China
| | - Rongrong Lv
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, China
| | - Pengyuan Niu
- Department of Neurosurgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, China
| | - Bao Zhao
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230071, Anhui, China
| | - Shiyin Ma
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, China.
| | - Hui Li
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, China.
| |
Collapse
|
40
|
Wang C, Zhong L, Xu J, Zhuang Q, Gong F, Chen X, Tao H, Hu C, Huang F, Yang N, Li J, Zhao Q, Sun X, Huo Y, Chen Q, Zhao Y, Peng R, Liu Z. Oncolytic mineralized bacteria as potent locally administered immunotherapeutics. Nat Biomed Eng 2024; 8:561-578. [PMID: 38514774 DOI: 10.1038/s41551-024-01191-w] [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: 06/22/2023] [Accepted: 02/17/2024] [Indexed: 03/23/2024]
Abstract
Oncolytic bacteria can trigger innate immune activity. However, the antitumour efficacy of inactivated bacteria is poor, and attenuated live bacteria pose substantial safety risks. Here we show that intratumourally injected paraformaldehyde-fixed bacteria coated with manganese dioxide potently activate innate immune activity, modulate the immunosuppressive tumour microenvironment and trigger tumour-specific immune responses and abscopal antitumour responses. A single intratumoural administration of mineralized Salmonella typhimurium suppressed the growth of multiple types of subcutaneous and orthotopic tumours in mice, rabbits and tree shrews and protected the cured animals against tumour rechallenge. We also show that mineralized bacteria can be administered via arterial embolization to treat orthotopic liver cancer in rabbits. Our findings support the further translational testing of oncolytic mineralized bacteria as potent and safe antitumour immunotherapeutics.
Collapse
Affiliation(s)
- Chenya Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Liping Zhong
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Jiachen Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Zhuang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Fei Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Xiaojing Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Huiquan Tao
- InnoBM Pharmaceuticals Co. Ltd., Suzhou, China
| | - Cong Hu
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Fuquan Huang
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Junyan Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Qi Zhao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
- InnoBM Pharmaceuticals Co. Ltd., Suzhou, China
| | - Xinjun Sun
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Yu Huo
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Yongxiang Zhao
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China.
| | - Rui Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China.
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China.
- InnoBM Pharmaceuticals Co. Ltd., Suzhou, China.
| |
Collapse
|
41
|
de Haas AM, Stolk DA, Schetters STT, Goossens-Kruijssen L, Keuning E, Ambrosini M, Boon L, Kalay H, Storm G, van der Vliet HJ, de Gruijl TD, van Kooyk Y. Vaccination with DC-SIGN-Targeting αGC Liposomes Leads to Tumor Control, Irrespective of Suboptimally Activated T-Cells. Pharmaceutics 2024; 16:581. [PMID: 38794243 PMCID: PMC11124829 DOI: 10.3390/pharmaceutics16050581] [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: 03/19/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer vaccines have emerged as a potent strategy to improve cancer immunity, with or without the combination of checkpoint blockade. In our investigation, liposomal formulations containing synthetic long peptides and α-Galactosylceramide, along with a DC-SIGN-targeting ligand, Lewis Y (LeY), were studied for their anti-tumor potential. The formulated liposomes boosted with anti-CD40 adjuvant demonstrated robust invariant natural killer (iNKT), CD4+, and CD8+ T-cell activation in vivo. The incorporation of LeY facilitated the targeting of antigen-presenting cells expressing DC-SIGN in vitro and in vivo. Surprisingly, mice vaccinated with LeY-modified liposomes exhibited comparable tumor reduction and survival rates to those treated with untargeted counterparts despite a decrease in antigen-specific CD8+ T-cell responses. These results suggest that impaired induction of antigen-specific CD8+ T-cells via DC-SIGN targeting does not compromise anti-tumor potential, hinting at alternative immune activation routes beyond CD8+ T-cell activation.
Collapse
Affiliation(s)
- Aram M. de Haas
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Dorian A. Stolk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sjoerd T. T. Schetters
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Laura Goossens-Kruijssen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Eelco Keuning
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Martino Ambrosini
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- LIPOSOMA BV, Meerpaalweg 5, 1332 BB Almere, The Netherlands
| | | | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Gert Storm
- LIPOSOMA BV, Meerpaalweg 5, 1332 BB Almere, The Netherlands
- Department of Biomaterials Science and Technology, University of Twente, 7500 AE Enschede, The Netherlands
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Hans J. van der Vliet
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- LAVA Therapeutics, 3584 CM Utrecht, The Netherlands
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
42
|
Zhang Y, Zhang H, Xu T, Zeng L, Liu F, Huang X, Liu Q. Interactions among microorganisms open up a new world for anti-infectious therapy. FEBS J 2024; 291:1615-1631. [PMID: 36527169 DOI: 10.1111/febs.16705] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
The human microbiome, containing bacteria, fungi, and viruses, is a community that coexists peacefully with humans most of the time, but with the potential to cause disease under certain conditions. When the environment changes or certain stimuli are received, microbes may interact with each other, causing or increasing the severity of disease in a host. With the appropriate methods, we can make these microbiota work for us, creating new applications for human health. This review discusses the wide range of interactions between microorganisms that result in an increase in susceptibility to, severity of, and mortality of diseases, and also briefly introduces how microorganisms interact with each other directly or indirectly. The study of microbial interactions and their mechanisms has revealed a new world of treatments for infectious disease. The regulation of the balance between intestinal flora, the correct application of probiotics, and the development of effective drugs by symbiosis all demonstrate the great contributions of the microbiota to human health and its powerful potential value. Consequently, the study of interactions between microorganisms plays an essential role in identifying the causes of diseases and the development of treatments.
Collapse
Affiliation(s)
- Yejia Zhang
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
| | - Hanchi Zhang
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
- The First Clinical Medical College, Nanchang University, China
| | - Tian Xu
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
| | - Lingbing Zeng
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
- The First Clinical Medical College, Nanchang University, China
| | - Fadi Liu
- The Department of Clinical Laboratory, Children's Hospital of Jiangxi Province, Nanchang, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
| |
Collapse
|
43
|
Rathod RJ, Sukumaran RK, Kedia N, Kumar J, Nair R, Chandy M, Gandikota L, Radhakrishnan VS. Chimeric Antigen Receptor T-cell based cellular therapies for cancer: An introduction and Indian perspective. Indian J Cancer 2024; 61:204-214. [PMID: 39152647 DOI: 10.4103/ijc.ijc_433_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/19/2021] [Indexed: 08/19/2024]
Abstract
Using one's own immune system for curing cancer has been an active field of research in cancer biology and therapeutics. One such opportunity in cellular immunotherapy is adoptive cell transfers. With the recent approval of CAR-T therapy as a cancer treatment, a whole new paradigm of cancer treatment has opened-up, with a ray of hope for relapsed/refractory cancer patients. Despite promising clinical outcomes, the therapy is in its early phase and remains out of reach for most patients due to its high cost and logistic challenges. In India, these therapies are unavailable and further confounded by the economic challenges and a large population. In this review, we discuss various aspects of T-cell immunotherapies with a special focus on CAR-T in the Indian scenario. We touch upon the basic scientific aspects, mechanism of action, manufacturing, clinical aspects and commercial aspects of the CAR-Tcell therapies and its future worldwide and in India.
Collapse
Affiliation(s)
- Reena J Rathod
- Cell and Gene Therapy Division, Intas Pharmaceuticals, Ahmedabad, Gujarat, India
| | - Reghu K Sukumaran
- Hematology Oncology and HCT, Tata Medical Center, Kolkata, West Bengal, India
| | - Neelam Kedia
- Cell and Gene Therapy Division, Intas Pharmaceuticals, Ahmedabad, Gujarat, India
| | - Jeevan Kumar
- Hematology Oncology and HCT, Tata Medical Center, Kolkata, West Bengal, India
| | - Reena Nair
- Hematology Oncology and HCT, Tata Medical Center, Kolkata, West Bengal, India
| | - Mammen Chandy
- Hematology Oncology and HCT, Tata Medical Center, Kolkata, West Bengal, India
| | | | | |
Collapse
|
44
|
Kane G, Lusi C, Brassil M, Atukorale P. Engineering approaches for innate immune-mediated tumor microenvironment remodeling. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 21:100406. [PMID: 38213392 PMCID: PMC10777078 DOI: 10.1016/j.iotech.2023.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Cancer immunotherapy offers transformative promise particularly for the treatment of lethal cancers, since a correctly trained immune system can comprehensively orchestrate tumor clearance with no need for continued therapeutic intervention. Historically, the majority of immunotherapies have been T cell-focused and have included immune checkpoint inhibitors, chimeric antigen receptor T cells, and T-cell vaccines. Unfortunately T-cell-focused therapies have failed to achieve optimal efficacy in most solid tumors largely because of a highly immunosuppressed 'cold' or immune-excluded tumor microenvironment (TME). Recently, a rapidly growing treatment paradigm has emerged that focuses on activation of tumor-resident innate antigen-presenting cells, such as dendritic cells and macrophages, which can drive a proinflammatory immune response to remodel the TME from 'cold' or immune-excluded to 'hot'. Early strategies for TME remodeling centered on free cytokines and agonists, but these approaches have faced significant hurdles in both delivery and efficacy. Systemic toxicity from off-target inflammation is a paramount concern in these therapies. To address this critical gap, engineering approaches have provided the opportunity to add 'built-in' capabilities to cytokines, agonists, and other therapeutic agents to mediate improved delivery and efficacy. Such capabilities have included protective encapsulation to shield them from degradation, targeting to direct them with high specificity to tumors, and co-delivery strategies to harness synergistic proinflammatory pathways. Here, we review innate immune-mediated TME remodeling engineering approaches that focus on cytokines, innate immune agonists, immunogenic viruses, and cell-based methods, highlighting emerging preclinical approaches and strategies that are either being tested in clinical trials or already Food and Drug Administration approved.
Collapse
Affiliation(s)
- G.I. Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - C.F. Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - M.L. Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - P.U. Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, USA
| |
Collapse
|
45
|
Shaha S, Rodrigues D, Mitragotri S. Locoregional drug delivery for cancer therapy: Preclinical progress and clinical translation. J Control Release 2024; 367:737-767. [PMID: 38325716 DOI: 10.1016/j.jconrel.2024.01.072] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Systemic drug delivery is the current clinically preferred route for cancer therapy. However, challenges associated with tumor localization and off-tumor toxic effects limit the clinical effectiveness of this route. Locoregional drug delivery is an emerging viable alternative to systemic therapies. With the improvement in real-time imaging technologies and tools for direct access to tumor lesions, the clinical applicability of locoregional drug delivery is becoming more prominent. Theoretically, locoregional treatments can bypass challenges faced by systemic drug delivery. Preclinically, locoregional delivery of drugs has demonstrated enhanced therapeutic efficacy with limited off-target effects while still yielding an abscopal effect. Clinically, an array of locoregional strategies is under investigation for the delivery of drugs ranging in target and size. Locoregional tumor treatment strategies can be classified into two main categories: 1) direct drug infusion via injection or implanted port and 2) extended drug elution via injected or implanted depot. The number of studies investigating locoregional drug delivery strategies for cancer treatment is rising exponentially, in both preclinical and clinical settings, with some approaches approved for clinical use. Here, we highlight key preclinical advances and the clinical relevance of such locoregional delivery strategies in the treatment of cancer. Furthermore, we critically analyze 949 clinical trials involving locoregional drug delivery and discuss emerging trends.
Collapse
Affiliation(s)
- Suyog Shaha
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Danika Rodrigues
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
46
|
Spalinger MR, Scharl M. Microbiota Manipulation as an Emerging Concept in Cancer Therapy. Visc Med 2024; 40:2-11. [PMID: 38312366 PMCID: PMC10836949 DOI: 10.1159/000534810] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/24/2023] [Indexed: 02/06/2024] Open
Abstract
Background The human body is colonized by billions of bacteria that provide nutrients to the host, train our immune system, and importantly affect our heath. It has long been suggested that microbes might play a role in tumor pathogenesis; however, compelling evidence was only provided in the past decades when novel detection methods that do not depend on culturing techniques had been developed. Summary The microbiome impacts tumor development and anti-tumor therapies on various levels. Bacteria can promote or suppress tumor growth via direct interactions with cancer cells, production of metabolites that promote or inhibit tumor growth, and via stimulation or suppression of the local and systemic immune response. Cancer patients harbor a distinct microbiome when compared to healthy controls, which could potentially be employed to detect, identify, and treat cancer. Manipulation of the microbiome either via supplementation of single strains, bacterial consortia, fecal microbiota transfer or the use of pre- and probiotics has been suggested as therapeutic approach to directly target tumor growth or to enhance the efficacy of current state-of-the-art treatment options. Key Messages (1) Bacteria have a tremendous impact on anti-cancer immune responses. (2) Cancer patients harbor a distinct microbiome when compared to healthy controls. (3) The microbiome seems to be cancer-type specific. (4) Exploitation of bacteria to promote anti-tumor therapy is a novel, very promising venue in cancer treatment.
Collapse
Affiliation(s)
| | - Michael Scharl
- Department for Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
47
|
Lu Y, Mei N, Ying Y, Wang D, Li X, Zhao Y, Zhu Y, Shen S, Yin B. Bacteria-Based Nanoprobes for Cancer Therapy. Int J Nanomedicine 2024; 19:759-785. [PMID: 38283198 PMCID: PMC10821665 DOI: 10.2147/ijn.s438164] [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: 09/05/2023] [Accepted: 01/04/2024] [Indexed: 01/30/2024] Open
Abstract
Surgical removal together with chemotherapy and radiotherapy has used to be the pillars of cancer treatment. Although these traditional methods are still considered as the first-line or standard treatments, non-operative situation, systemic toxicity or resistance severely weakened the therapeutic effect. More recently, synthetic biological nanocarriers elicited substantial interest and exhibited promising potential for combating cancer. In particular, bacteria and their derivatives are omnipotent to realize intrinsic tumor targeting and inhibit tumor growth with anti-cancer agents secreted and immune response. They are frequently employed in synergistic bacteria-mediated anticancer treatments to strengthen the effectiveness of anti-cancer treatment. In this review, we elaborate on the development, mechanism and advantage of bacterial therapy against cancer and then systematically introduce the bacteria-based nanoprobes against cancer and the recent achievements in synergistic treatment strategies and clinical trials. We also discuss the advantages as well as the limitations of these bacteria-based nanoprobes, especially the questions that hinder their application in human, exhibiting this novel anti-cancer endeavor comprehensively.
Collapse
Affiliation(s)
- Yiping Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Nan Mei
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yinwei Ying
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Dongdong Wang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Xuanxuan Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yajing Zhao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yuqi Zhu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People’s Republic of China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People’s Republic of China
| | - Bo Yin
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
48
|
Ricker CA, Meli K, Van Allen EM. Historical perspective and future directions: computational science in immuno-oncology. J Immunother Cancer 2024; 12:e008306. [PMID: 38191244 PMCID: PMC10826578 DOI: 10.1136/jitc-2023-008306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 01/10/2024] Open
Abstract
Immuno-oncology holds promise for transforming patient care having achieved durable clinical response rates across a variety of advanced and metastatic cancers. Despite these achievements, only a minority of patients respond to immunotherapy, underscoring the importance of elucidating molecular mechanisms responsible for response and resistance to inform the development and selection of treatments. Breakthroughs in molecular sequencing technologies have led to the generation of an immense amount of genomic and transcriptomic sequencing data that can be mined to uncover complex tumor-immune interactions using computational tools. In this review, we discuss existing and emerging computational methods that contextualize the composition and functional state of the tumor microenvironment, infer the reactivity and clonal dynamics from reconstructed immune cell receptor repertoires, and predict the antigenic landscape for immune cell recognition. We further describe the advantage of multi-omics analyses for capturing multidimensional relationships and artificial intelligence techniques for integrating omics data with histopathological and radiological images to encapsulate patterns of treatment response and tumor-immune biology. Finally, we discuss key challenges impeding their widespread use and clinical application and conclude with future perspectives. We are hopeful that this review will both serve as a guide for prospective researchers seeking to use existing tools for scientific discoveries and inspire the optimization or development of novel tools to enhance precision, ultimately expediting advancements in immunotherapy that improve patient survival and quality of life.
Collapse
Affiliation(s)
- Cora A Ricker
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kevin Meli
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
49
|
Yamamoto M, Tanaka Y, Takeda R, Nakamoto A, Nakamoto M, Yagita H, Sakai T. Soy isoflavone genistein attenuates the efficacy of immune checkpoint therapy in C57BL/6 mice inoculated with B16F1 melanoma and a high PD-L1 expression level reflects tumor resistance. J Clin Biochem Nutr 2024; 74:57-62. [PMID: 38292119 PMCID: PMC10822757 DOI: 10.3164/jcbn.23-76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 09/28/2023] [Indexed: 02/01/2024] Open
Abstract
Immune checkpoint therapy has been shown to be an effective therapy for many types of tumors. Much attention has been paid to the development of an effector target would be helpful for immune checkpoint therapy. Genistein has been shown to have an anti-tumor effect both in vitro and in vivo. In this study, we examined the effect of genistein on immune checkpoint blockade therapy against B16F1 melanoma tumors. Mice treated with genistein or anti-programmed death (PD)-1 antibody showed a significant decrease in tumor growth. However, treatment with genistein had no effect on or attenuated the efficacy of immune checkpoint therapy. The percentages of T cell receptor (TCR)β+CD4+ and TCRβ+CD8+ cells and the concentrations of interferon-γ and tumor necrosis factor-α in tumor tissue were not different among the experimental groups. A significant difference was also not found in microbe composition. Interestingly, a high expression level of PD-ligand (L)1 closely reflected the outcome of therapy by genistein or anti-PD-1 antibody. The study showed that a combination of genistein treatment does not improve the effect of immune blockade therapy. It also showed that a high PD-L1 expression level in tumors is a good prediction maker for the outcome of tumor therapy.
Collapse
Affiliation(s)
- Mako Yamamoto
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuko Tanaka
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Risako Takeda
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Akiko Nakamoto
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Mariko Nakamoto
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hideo Yagita
- Department of Immunology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Tohru Sakai
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
50
|
NISHIKAWA H. Establishment of immune suppression by cancer cells in the tumor microenvironment. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2024; 100:114-122. [PMID: 38346752 PMCID: PMC10978970 DOI: 10.2183/pjab.100.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/12/2023] [Indexed: 02/15/2024]
Abstract
With the clinical success of immune checkpoint inhibitors (ICIs), cancer immunotherapy has become an important pillar of cancer treatment in various types of cancer. However, more than half of patients fail to respond to ICIs, even in combination, uncovering a limited window of clinical responses. Therefore, it is essential to develop more effective cancer immunotherapies and to define biomarkers for stratifying responders and nonresponders by exploring the immunological landscape in the tumor microenvironment (TME). It has become clear that differences in immune responses in the TME determine the clinical efficacy of cancer immunotherapies. Additionally, gene alterations in cancer cells contribute to the development of the immunological landscape, particularly immune suppression in the TME. Therefore, integrated analyses of immunological and genomic assays are key for understanding diverse immune suppressive mechanisms in the TME. Developing novel strategies to control immune suppression in the TME from the perspective of immunology and the cancer genome is crucial for effective cancer immunotherapy (immune-genome precision medicine).
Collapse
Affiliation(s)
- Hiroyoshi NISHIKAWA
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|