1
|
Ma S, Yu J, Caligiuri MA. Natural killer cell-based immunotherapy for cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf036. [PMID: 40246292 DOI: 10.1093/jimmun/vkaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 04/19/2025]
Abstract
Natural killer (NK) cells are emerging as a promising tool for cancer immunotherapy due to their innate ability to selectively recognize and eliminate cancer cells. Over the past 3 decades, strategies to harness NK cells have included cytokines, small molecules, antibodies, and the adoptive transfer of autologous or allogeneic NK cells, both unmodified and genetically engineered. Despite favorable safety profiles in clinical trials, challenges such as limited in vivo persistence, exhaustion, and the suppressive tumor microenvironment continue to hinder their efficacy and durability. This review categorizes NK cell-based therapies into 3 major approaches: (i) cellular therapies, including unmodified and chimeric antigen receptor-engineered NK cells; (ii) cytokine-based strategies such as interleukin-2 and interleukin-15 derivatives; and (iii) antibody-based therapies, including immune checkpoint inhibitors and NK cell engagers. We highlight these advancements, discuss current limitations, and propose strategies to optimize NK cell-based therapies for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Jianhua Yu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, United States
- Institute for Precision Cancer Therapeutics and Immuno-Oncology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, United States
- Clemons Family Center for Transformative Cancer Research, University of California, Irvine, Irvine, CA, United States
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
2
|
Balkhi S, Zuccolotto G, Di Spirito A, Rosato A, Mortara L. CAR-NK cell therapy: promise and challenges in solid tumors. Front Immunol 2025; 16:1574742. [PMID: 40260240 PMCID: PMC12009813 DOI: 10.3389/fimmu.2025.1574742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Over the past few years, cellular immunotherapy has emerged as a promising treatment for certain hematologic cancers, with various CAR-T therapies now widely used in clinical settings. However, challenges related to the production of autologous cell products and the management of CAR-T cell toxicity highlight the need for new cell therapy options that are universal, safe, and effective. Natural killer (NK) cells, which are part of the innate immune system, offer unique advantages, including the potential for off-the-shelf therapy. A recent first-in-human trial of CD19-CAR-NK infusion in patients with relapsed/refractory lymphoid malignancies demonstrated safety and promising clinical activity. Building on these positive clinical outcomes, current research focuses on enhancing CAR-NK cell potency by increasing their in vivo persistence and addressing functional exhaustion. There is also growing interest in applying the successes seen in hematologic malignancies to solid tumors. This review discusses current trends and emerging concepts in the engineering of next-generation CAR- NK therapies. It will cover the process of constructing CAR-NK cells, potential targets for their manufacturing, and their role in various solid tumors. Additionally, it will examine the mechanisms of action and the research status of CAR-NK therapies in the treatment of solid tumors, along with their advantages, limitations, and future challenges. The insights provided may guide future investigations aimed at optimizing CAR-NK therapy for a broader range of malignancies.
Collapse
Affiliation(s)
- Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
3
|
Shen Y, Zhang H, Xue M, Zheng C, Chen Q. HSV-1 as a gene delivery platform for cancer gene therapy. Trends Pharmacol Sci 2025; 46:324-336. [PMID: 40069043 DOI: 10.1016/j.tips.2025.02.006] [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: 01/23/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 04/06/2025]
Abstract
Herpes simplex virus type 1 (HSV-1) is a DNA virus with strong replication capabilities, a large genomic payload (≥30 kb), and low toxicity, making it a prominent vector in cancer gene therapy. Clinically approved oncolytic HSV-1 (oHSV-1) variants, such as T-VEC and G47Δ, demonstrate safety and efficacy in localized tumors, but face challenges in treating metastatic disease. To address this issue, next-generation oHSV-1 designs focus on precision targeting and immune remodeling through the delivery of multiple exogenous genes. In this review, we provide an overview of the inherent characteristics of oHSV-1 as a gene delivery platform, focusing on its genetic modification strategies, safety challenges in clinical applications, and future directions to maximize its therapeutic potential.
Collapse
Affiliation(s)
- Yangkun Shen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Hucheng Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
4
|
Alwithenani A, Hengswat P, Chiocca EA. Oncolytic viruses as cancer therapeutics: From mechanistic insights to clinical translation. Mol Ther 2025:S1525-0016(25)00209-6. [PMID: 40143547 DOI: 10.1016/j.ymthe.2025.03.035] [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: 01/31/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 03/28/2025] Open
Abstract
Oncolytic virotherapy is a therapeutic approach that leverages genetically engineered or naturally occurring viruses to selectively target and destroy cancer cells while sparing normal tissues. This review provides an overview of the mechanisms of action by oncolytic viruses (OVs), including direct oncolysis, immune activation, and tumor microenvironment (TME) modulation. Despite significant progress, challenges such as immune resistance, tumor evasion mechanisms, and delivery barriers continue to limit the efficacy of OVs. To address these obstacles, recent advances in OV engineering have focused on arming viruses with immunomodulatory molecules, utilizing tumor-specific promoters, and employing CRISPR-based genome editing. Emerging strategies, such as dual-targeting OVs and viral enhancer drugs, have demonstrated promising potential in preclinical and clinical settings. This review also highlights findings from recent clinical trials, underscoring the translational challenges in scaling OVs for widespread therapeutic application. By exploring these innovations and their implications, we aim to shed light on the future directions of oncolytic virotherapy and its transformative potential in cancer treatment.
Collapse
Affiliation(s)
- Akram Alwithenani
- Harvey Cushing Neuro-oncology Laboratories, Department of Neurosurgery, Mass General Brigham, Harvard Medical School, Boston, MA, USA
| | - Pranaidej Hengswat
- Harvey Cushing Neuro-oncology Laboratories, Department of Neurosurgery, Mass General Brigham, Harvard Medical School, Boston, MA, USA
| | - E Antonio Chiocca
- Harvey Cushing Neuro-oncology Laboratories, Department of Neurosurgery, Mass General Brigham, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Dhital R, Kim Y, Kim D, Hernandez-Aguirre I, Hedberg J, Martin A, Cassady KA. Ruxolitinib and oHSV combination therapy increases CD4 T cell activity and germinal center B cell populations in murine sarcoma. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200929. [PMID: 39895689 PMCID: PMC11787636 DOI: 10.1016/j.omton.2024.200929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are a highly aggressive neoplasm of the peripheral nervous system and are resistant to most conventional cancer therapies. We previously showed that pretreatment with ruxolitinib (RUX) enhanced the efficacy of oncolytic herpes simplex virus (oHSV) virotherapy in this murine sarcoma model. A low abundance of tumor-infiltrating leukocytes and limitations in conventional flow cytometry restrict analyses to a narrow subset of immune cells, potentially introducing a confirmation bias. To address these limitations, we developed a 46-color spectral flow cytometry panel for the detailed analysis of immune cell dynamics following repeated oHSV dosing. Beyond the cytotoxic T lymphocyte (CTL) and regulatory T cell (Treg) changes reported in our earlier studies, RUX+oHSV treatment modulates myeloid and other lymphoid compartments, including germinal center B cell populations with enhanced activation. RUX+oHSV therapy also increased cytokine-expressing CD4(+) populations, predominantly granzyme B(+) cytotoxic-like, interferon (IFN)-γ(+) T helper type 1 (Th1)-like, and interleukin (IL)-21(+) T follicular helper (Tfh)-like phenotypes, within the tumor infiltrates, suggestive of potential tertiary lymphoid structure development in the treated tumors. Here, we illustrate the utility of a high-dimensional spectral flow cytometry panel that permits simultaneous evaluation of intratumoral CD4/CD8 T cell, Treg, γδ-T cell, natural killer T (NKT) cell, B cell, NK cell, monocyte, macrophage, granulocyte, myeloid-derived suppressor cell (MDSC), and dendritic cell functional changes from RUX+oHSV-treated MPNSTs.
Collapse
Affiliation(s)
- Ravi Dhital
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Yeaseul Kim
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Doyeon Kim
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Ilse Hernandez-Aguirre
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jack Hedberg
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Alexia Martin
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kevin A. Cassady
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Nationwide Children’s Hospital, Columbus, OH, USA
| |
Collapse
|
6
|
Poorva P, Mast J, Cao B, Shah MV, Pollok KE, Shen J. Killing the killers: Natural killer cell therapy targeting glioma stem cells in high-grade glioma. Mol Ther 2025:S1525-0016(25)00168-6. [PMID: 40040281 DOI: 10.1016/j.ymthe.2025.02.043] [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: 10/28/2024] [Revised: 01/22/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
High-grade gliomas (HGGs), including glioblastoma (GBM) in adults and diffuse intrinsic pontine glioma (DIPG) in children, are among the most aggressive and deadly brain tumors. A key factor in their resilience is the presence of glioma stem cells (GSCs), which drive tumor initiation, progression, and resistance to treatment. Targeting and eradicating GSCs holds potential for curing both GBM and DIPG. Natural killer (NK) cells, as part of the innate immune system, naturally recognize and destroy malignant cells. Recent advances in NK cell-based therapies, such as chimeric antigen receptor (CAR)-NK cells, NK cell engagers, and NK cell-derived exosomes, offer promising approaches for treating GBM and DIPG, particularly by addressing the persistence of GSCs. This review highlights these advancements, explores challenges such as the blood-brain barrier and the immunosuppressive tumor microenvironment, and proposes future directions for improving and clinically advancing these NK cell-based therapies for HGGs.
Collapse
Affiliation(s)
- Poorva Poorva
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Jensen Mast
- Biochemistry Graduate Program, Indiana University, Bloomington, IN 47405, USA
| | - Bihui Cao
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Mitesh V Shah
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karen E Pollok
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Jia Shen
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
7
|
Luo J, Guo M, Huang M, Liu Y, Qian Y, Liu Q, Cao X. Neoleukin-2/15-armored CAR-NK cells sustain superior therapeutic efficacy in solid tumors via c-Myc/NRF1 activation. Signal Transduct Target Ther 2025; 10:78. [PMID: 40025022 PMCID: PMC11873268 DOI: 10.1038/s41392-025-02158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/22/2024] [Accepted: 01/22/2025] [Indexed: 03/04/2025] Open
Abstract
Adoptive transfer of chimeric antigen receptor (CAR)-modified natural killer (NK) cells represents a transformative approach that has significantly advanced clinical outcomes in patients with malignant hematological conditions. However, the efficacy of CAR-NK cells in treating solid tumors is limited by their exhaustion, impaired infiltration and poor persistence in the immunosuppressive tumor microenvironment (TME). As NK cell functional states are associated with IL-2 cascade, we engineered mesothelin-specific CAR-NK cells that secrete neoleukin-2/15 (Neo-2/15), an IL-2Rβγ agonist, to resist immunosuppressive polarization within TME. The adoptively transferred Neo-2/15-armored CAR-NK cells exhibited enhanced cytotoxicity, less exhaustion and longer persistence within TME, thereby having superior antitumor activity against pancreatic cancer and ovarian cancer. Mechanistically, Neo-2/15 provided sustained and enhanced downstream IL-2 receptor signaling, which promotes the expression of c-Myc and nuclear respiratory factor 1 (NRF1) in CAR-NK cells. This upregulation was crucial for maintaining mitochondrial adaptability and metabolic resilience, ultimately leading to increased cytotoxicity and pronounced persistence of CAR-NK cells within the TME. The resistance against TME immunosuppressive polarization necessitated the upregulation of NRF1, which is essential to the augmentative effects elicited by Neo-2/15. Overexpression of NRF1 significantly bolsters the antitumor efficacy of CAR-NK cells both in vitro and in vivo, with increased ATP production. Collectively, Neo-2/15-expressing CAR-NK cells exerts superior antitumor effects by exhaustion-resistance and longer survival in solid tumors.
Collapse
Affiliation(s)
- Jianhua Luo
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Meng Guo
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
| | - Mingyan Huang
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Yanfang Liu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Yuping Qian
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Qiuyan Liu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
| | - Xuetao Cao
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
8
|
Xiong Y, Sun M, Yang Q, Zhang W, Song A, Tan Y, Mao J, Liu G, Xue P. Nanoparticle-based drug delivery systems to modulate tumor immune response for glioblastoma treatment. Acta Biomater 2025; 194:38-57. [PMID: 39884522 DOI: 10.1016/j.actbio.2025.01.050] [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: 10/16/2024] [Revised: 12/28/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
Glioblastoma (GBM) is a primary central nervous system neoplasm, characterized by a grim prognosis and low survival rates. This unfavorable therapeutic outcome is partially attributed to the inadequate immune infiltration and an immunosuppressive microenvironment, which compromises the effectiveness of conventional radiotherapy and chemotherapy. To this end, precise modulation of cellular dynamics in the immune system has emerged as a promising approach for therapeutic intervention. The advent of nanoparticle-based therapies has revolutionized cancer treatment and provided highly effective options. Consequently, various strategically designed nano-delivery platforms have been established to promote the efficacy of immune therapy against GBM. This review delves into the recent advancements in nano-based delivery systems that are designed to modulate immune cells in GBM microenvironment, and explores their multifaceted mechanisms, including the blockade of immune checkpoints, the restraint of immunosuppressive cells, the coordination of tumor-associated macrophages, the activation of innate immune cells, and the stimulation of adaptive immunity. Collectively, this summary not only advances the comprehension involved in modulating antitumor immune responses in GBM, but also paves the way for the development of innovative therapeutic strategies to conquer GBM. STATEMENT OF SIGNIFICANCE: Glioblastoma (GBM) is the most lethal brain tumor, with a median survival rate of merely 12-16 months after diagnosis. Despite surgical, radiation and chemotherapy treatments, the two-year survival rate for GBM patients is less than 10 %. The treatment of GBM is challenging mainly because several issues associated with the GBM microenvironment have not yet been resolved. Most recently, novel drug delivery approaches, based on the clear understanding of the intrinsic properties of GBM, have shown promise in overcoming some of the obstacles. In particular, taking account of the highly immunosuppressive tumor microenvironment in GBM, recent advancements in nano-based delivery systems are put forward to stimulate immune cells in GBM and unravel their multifaceted mechanisms. This review summarizes the latest nanoparticle-based drug delivery systems to modulate tumor immune response for glioblastoma treatment. Moreover, the development trends and challenges of nanoparticle-based drug delivery systems in modulating the immunity of GBM are predicted, which may facilitate widespread regimens springing up for successfully treating GBM.
Collapse
Affiliation(s)
- Yongqi Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Maoyuan Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qinhao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wenli Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Anchao Song
- College of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Jinning Mao
- Health Medical Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Peng Xue
- School of Materials and Energy, Southwest University, Chongqing 400715, China; Yibin Academy of Southwest University, Yibin 644005, China.
| |
Collapse
|
9
|
Park JD, Shin HE, An YS, Jang HJ, Park J, Kim SN, Park CG, Park W. Advancing Natural Killer Cell Therapy: Genetic Engineering Strategies for Enhanced Cancer Immunotherapy. Ann Lab Med 2025; 45:146-159. [PMID: 39774132 PMCID: PMC11788708 DOI: 10.3343/alm.2024.0380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/06/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Natural killer (NK) cells are pivotal innate immune system components that exhibit spontaneous cytolytic activity against abnormal cells, such as infected and tumor cells. NK cells have shown significant promise in adoptive cell therapy because of their favorable safety profiles and minimal toxicity in clinical settings. Despite their advantages, the therapeutic application of unmodified NK cells faces challenges, including limited in vivo persistence, particularly in the immunosuppressive tumor microenvironment. Recent advances in genetic engineering have enhanced the therapeutic potential of NK cells by addressing these limitations and improving their therapeutic efficacy. In this review, we have described various methodologies for the genetic modification of NK cells, including viral vectors, electroporation, and nanoparticle-based approaches. The ongoing research on nanomaterialbased approaches highlights their potential to overcome current limitations in NK cell therapy, paving the way for advanced cancer therapy and improved clinical outcomes. In this review, we also emphasize the potential of engineered NK cells in cancer immunotherapy and other clinical applications, highlighting the expanding scope of NK cell-based treatments and the critical role of innovative genetic engineering techniques.
Collapse
Affiliation(s)
- Joo Dong Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Ha Eun Shin
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Manoa, Honolulu, USA
| | - Yeon Su An
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Hye Jung Jang
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Manoa, Honolulu, USA
| | - Se-Na Kim
- Department of Industrial Cosmetic Science, Chungbuk National University, Cheongju, Korea
- Research and Development Center, MediArk Inc., Cheongju, Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, Institute for Cross-disciplinary Studies, Sungkyunkwan University, Suwon, Korea
- Department of Intelligent Precision Healthcare Convergence, Institute for Cross-disciplinary Studies, Sungkyunkwan University, Suwon, Korea
- Korea Institute of Science and Technology, Seoul, Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Korea
- Korea Institute of Science and Technology, Seoul, Korea
- Department of MetaBioHealth, Institute for Cross-disciplinary Studies, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
10
|
Du W, Na J, Zhong L, Zhang P. Advances in preclinical and clinical studies of oncolytic virus combination therapy. Front Oncol 2025; 15:1545542. [PMID: 39990685 PMCID: PMC11842258 DOI: 10.3389/fonc.2025.1545542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
Oncolytic viruses represent a distinct class of viruses that selectively infect and destroy tumor cells while sparing normal cells. Despite their potential, oncolytic viruses encounter several challenges as standalone therapies. Consequently, the combination of oncolytic viruses with other therapeutic modalities has emerged as a prominent research focus. This paper summarizes the tumor-killing mechanisms of oncolytic viruses, explores their integration with radiotherapy, chemotherapy, immune checkpoint inhibitors, CAR-T, and CAR-NK therapies, and provides an overview of related clinical trials. By synthesizing these advancements, this study seeks to offer valuable insights for the clinical translation of oncolytic virus combination therapies.
Collapse
Affiliation(s)
- Wenlong Du
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Pumin Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
| |
Collapse
|
11
|
Ren Y, Xue M, Hui X, Liu X, Farooq MA, Chen Y, Ji Y, Duan Y, Ajmal I, Yao J, Jiang W. Chimeric cytokine receptor TGF-β RⅡ/IL-21R improves CAR-NK cell function by reversing the immunosuppressive tumor microenvironment of gastric cancer. Pharmacol Res 2025; 212:107637. [PMID: 39884449 DOI: 10.1016/j.phrs.2025.107637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
Gastric cancer remains a significant global health burden, characterized by regional variations in incidence and poor survival prospects in advanced stages. Natural killer (NK) cells play a crucial role in the body's anti-cancer defense, and chimeric antigen receptor (CAR)-NK cell therapy is gaining attention as a cutting-edge and promising treatment method. This study aims to tackle the challenge of TGF-β-mediated tumor immune evasion within the immunosuppressive tumor microenvironment by designing a novel chimeric cytokine receptor TRII/21 R, which consists of extracellular domains of TGF-β receptor II (TRII) and transmembrane and intracellular domains of IL-21 receptor (21 R) and can convert the immunosuppressive signal from TGF-β in the tumor microenvironment (TME) into an NK cell activation signal through the IL-21R-STAT3 pathway. We successfully constructed NKG2D-CAR-NK cells expressing TRII/21 R and demonstrated strong anti-tumor activity against cancer cells both in vitro and in vivo. The co-expression of TRII/21 R in CAR-NK cells enhanced the cytotoxicity, promoted proliferation and survival capabilities, and reduced the expression of exhaustion markers. In the xenograft mouse model, TRII/21R-CAR-NK cells significantly inhibited tumor growth and improved the survival rate of tumor-bearing mice compared to the mice receiving control CAR-NK cells. Additionally, TRII/21 R co-expression enhanced NK cells' infiltration, activation, and persistence within the tumor, indicating a robust anti-tumor response mediated by the JAK-STAT3 signaling pathway. This study underscores the therapeutic potential of TRII/21R-modified CAR-NK cells as a breakthrough strategy for combating cancer.
Collapse
MESH Headings
- Killer Cells, Natural/immunology
- Animals
- Tumor Microenvironment/immunology
- Humans
- Stomach Neoplasms/immunology
- Stomach Neoplasms/therapy
- Stomach Neoplasms/pathology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Cell Line, Tumor
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Receptor, Transforming Growth Factor-beta Type II/immunology
- Mice
- Immunotherapy, Adoptive/methods
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- NK Cell Lectin-Like Receptor Subfamily K/immunology
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/immunology
- Receptors, Interleukin-21/metabolism
- Receptors, Interleukin-21/genetics
- Mice, Inbred BALB C
- Signal Transduction
- Mice, Nude
Collapse
Affiliation(s)
- Yaojun Ren
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China; College of Life Science, Xinjiang Normal University, Urumqi 830054, China
| | - Min Xue
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xinhui Hui
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiuyu Liu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Muhammad Asad Farooq
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yiran Chen
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuzhou Ji
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yixin Duan
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Iqra Ajmal
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jie Yao
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
12
|
Wu H, Liu Q, Wang F, Gao W, Zhou F, Zhao H. Research Progress of NK Cells in Glioblastoma Treatment. Onco Targets Ther 2025; 18:87-106. [PMID: 39845286 PMCID: PMC11752833 DOI: 10.2147/ott.s486411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/01/2025] [Indexed: 01/24/2025] Open
Abstract
NK cells are a type of antitumor immune cell with promising clinical application, following T cells. The activity of NK cells is primarily regulated by their surface receptors and immune microenvironment. In gliomas, the tumor microenvironment exerts a strong immunosuppressive effect, which significantly reduces the clinical efficacy of NK cell immunotherapy. Therefore, this review aims to discuss the latest research on the role of NK cells in glioma immunotherapy, focusing on aspects such as NK cell development, function, and localization. It summarizes information on the compounds, monoclonal antibodies, and cytokine therapies targeting NK cells while emphasizing the current status and trends of gene-modified NK cells in glioma treatment. Additionally, it explores the molecular mechanisms underlying immune escape in glioma cells, providing a theoretical foundation and new perspectives for NK cell-based immunotherapy in gliomas.
Collapse
Affiliation(s)
- Hao Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, People’s Republic of China
| | - Qi Liu
- Department of Neurosurgery, The First Hospital of Yulin, Yulin, People’s Republic of China
| | - Fenglu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, People’s Republic of China
| | - Wenwen Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, People’s Republic of China
| | - Feng Zhou
- Department of Neurosurgery, The First Hospital of Yulin, Yulin, People’s Republic of China
| | - Haikang Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, People’s Republic of China
| |
Collapse
|
13
|
Rao ZY, Kuang J, Pan T, Qin YT, Huang QX, Sun YL, Zhao K, Jin XK, Yang CH, Zhang SM, Yan Y, Zhang XZ. Cationic Magnetic Nanoparticles Activate Natural Killer Cells for the Treatment of Glioblastoma. ACS NANO 2025; 19:649-661. [PMID: 39711185 DOI: 10.1021/acsnano.4c11250] [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: 12/24/2024]
Abstract
The blood-brain barrier (BBB) and the immunosuppressive microenvironment of glioblastoma (GBM) severely hinder the infiltration and activity of natural killer (NK) cells, thereby reducing their clinical efficacy in GBM treatment. To address this challenge, we introduced an engineered living material, HEFDS-NK cells, designed to enhance the penetration of NK cells across the BBB and improve their cytotoxicity against GBM. HEFDS comprises magnetic nanoparticles modified using cationic polyethylenimine (PEI), selenocysteine (Sec), and sodium hyaluronate (HA) and cocultured with NK cells to form HEFDS-NK cells. With the assistance of HA and magnet targeting, HEFDS-NK cells can effectively cross the BBB and localize at the GBM site. Moreover, PEI enhances the expression of C-X-C chemokine receptor type 4 (CXCR4) and C-C chemokine receptor type 4 (CCR4) on NK cells, thereby improving their recognition and cytotoxicity against GBM. Additionally, Sec boosts the immune activity of NK cells against GBM. Upon recognizing GBM, the activated HEFDS-NK cells produce Granzyme B, Perforin, and IFN-γ, ultimately achieving effective therapy for GBM. This study demonstrates an effective treatment of GBM while enhancing NK cell activity and their ability to penetrate the BBB, providing an innovative and high-precision therapeutic approach for GBM.
Collapse
Affiliation(s)
- Zhi-Yong Rao
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Jing Kuang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Ting Pan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - You-Teng Qin
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Qian-Xiao Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yu-Liang Sun
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Kai Zhao
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xiao-Kang Jin
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Chi-Hui Yang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Shi-Man Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yu Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
14
|
Hosseinalizadeh H, Wang LS, Mirzaei H, Amoozgar Z, Tian L, Yu J. Emerging combined CAR-NK cell therapies in cancer treatment: Finding a dancing partner. Mol Ther 2025:S1525-0016(24)00895-5. [PMID: 39754357 DOI: 10.1016/j.ymthe.2024.12.057] [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: 03/21/2024] [Revised: 09/21/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025] Open
Abstract
In recent decades, immunotherapy with chimeric antigen receptors (CARs) has revolutionized cancer treatment and given hope where other cancer therapies have failed. CAR-natural killer (NK) cells are NK cells that have been engineered ex vivo with a CAR on the cell membrane with high specificity for specific target antigens of tumor cells. The impressive results of several studies suggest that CAR-NK cell therapy has significant potential and successful performance in cancer treatment. Despite its effectiveness, CAR-NK cell therapy can have significant challenges when it comes to treating cancer. These challenges include tumor heterogeneity, antigen escape, an immunosuppressive tumor microenvironment, limited tissue migration from blood, exhaustion of CAR-NK cells, and inhibition by immunosuppressive checkpoint molecule signaling, etc. In CAR-T cell therapy, the use of combined approaches has shown encouraging outcomes for tumor regression and improved cancer treatment compared to single therapies. Therefore, to overcome these significant challenges in CAR-NK cells, innovative combination therapies of CAR-NK cells with other conventional therapies (e.g., chemotherapy and radiotherapy) or other immunotherapies are needed to counteract the above challenges and thereby increase the activity of CAR-NK cells. This review comprehensively discusses various cancer-treatment approaches in combination with CAR-NK cell therapy in the hope of providing valuable insights that may improve cancer treatment in the near future.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Li-Shu Wang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lei Tian
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| | - Jianhua Yu
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
15
|
Tao JH, Zhang J, Li HS, Zhou Y, Guan CX. Nature killer cell for solid tumors: Current obstacles and prospective remedies in NK cell therapy and beyond. Crit Rev Oncol Hematol 2025; 205:104553. [PMID: 39515404 DOI: 10.1016/j.critrevonc.2024.104553] [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: 11/25/2023] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, cell therapy has emerged as an innovative treatment method for the management of clinical tumors following immunotherapy. Among them, Natural killer (NK) cell therapy has achieved a significant breakthrough in the treatment of hematological tumors. However, the therapeutic effectiveness of NK cells in the treatment of solid tumors remains challenging. With the progress of gene editing and culture techniques and their application to NK cell engineering, it is expected that NK cell therapy will revolutionize the treatment of solid tumors. In this review, we explore the discovery and biological properties of NK cells, their role in the tumor microenvironment, and the therapeutic strategies, clinical trials, challenges, and prospects of NK cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Jia-Hao Tao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Jun Zhang
- Ascle Therapeutics, Suzhou, Jiangsu 215000, China
| | - Hua-Shun Li
- Ascle Therapeutics, Suzhou, Jiangsu 215000, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
16
|
Zhai Y, Zhang W, Wang J, Kong Y, Rong R, Lang T, Zheng C, Wang Y, Yu Y, Zhu HH, Cai Y, Zhang P, Li Y. Interleukin 15-Presenting Nanovesicles with Doxorubicin-Loaded Ferritin Cores for Cancer Immunochemotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409194. [PMID: 39625860 PMCID: PMC11789581 DOI: 10.1002/advs.202409194] [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: 08/05/2024] [Revised: 11/13/2024] [Indexed: 01/30/2025]
Abstract
Interleukin 15 (IL15) is crucial for fostering the survival and proliferation of nature killer (NK) cells and cytotoxic T lymphocytes (CTLs), playing a pivotal role in tumor control. However, IL15 supplementary therapy encounters challenges such as systemic inflammation and non-specific stimulation of cancer cells. Herein, a nanovesicle termed DoxFILN, comprising a membrane presenting IL15/IL15 receptor α complexes (IL15c) and a core of doxorubicin-loaded ferritin (Dox-Fn) are reported. The DoxFILN significantly enhances the densities and activities of intratumoral CTLs and NK cells. Mechanistically, DoxFILN undergoes deshelling in the acidic tumor microenvironment, releasing Dox-Fn and membrane-bound IL15c. Dox-Fn selectively target transferrin receptors on cancerous cells, facilitating intracellular Dox release and inducing immunogenic cell death. Concurrently, membrane-bound IL15c recognizes and activates IL15 receptor β/γc heterodimers, leading to a remarkable increase in the proliferation and activation of CTLs (16-fold and 28-fold) and NK cells (37-fold and 50-fold). The IL15-displaying nanovesicle introduced here holds promise as a potential platform for immunochemotherapy in the treatment of cancer.
Collapse
Affiliation(s)
- Yihui Zhai
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wen Zhang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- China State Institute of Pharmaceutical IndustryShanghai201203China
| | - Jinming Wang
- State Key Laboratory of Oncogenes and Related GenesRenji‐Med‐X Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200127China
| | - Ying Kong
- Yantai Institute of Materia MedicaShandong264000China
| | - Rong Rong
- Yantai Institute of Materia MedicaShandong264000China
| | - Tianqun Lang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Chao Zheng
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- China State Institute of Pharmaceutical IndustryShanghai201203China
| | - Yanke Wang
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai201210China
| | - Yang Yu
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai201210China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related GenesRenji‐Med‐X Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200127China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine & Advanced PreparationsYantai Institute of Materia MedicaShandong264000China
| | - Pengcheng Zhang
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and DevicesShanghaiTech UniversityShanghai201210China
- Shanghai Clinical Research and Trial CenterShanghai201203China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine & Advanced PreparationsYantai Institute of Materia MedicaShandong264000China
| |
Collapse
|
17
|
Karamivandishi A, Hatami A, Eslami MM, Soleimani M, Izadi N. Chimeric antigen receptor natural killer cell therapy: A systematic review of preclinical studies for hematologic and solid malignancies. Hum Immunol 2025; 86:111207. [PMID: 39667204 DOI: 10.1016/j.humimm.2024.111207] [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: 09/02/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 12/14/2024]
Abstract
Advancements in the field of CAR-T therapy have brought about a revolution in the treatment of numerous types of cancer in the past ten years. However, despite the remarkable success achieved thus far, certain barriers impede the widespread implementation of this therapy such as intricate manufacturing processes and treatment-associated toxicities. As an alternative, chimeric antigen receptor-engineered natural killer cell (CAR-NK) therapy presents a viable opportunity for a simpler and more cost-effective "off-the-shelf" treatment option, which is likely to result in fewer adverse reactions. A total of 71 studies were included in this review. Eligible studies were searched and reviewed from the databases of PubMed, Web of Science and Scopus. Based on data extracted from articles, we concluded that CAR-NK cell efficiency can vary considerably depending on factors such as tumor model, dosage, CAR generation and expansion method. Furthermore, investigating consequences of utilizing various constructs and generations of CAR-NK cells on their anti-tumor activity examined in this review.
Collapse
Affiliation(s)
- Arezoo Karamivandishi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Hatami
- Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Masoud Eslami
- Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Neda Izadi
- Research Center for Social Determinants of Health,Research institute for metabolic and obesity disorders, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Tian H, Yao J, Ba Q, Meng Y, Cui Y, Quan L, Gong W, Wang Y, Yang Y, Yang M, Gao C. Cerebral biomimetic nano-drug delivery systems: A frontier strategy for immunotherapy. J Control Release 2024; 376:1039-1067. [PMID: 39505218 DOI: 10.1016/j.jconrel.2024.10.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/19/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Brain diseases are a significant threat to human health, especially in the elderly, and this problem is growing as the aging population increases. Efficient brain-targeted drug delivery has been the greatest challenge in treating brain disorders due to the unique immune environment of the brain, including the blood-brain barrier (BBB). Recently, cerebral biomimetic nano-drug delivery systems (CBNDSs) have provided a promising strategy for brain targeting by mimicking natural biological materials. Herein, this review explores the latest understanding of the immune microenvironment of the brain, emphasizing the immune mechanisms of the occurrence and progression of brain disease. Several brain targeting systems are summarized, including cell-based, exosome-based, protein-based, and microbe-based CBNDSs, and their immunological mechanisms are highlighted. Moreover, given the rise of immunotherapy, the latest applications of CBNDSs in immunotherapy are also discussed. This review provides a comprehensive understanding of CBNDSs and serves as a guideline for immunotherapy in treating brain diseases. In addition, it provides inspiration for the future of CBNDSs.
Collapse
Affiliation(s)
- Hao Tian
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Jiaxin Yao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Qi Ba
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Yuanyuan Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanan Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Liangzhu Quan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
19
|
Ahuja S, Zaheer S. The evolution of cancer immunotherapy: a comprehensive review of its history and current perspectives. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:51-73. [PMID: 39778508 PMCID: PMC11717579 DOI: 10.14216/kjco.24009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy uses the body's immune system to combat cancer, marking a significant advancement in treatment. This review traces its evolution from the late 19th century to its current status. It began with William Coley's pioneering work using bacterial toxins to stimulate the immune system against cancer cells, establishing the foundational concept of immunotherapy. In the mid-20th century, cytokine therapies like interferons and interleukins emerged, demonstrating that altering the immune response could reduce tumors and highlighting the complex interplay between cancer and the immune system. The discovery of immune checkpoints, regulatory pathways that prevent autoimmunity but are exploited by cancer cells to evade detection, was a pivotal development. Another major breakthrough is CAR-T cell therapy, which involves modifying a patient's T cells to target cancer-specific antigens. This personalized treatment has shown remarkable success in certain blood cancers. Additionally, cancer vaccines aim to trigger immune responses against tumor-specific or associated antigens, and while challenging, ongoing research is improving their efficacy. The historical progression of cancer immunotherapy, from Coley's toxins to modern innovations like checkpoint inhibitors and CAR-T cell therapy, underscores its transformative impact on cancer treatment. As research delves deeper into the immune system's complexities, immunotherapy is poised to become even more crucial in oncology, offering renewed hope to patients globally.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| |
Collapse
|
20
|
Liu Y, Chen J, Tian J, Hao Y, Ma X, Zhou Y, Feng L. Engineered CAR-NK Cells with Tolerance to H2O2 and Hypoxia Can Suppress Postoperative Relapse of Triple-Negative Breast Cancers. Cancer Immunol Res 2024; 12:1574-1588. [PMID: 39023168 DOI: 10.1158/2326-6066.cir-23-1017] [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: 11/30/2023] [Revised: 04/19/2024] [Accepted: 07/16/2024] [Indexed: 07/20/2024]
Abstract
Surgical resection is a primary treatment option for patients with triple-negative breast cancer (TNBC), but it is associated with a high rate of postoperative local and metastatic relapse. Although chimeric antigen receptor-engineered NK (CAR-NK) cell therapy can specifically recognize and eradicate tumor cells, its therapeutic potency toward TNBCs is markedly suppressed by the hostile tumor microenvironment, which restricts the infiltration, survival, and effector functions of CAR-NK cells inside tumor masses. In this study, HER1-overexpressing TNBC-targeted CAR-NK (HER1-CAR-NK) cells were genetically engineered with catalase to endow them with tolerance toward the high levels of oxidative stress and hypoxia inside TNBC tumors through the catalytic decomposition of hydrogen peroxide, which is a principle reactive oxygen species inside tumors, into O2. We refer to these cells as HER1-CAR-CAT-NK cells. Upon intratumoral fixation with an injectable alginate hydrogel, HER1-CAR-CAT-NK cells enabled sustained tumor hypoxia attenuation and exhibited markedly enhanced persistence and effector functions inside TNBC tumors. As a result, locoregional HER1-CAR-CAT-NK cell therapy not only inhibited the growth of local primary residual tumors but also elicited systemic antitumor activity to suppress the growth of distant tumors. This study highlights that genetic engineering of HER1-CAR-NK cells with catalase is a promising strategy to suppress the postoperative local and distant relapse of TNBC tumors.
Collapse
Affiliation(s)
- Yan Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Cancer Institute, Department of Biochemistry, College of Life Science, Nanjing Normal University, Nanjing, P. R. China
| | - Jiahui Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Cancer Institute, Department of Biochemistry, College of Life Science, Nanjing Normal University, Nanjing, P. R. China
| | - Jia Tian
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Cancer Institute, Department of Biochemistry, College of Life Science, Nanjing Normal University, Nanjing, P. R. China
| | - Yu Hao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, P. R. China
| | - Xinxing Ma
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
| | - Yehui Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
| | - Liangzhu Feng
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, P. R. China
| |
Collapse
|
21
|
Qi Z, Gu J, Qu L, Shi X, He Z, Sun J, Tan L, Sun M. Advancements of engineered live oncolytic biotherapeutics (microbe/virus/cells): Preclinical research and clinical progress. J Control Release 2024; 375:209-235. [PMID: 39244159 DOI: 10.1016/j.jconrel.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The proven efficacy of immunotherapy in fighting tumors has been firmly established, heralding a new era in harnessing both the innate and adaptive immune systems for cancer treatment. Despite its promise, challenges such as inefficient delivery, insufficient tumor penetration, and considerable potential toxicity of immunomodulatory agents have impeded the advancement of immunotherapies. Recent endeavors in the realm of tumor prophylaxis and management have highlighted the use of living biological entities, including bacteria, oncolytic viruses, and immune cells, as a vanguard for an innovative class of live biotherapeutic products (LBPs). These LBPs are gaining recognition for their inherent ability to target tumors. However, these LBPs must contend with significant barriers, including robust immune clearance mechanisms, cytotoxicity and other in vivo adverse effects. Priority must be placed on enhancing their safety and therapeutic indices. This review consolidates the latest preclinical research and clinical progress pertaining to the exploitation of engineered biologics, spanning bacteria, oncolytic viruses, immune cells, and summarizes their integration with combination therapies aimed at circumventing current clinical impasses. Additionally, the prospective utilities and inherent challenges of the biotherapeutics are deliberated, with the objective of accelerating their clinical application in the foreseeable future.
Collapse
Affiliation(s)
- Zhengzhuo Qi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Junmou Gu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lihang Qu
- The 4th People's Hospital of Shenyang, China Medical University, Shenyang, Liaoning, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Lingchen Tan
- School of Life Sciences and Biopharmaceutical, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| |
Collapse
|
22
|
Li X, Zhang Y, Mao Z, Zhao H, Cao H, Wang J, Liu W, Dai S, Yang Y, Huang Y, Wang H. Decorin-armed oncolytic adenovirus promotes natural killers (NKs) activation and infiltration to enhance NK therapy in CRC model. MOLECULAR BIOMEDICINE 2024; 5:48. [PMID: 39482550 PMCID: PMC11527862 DOI: 10.1186/s43556-024-00212-z] [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: 03/25/2024] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignant tumor of the gastrointestinal system, with the third and second highest incidence and mortality rates globally in 2020, respectively. Immunotherapy has developed rapidly in recent years. Natural killer (NK) cells have received increasing attention in the field of tumor immunotherapy due to their recognition and killing tumor cells without the limitations of major histocompatibility complexes. However, constraints within the tumor microenvironment that impede the infiltration and proliferation of NK cells result in poor efficacy of NK cell therapy for solid tumors. Oncolytic viral therapy is an immunogenic treatment with the potential to enhance anti-tumour immune responses and promote immune cell infiltration. In this study, we synergistically combine NK cells with an oncolytic adenovirus carrying Decorin (rAd.DCN) for the treatment of colorectal cancer (CRC) in a xenograft mouse model. By using Flow cytometry, real-time quantitative PCR and Calcein-AM release assay, we found that rAd.DCN could effectively promote proliferation, activation and degranulation of NK cells, up-regulate expression and secretion of NK cell killing activity-related factors, and enhance their killing activity. The efficacy is better than that of the blank control oncolytic virus rAd.Null. Combined treatment significantly inhibited tumor growth, increased the number of NK cells in peripheral blood, promoted the killing function of NK cells, and increased the expression levels of perforin and IFN-γ. At the same time, more NK cells were recruited to infiltrate tumor tissue. Our study established the feasibility of combination NK cells and oncolytic adenovirus application, thus expanding the scope of potentially curative treatments for NK cells in CRC.
Collapse
Affiliation(s)
- Xue Li
- College of Life Science, Anhui Medical University, Hefei, 230032, P.R. China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yuning Zhang
- Department of Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, 100071, China
| | - Zhuang Mao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Huiqiang Zhao
- Department of Healthcare, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hu Cao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jingyi Wang
- Beijing Jingda Biotechnology Co. Ltd, Beijing, 102629, China
| | - Wei Liu
- Beijing Jingda Biotechnology Co. Ltd, Beijing, 102629, China
| | - Shiyun Dai
- Beijing Jingda Biotechnology Co. Ltd, Beijing, 102629, China
| | - Yuefeng Yang
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yuanyuan Huang
- Beijing Jingda Biotechnology Co. Ltd, Beijing, 102629, China.
| | - Hua Wang
- College of Life Science, Anhui Medical University, Hefei, 230032, P.R. China.
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
23
|
Jacome MA, Wu Q, Piña Y, Etame AB. Evolution of Molecular Biomarkers and Precision Molecular Therapeutic Strategies in Glioblastoma. Cancers (Basel) 2024; 16:3635. [PMID: 39518074 PMCID: PMC11544870 DOI: 10.3390/cancers16213635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma is the most commonly occurring malignant brain tumor, with a high mortality rate despite current treatments. Its classification has evolved over the years to include not only histopathological features but also molecular findings. Given the heterogeneity of glioblastoma, molecular biomarkers for diagnosis have become essential for initiating treatment with current therapies, while new technologies for detecting specific variations using computational tools are being rapidly developed. Advances in molecular genetics have made possible the creation of tailored therapies based on specific molecular targets, with various degrees of success. This review provides an overview of the latest advances in the fields of histopathology and radiogenomics and the use of molecular markers for management of glioblastoma, as well as the development of new therapies targeting the most common molecular markers. Furthermore, we offer a summary of the results of recent preclinical and clinical trials to recognize the current trends of investigation and understand the possible future directions of molecular targeted therapies in glioblastoma.
Collapse
Affiliation(s)
- Maria A. Jacome
- Departamento de Ciencias Morfológicas Microscópicas, Universidad de Carabobo, Valencia 02001, Venezuela
| | - Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA; (Q.W.); (Y.P.)
| | - Yolanda Piña
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA; (Q.W.); (Y.P.)
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA; (Q.W.); (Y.P.)
| |
Collapse
|
24
|
Hu Z, Li Y, Yang J, Liu J, Zhou H, Sun C, Tian C, Zhu C, Shao M, Wang S, Wei L, Liu M, Li S, Wang J, Xu H, Zhu W, Li X, Li J. Improved antitumor effectiveness of oncolytic HSV-1 viruses engineered with IL-15/IL-15Rα complex combined with oncolytic HSV-1-aPD1 targets colon cancer. Sci Rep 2024; 14:23671. [PMID: 39389985 PMCID: PMC11467195 DOI: 10.1038/s41598-024-72888-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Oncolytic virotherapy is emerging as a promising therapeutic avenue for cancer treatment, harnessing both innate and tumor-specific immune responses for targeted tumor elimination. In this study, we present a novel oncolytic virus (oHSV1-IL15B) derived from herpes simplex virus-1 (HSV-1), armed with IL-15/IL-15Rα complex, with a focus on treating colon cancer combined with oncolytic HSV-1 expressing anti-PD-1 antibody (oHSV1-aPD1). Results from our study reveal that recombinant oHSV-1 virus equipped with IL-15/IL-15Rα complex exhibited significant anti-tumor effects in a murine CT26 colon adenocarcinoma model. Notably, oHSV1-IL15B combined with oHSV-1-aPD1 demonstrates superior tumor inhibition and prolonged overall survival compared to oHSV1-mock and monotherapy groups. Further exploration highlights the impact of oHSV1-IL15B, oHSV-1-aPD1 and combined group on antitumor capacity, revealing a substantial increase in CD8+ T and CD4+ T cell proportions of CT26-bearing BALB/c mice and promoting apoptosis in tumor tissue. The study emphasizes the pivotal role of cytotoxic CD8+T cells in oncolytic virotherapy, demonstrating that recombinant oHSV1-IL15B combined with oncolytic HSV-1-aPD1 induces a robust tumor-specific T cell response. RNA sequence analysis highlighted oHSV1-IL15B combined with oHSV1-aPD1 improved tumors immune microenvironment on immune response, antiviral response-related genes and apoptosis-related genes, which contributed to anti-tumor immunotherapy. The findings underscore the promising antitumor activity achieved through the combination of IL-15/IL-15Rα complex and anti-PD-1 antibody with oHSV-1. This research opens avenues for diverse therapeutic strategies, suggesting the potential of synergistically utilizing cytokines and anti-PD-1 antibody with oncolytic viruses to enhance immunotherapy for cancer management.
Collapse
Affiliation(s)
- Zongfeng Hu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Yixiao Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | | | - Jiajia Liu
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Hua Zhou
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chunyang Sun
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chao Tian
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chengyang Zhu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Mingxia Shao
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Shengrun Wang
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Lijun Wei
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Min Liu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Shuzhen Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Jinyu Wang
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Haitian Xu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Wei Zhu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Xiaopeng Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China.
- Beijing WellGene Company, Ltd, Beijing, 100085, China.
| | - Jingfeng Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China.
| |
Collapse
|
25
|
Tachi T, Kijima N, Kuroda H, Ikeda S, Murakami K, Nakagawa T, Yaga M, Nakagawa K, Utsugi R, Hirayama R, Okita Y, Kagawa N, Kishima H, Imai C, Hosen N. Antitumor effects of intracranial injection of B7-H3-targeted Car-T and Car-Nk cells in a patient-derived glioblastoma xenograft model. Cancer Immunol Immunother 2024; 73:256. [PMID: 39367952 PMCID: PMC11456075 DOI: 10.1007/s00262-024-03808-0] [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: 04/01/2024] [Accepted: 08/12/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most lethal primary brain tumor for which novel therapies are needed. Recently, chimeric antigen receptor (CAR) T cell therapy has been shown to be effective against GBM, but it is a personalized medicine and requires high cost and long time for the cell production. CAR-transduced natural killer (NK) cells can be used for "off-the-shelf" cellular immunotherapy because they do not induce graft-versus-host disease. Therefore, we aimed to analyze the anti-GBM effect of CAR-T or NK cells targeting B7-H3, which is known to be highly expressed in GBM. METHODS CAR-T cells targeting B7-H3 were generated using previously reported anti-B7-H3 scFv sequences. Cord blood (CB)-derived NK cells transduced with the B7-H3 CAR were also generated. Their anti-GBM effect was analyzed in vitro. The antitumor effect of intracranial injection of the B7-H3 CAR-T or NK cells was investigated in an in vivo xenograft model with patient-derived GBM cells. RESULTS Both B7-H3 CAR-T cells and CAR-NK cells exhibited marked cytotoxicity against patient-derived GBM cells in vitro. Furthermore, intracranial injection of CAR-T cells and CAR-NK cells targeting B7-H3 resulted in a significant antitumor effect against patient-derived GBM xenografts. CONCLUSION Not only CAR-T cells but also CB-derived CAR-NK cells targeting B7-H3 may have the potential to eliminate GBM cells.
Collapse
Affiliation(s)
- Tetsuro Tachi
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Noriyuki Kijima
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan.
| | - Hideki Kuroda
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Syunya Ikeda
- World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Koki Murakami
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Tomoyoshi Nakagawa
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Moto Yaga
- Department of Respiratory Medicine, Osaka General Hospital, Osaka, Japan
| | - Kanji Nakagawa
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Reina Utsugi
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Ryuichi Hirayama
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Yoshiko Okita
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Naoki Kagawa
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Chihaya Imai
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Naoki Hosen
- World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan.
| |
Collapse
|
26
|
Ghemrawi R, Abuamer L, Kremesh S, Hussien G, Ahmed R, Mousa W, Khoder G, Khair M. Revolutionizing Cancer Treatment: Recent Advances in Immunotherapy. Biomedicines 2024; 12:2158. [PMID: 39335671 PMCID: PMC11429153 DOI: 10.3390/biomedicines12092158] [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: 06/19/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer immunotherapy has emerged as a transformative approach in oncology, utilizing the body's immune system to specifically target and destroy malignant cells. This review explores the scope and impact of various immunotherapeutic strategies, including monoclonal antibodies, chimeric antigen receptor (CAR)-T cell therapy, checkpoint inhibitors, cytokine therapy, and therapeutic vaccines. Monoclonal antibodies, such as Rituximab and Trastuzumab, have revolutionized treatment paradigms for lymphoma and breast cancer by offering targeted interventions that reduce off-target effects. CAR-T cell therapy presents a potentially curative option for refractory hematologic malignancies, although challenges remain in effectively treating solid tumors. Checkpoint inhibitors have redefined the management of cancers like melanoma and lung cancer; however, managing immune-related adverse events and ensuring durable responses are critical areas of focus. Cytokine therapy continues to play a vital role in modulating the immune response, with advancements in cytokine engineering improving specificity and reducing systemic toxicity. Therapeutic vaccines, particularly mRNA-based vaccines, represent a frontier in personalized cancer treatment, aiming to generate robust, long-lasting immune responses against tumor-specific antigens. Despite these advancements, the field faces significant challenges, including immune resistance, tumor heterogeneity, and the immunosuppressive tumor microenvironment. Future research should address these obstacles through emerging technologies, such as next-generation antibodies, Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-based gene editing, and AI-driven drug discovery. By integrating these novel approaches, cancer immunotherapy holds the promise of offering more durable, less toxic, and highly personalized treatment options, ultimately improving patient outcomes and survival rates.
Collapse
Affiliation(s)
- Rose Ghemrawi
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Lama Abuamer
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Sedra Kremesh
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Ghadeer Hussien
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Rahaf Ahmed
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Walaa Mousa
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Ghalia Khoder
- Department of Pharmaceutics and Pharmaceuticals Technology, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates
| |
Collapse
|
27
|
Shi Z, Li Z, Zhang M. Emerging roles of intratumor microbiota in cancer: tumorigenesis and management strategies. J Transl Med 2024; 22:837. [PMID: 39261861 PMCID: PMC11391643 DOI: 10.1186/s12967-024-05640-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
The intricate interplay between the host and its microbiota has garnered increasing attention in the past decade. Specifically, the emerging recognition of microorganisms within diverse cancer tissues, previously presumed sterile, has ignited a resurgence of enthusiasm and research endeavors. Four potential migratory routes have been identified as the sources of intratumoral microbial "dark matter," including direct invasion of mucosal barriers, spreading from normal adjacent tissue, hematogenous spread, and lymphatic drainage, which contribute to the highly heterogeneous features of intratumor microbiota. Importantly, multitudes of studies delineated the roles of intratumor microbiota in cancer initiation and progression, elucidating underlying mechanisms such as genetic alterations, epigenetic modifications, immune dysfunctions, activating oncogenic pathways, and inducing metastasis. With the deepening understanding of intratumoral microbial composition, novel microbiota-based strategies for early cancer diagnosis and prognostic stratification continue to emerge. Furthermore, intratumor microbiota exerts significant influence on the efficacy of cancer therapeutics, particularly immunotherapy, making it an enticing target for intervention in cancer treatment. In this review, we present a comprehensive discussion of the current understanding pertaining to the developmental history, heterogeneous profiles, underlying originations, and carcinogenic mechanisms of intratumor microbiota, and uncover its potential predictive and intervention values, as well as several inevitable challenges as a target for personalized cancer management strategies.
Collapse
Affiliation(s)
- Zhuangzhuang Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, 450000, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, 450000, China.
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, 450000, China.
| |
Collapse
|
28
|
Quinn CH, Julson JR, Markert HR, Nazam N, Butey S, Stewart JE, Coleman JC, Markert JM, Leavenworth JW, Beierle EA. Oncolytic virotherapy augments self-maintaining natural killer cell line cytotoxicity against neuroblastoma. Cancer Immunol Immunother 2024; 73:221. [PMID: 39235531 PMCID: PMC11377387 DOI: 10.1007/s00262-024-03818-y] [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: 05/16/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Neuroblastoma is the most common extracranial solid tumor in children and accounts for 15% of pediatric cancer related deaths. Targeting neuroblastoma with immunotherapies has proven challenging due to a paucity of immune cells in the tumor microenvironment and the release of immunosuppressive cytokines by neuroblastoma tumor cells. We hypothesized that combining an oncolytic Herpes Simplex Virus (oHSV) with natural killer (NK) cells might overcome these barriers and incite tumor cell death. METHODS We utilized MYCN amplified and non-amplified neuroblastoma cell lines, the IL-12 expressing oHSV, M002, and the human NK cell line, NK-92 MI. We assessed the cytotoxicity of NK cells against neuroblastoma with and without M002 infection, the effects of M002 on NK cell priming, and the impact of M002 and priming on the migratory capacity and CD107a expression of NK cells. To test clinical applicability, we then investigated the effects of M002 and NK cells on neuroblastoma in vivo. RESULTS NK cells were more attracted to neuroblastoma cells that were infected with M002. There was an increase in neuroblastoma cell death with the combination treatment of M002 and NK cells both in vitro and in vivo. Priming the NK cells enhanced their cytotoxicity, migratory capacity and CD107a expression. CONCLUSIONS To the best of our knowledge, these investigations are the first to demonstrate the effects of an oncolytic virus combined with self-maintaining NK cells in neuroblastoma and the priming effect of neuroblastoma on NK cells. The current studies provide a deeper understanding of the relation between NK cells and neuroblastoma and these data suggest that oHSV increases NK cell cytotoxicity towards neuroblastoma.
Collapse
Affiliation(s)
- Colin H Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. South, Lowder, Room 300, Birmingham, AL, 35233, UK
| | - Janet R Julson
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. South, Lowder, Room 300, Birmingham, AL, 35233, UK
| | - Hooper R Markert
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. South, Lowder, Room 300, Birmingham, AL, 35233, UK
| | - Nazia Nazam
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. South, Lowder, Room 300, Birmingham, AL, 35233, UK
| | - Swatika Butey
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. South, Lowder, Room 300, Birmingham, AL, 35233, UK
| | - Jerry E Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. South, Lowder, Room 300, Birmingham, AL, 35233, UK
| | - Jennifer C Coleman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Elizabeth A Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. South, Lowder, Room 300, Birmingham, AL, 35233, UK.
| |
Collapse
|
29
|
Wang K, Wang L, Wang Y, Xiao L, Wei J, Hu Y, Wang D, Huang H. Reprogramming natural killer cells for cancer therapy. Mol Ther 2024; 32:2835-2855. [PMID: 38273655 PMCID: PMC11403237 DOI: 10.1016/j.ymthe.2024.01.027] [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: 10/13/2023] [Revised: 01/05/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
The last decade has seen rapid development in the field of cellular immunotherapy, particularly in regard to chimeric antigen receptor (CAR)-modified T cells. However, challenges, such as severe treatment-related toxicities and inconsistent quality of autologous products, have hindered the broader use of CAR-T cell therapy, highlighting the need to explore alternative immune cells for cancer targeting. In this regard, natural killer (NK) cells have been extensively studied in cellular immunotherapy and were found to exert cytotoxic effects without being restricted by human leukocyte antigen and have a lower risk of causing graft-versus-host disease; making them favorable for the development of readily available "off-the-shelf" products. Clinical trials utilizing unedited NK cells or reprogrammed NK cells have shown early signs of their effectiveness against tumors. However, limitations, including limited in vivo persistence and expansion potential, remained. To enhance the antitumor function of NK cells, advanced gene-editing technologies and combination approaches have been explored. In this review, we summarize current clinical trials of antitumor NK cell therapy, provide an overview of innovative strategies for reprogramming NK cells, which include improvements in persistence, cytotoxicity, trafficking and the ability to counteract the immunosuppressive tumor microenvironment, and also discuss some potential combination therapies.
Collapse
Affiliation(s)
- Kexin Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Linqin Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Yiyun Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Lu Xiao
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jieping Wei
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| | - Dongrui Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
30
|
Keshavarz M, Dianat-Moghadam H, Ghorbanhosseini SS, Sarshari B. Oncolytic virotherapy improves immunotherapies targeting cancer stemness in glioblastoma. Biochim Biophys Acta Gen Subj 2024; 1868:130662. [PMID: 38901497 DOI: 10.1016/j.bbagen.2024.130662] [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: 02/11/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Despite advances in cancer therapies, glioblastoma (GBM) remains the most resistant and recurrent tumor in the central nervous system. GBM tumor microenvironment (TME) is a highly dynamic landscape consistent with alteration in tumor infiltration cells, playing a critical role in tumor progression and invasion. In addition, glioma stem cells (GSCs) with self-renewal capability promote tumor recurrence and induce therapy resistance, which all have complicated eradication of GBM with existing therapies. Oncolytic virotherapy is a promising field of therapy that can kill tumor cells in a targeted manner. Manipulated oncolytic viruses (OVs) improve cancer immunotherapy by directly lysis tumor cells, infiltrating antitumor cells, inducing immunogenic cell death, and sensitizing immune-resistant TME to an immune-responsive hot state. Importantly, OVs can target stemness-driven GBM progression. In this review, we will discuss how OVs as a therapeutic option target GBM, especially the GSC subpopulation, and induce immunogenicity to remodel the TME, which subsequently enhances immunotherapies' efficiency.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- Department of Medical Virology, The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran; Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran.
| | - Seyedeh Sara Ghorbanhosseini
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrang Sarshari
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Gujar S, Pol JG, Kumar V, Lizarralde-Guerrero M, Konda P, Kroemer G, Bell JC. Tutorial: design, production and testing of oncolytic viruses for cancer immunotherapy. Nat Protoc 2024; 19:2540-2570. [PMID: 38769145 DOI: 10.1038/s41596-024-00985-1] [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: 05/10/2023] [Accepted: 02/12/2024] [Indexed: 05/22/2024]
Abstract
Oncolytic viruses (OVs) represent a novel class of cancer immunotherapy agents that preferentially infect and kill cancer cells and promote protective antitumor immunity. Furthermore, OVs can be used in combination with established or upcoming immunotherapeutic agents, especially immune checkpoint inhibitors, to efficiently target a wide range of malignancies. The development of OV-based therapy involves three major steps before clinical evaluation: design, production and preclinical testing. OVs can be designed as natural or engineered strains and subsequently selected for their ability to kill a broad spectrum of cancer cells rather than normal, healthy cells. OV selection is further influenced by multiple factors, such as the availability of a specific viral platform, cancer cell permissivity, the need for genetic engineering to render the virus non-pathogenic and/or more effective and logistical considerations around the use of OVs within the laboratory or clinical setting. Selected OVs are then produced and tested for their anticancer potential by using syngeneic, xenograft or humanized preclinical models wherein immunocompromised and immunocompetent setups are used to elucidate their direct oncolytic ability as well as indirect immunotherapeutic potential in vivo. Finally, OVs demonstrating the desired anticancer potential progress toward translation in patients with cancer. This tutorial provides guidelines for the design, production and preclinical testing of OVs, emphasizing considerations specific to OV technology that determine their clinical utility as cancer immunotherapy agents.
Collapse
Affiliation(s)
- Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Jonathan G Pol
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
| | - Vishnupriyan Kumar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Manuela Lizarralde-Guerrero
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
- Ecole Normale Supérieure de Lyon, Lyon, France
| | - Prathyusha Konda
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Guido Kroemer
- INSERM, U1138, Paris, France.
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.
- Université Paris Cité, Paris, France.
- Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France.
- Institut Universitaire de France, Paris, France.
- Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - John C Bell
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada.
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
32
|
Testa U, Castelli G, Pelosi E. CAR-T Cells in the Treatment of Nervous System Tumors. Cancers (Basel) 2024; 16:2913. [PMID: 39199683 PMCID: PMC11352247 DOI: 10.3390/cancers16162913] [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: 07/18/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Chimeric antigen receptor T cells (CAR-Ts) have shown a remarkable efficacy in hematological malignancies but limited responses in solid tumors. Among solid tumors, CAR-T cell therapy has been particularly explored in brain tumors. CAR-T cells have shown a limited clinical efficacy in various types of brain tumors due to several factors that have hampered their activity, including tumor antigen heterogeneity, the limited access of CAR-T cells to brain tumor cells, limited CAR-T cell trafficking and in vivo persistence and the presence of a highly immunosuppressive tumor microenvironment. Despite these considerations, some recent studies have shown promising antitumor activity of GD2-CAR-T cells on diffuse midline gliomas and neuroblastomas and of CARv3-TEAM-E cells in glioblastomas. However, strategies are required to improve the effect of CAR-T cells in brain tumors, including advanced CAR-T cell design with multiple antigenic targeting and incorporation of combination therapies.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (G.C.); (E.P.)
| | | | | |
Collapse
|
33
|
Mai Q, He B, Deng S, Zeng Q, Xu Y, Wang C, Pang Y, Zhang S, Li J, Zeng J, Huang L, Fu Y, Li C, Li T, Xu X, Zhang L. Efficacy of NKG2D CAR-T cells with IL-15/IL-15Rα signaling for treating Epstein-Barr virus-associated lymphoproliferative disorder. Exp Hematol Oncol 2024; 13:85. [PMID: 39160631 PMCID: PMC11334566 DOI: 10.1186/s40164-024-00553-z] [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: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024] Open
Abstract
Epstein-Barr virus (EBV) related post-transplant lymphoproliferative disorder (EBV-PTLD) is a life-threatening complication after hematopoietic stem cell transplantation (HSCT) or solid organ transplantation (SOT), for which no standard therapeutic means have been developed. Significant increase expression of natural killer group 2 member D ligands (NKG2DLs) was observed on B-lymphoblastoid cells of EBV-PTLD, indicating NKG2DLs as potential therapeutic targets for treatment of EBV-PTLD. In this study, the recombinant constructs of NKG2D CAR and IL-15/IL-15Rα-NKG2D CAR were generated with a retroviral vector and then transduced to human T cells to produce NKG2D CAR-T and IL-15/IL-15Rα-NKG2D CAR-T cells, respectively. B-lymphoblastoid cell lines (B-LCLs) and the xenografted mouse models were established to evaluate the efficacy of these CAR-T cells. IL-15/IL-15Rα-NKG2D CAR-T cells exhibited superior proliferation and antigen-specific cytotoxic effect compared to NKG2D CAR-T, as IL-15/IL-15Rα signaling promoted the expansion of less differentiated central memory T cells (TCM) and increased expression of CD107a and IFN-γ. Moreover, EBV DNA load was dramatically reduced, and 80% B-LCL cells were eliminated by IL-15/IL-15Rα-NKG2D CAR-T cells after co-culturing. In-vivo study confirmed that IL-15/IL-15Rα-NKG2D CAR-T cell therapy significantly enhanced antiviral efficacy in mice, as the serum load of EBV after IL-15/IL-15Rα-NKG2D CAR-T cell infusion was 1500 times lower than the untreated control (P < 0.001). The enhanced efficacy of IL-15/IL-15Rα-NKG2D CAR T cells was probably due to the IL-15/IL-15Rα signaling improved homing and persistence of NKG2D CAR-T cells in vivo, and increased the production of IFN-γ, Perforin, and Granulysin. In conclusion, NKG2D CAR-T cells co-expressing IL-15/IL-15Rα promoted the central memory CAR T cell proliferation and improved the homing and persistence of CAR T cells in vivo, resulting in enhanced anti-tumor and anti-viral effects in treating EBV-PTLD.
Collapse
Affiliation(s)
- Qiusui Mai
- Department of Blood Transfusion, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Bailin He
- Department of Hematology, Nanfang Hospital, Southern Medical Universit, Guangzhou, 510515, China
| | - Shikai Deng
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Qing Zeng
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Yanwen Xu
- Department of Obstetrics, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, 511402, China
| | - Cong Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou, 510555, China
| | - Yunyi Pang
- Department of Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Sheng Zhang
- Shenzhen Bao'an District Central Blood Station, Shenzhen, 518101, China
| | - Jinfeng Li
- Shenzhen Bao'an District Central Blood Station, Shenzhen, 518101, China
| | | | - Liqin Huang
- Shenzhen Blood Center, Shenzhen, 518035, China
| | - Yongshui Fu
- Guangzhou Blood Center, Guangzhou, 510095, China
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China.
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou, 510555, China.
| | - Tingting Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China.
- Shenzhen Bao'an District Central Blood Station, Shenzhen, 518101, China.
| | - Xiaojun Xu
- Department of Blood Transfusion, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
34
|
Mohammad Taheri M, Javan F, Poudineh M, Athari SS. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 2024; 22:736. [PMID: 39103889 PMCID: PMC11302387 DOI: 10.1186/s12967-024-05534-8] [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: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for "off-the-shelf" production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Shamseddin Athari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, Zanjan School of Medicine, Zanjan University of Medical Sciences, 12th Street, Shahrake Karmandan, Zanjan, 45139-561111, Iran.
| |
Collapse
|
35
|
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
|
36
|
Gao X, Liu J, Sun R, Zhang J, Cao X, Zhang Y, Zhao M. Alliance between titans: combination strategies of CAR-T cell therapy and oncolytic virus for the treatment of hematological malignancies. Ann Hematol 2024; 103:2569-2589. [PMID: 37853078 DOI: 10.1007/s00277-023-05488-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
There have been several clinical studies using chimeric antigen receptor (CAR)-T cell therapy for different hematological malignancies. It has transformed the therapy landscape for hematologic malignancies dramatically. Nonetheless, in acute myeloid leukemia (AML) and T cell malignancies, it still has a dismal prognosis. Even in the most promising locations, recurrence with CAR-T treatment remains a big concern. Oncolytic viruses (OVs) can directly lyse tumor cells or cause immune responses, and they can be manipulated to create therapeutic proteins, increasing anticancer efficacy. Oncolytic viruses have been proven in a rising number of studies to be beneficial in hematological malignancies. There are limitations that cannot be avoided by using either treatment alone, and the combination of CAR-T cell therapy and oncolytic virus therapy may complement the disadvantages of individual application, enhance the advantages of their respective treatment methods and improve the treatment effect. The alternatives for combining two therapies in hematological malignancies are discussed in this article.
Collapse
Affiliation(s)
- Xuejin Gao
- Emergency, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Jile Liu
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Rui Sun
- Nankai University School of Medicine, Tianjin, 300192, China
| | - Jingkun Zhang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xinping Cao
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Yi Zhang
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
37
|
Qi X. Advances in antitumour therapy with oncolytic herpes simplex virus combinations. Discov Oncol 2024; 15:302. [PMID: 39046631 PMCID: PMC11269532 DOI: 10.1007/s12672-024-01165-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Oncolytic Virus (OVs) is an emerging approach to tumour immunity that allows the use of natural or genetically modified viruses to specifically infect and lyse tumour cells without damaging normal cells. Oncolytic herpes simplex virus (oHSV) is one of the more widely researched and applied OVs in the field of oncology, which can directly kill tumour cells to promote anti-tumour immune responses. oHSV is one of the few viruses with good antiviral drugs, so oHSV is also more clinically safe. In recent years, in addition to monotherapy of oHSV in tumours, more and more studies have been devoted to exploring the anti-tumour effects of oHSV in combination with other therapeutic approaches. In this article we describe the progress of oHSV combination therapy against tumours in the nervous system, digestive system, reproductive system and other systems.
Collapse
Affiliation(s)
- Xuejiao Qi
- College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China.
| |
Collapse
|
38
|
Yuan Z, Zhang Y, Wang X, Wang X, Ren S, He X, Su J, Zheng A, Guo S, Chen Y, Deng S, Wu X, Li M, Du F, Zhao Y, Shen J, Wang Z, Xiao Z. The investigation of oncolytic viruses in the field of cancer therapy. Front Oncol 2024; 14:1423143. [PMID: 39055561 PMCID: PMC11270537 DOI: 10.3389/fonc.2024.1423143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Oncolytic viruses (OVs) have emerged as a potential strategy for tumor treatment due to their ability to selectively replicate in tumor cells, induce apoptosis, and stimulate immune responses. However, the therapeutic efficacy of single OVs is limited by the complexity and immunosuppressive nature of the tumor microenvironment (TME). To overcome these challenges, engineering OVs has become an important research direction. This review focuses on engineering methods and multi-modal combination therapies for OVs aimed at addressing delivery barriers, viral phagocytosis, and antiviral immunity in tumor therapy. The engineering approaches discussed include enhancing in vivo immune response, improving replication efficiency within the tumor cells, enhancing safety profiles, and improving targeting capabilities. In addition, this review describes the potential mechanisms of OVs combined with radiotherapy, chemotherapy, cell therapy and immune checkpoint inhibitors (ICIs), and summarizes the data of ongoing clinical trials. By continuously optimizing engineering strategies and combination therapy programs, we can achieve improved treatment outcomes and quality of life for cancer patients.
Collapse
Affiliation(s)
- Zijun Yuan
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anfu Zheng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Sipeng Guo
- Research And Experiment Center, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Zechen Wang
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
| | - Zhangang Xiao
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| |
Collapse
|
39
|
Zhong Y, Liu J. Emerging roles of CAR-NK cell therapies in tumor immunotherapy: current status and future directions. Cell Death Discov 2024; 10:318. [PMID: 38987565 PMCID: PMC11236993 DOI: 10.1038/s41420-024-02077-1] [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: 03/29/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024] Open
Abstract
Cancer immunotherapy harnesses the body's immune system to combat malignancies, building upon an understanding of tumor immunosurveillance and immune evasion mechanisms. This therapeutic approach reactivates anti-tumor immune responses and can be categorized into active, passive, and combined immunization strategies. Active immunotherapy engages the immune system to recognize and attack tumor cells by leveraging host immunity with cytokine supplementation or vaccination. Conversely, passive immunotherapy employs exogenous agents, such as monoclonal antibodies (anti-CTLA4, anti-PD1, anti-PD-L1) or adoptive cell transfers (ACT) with genetically engineered chimeric antigen receptor (CAR) T or NK cells, to exert anti-tumor effects. Over the past decades, CAR-T cell therapies have gained significant traction in oncological treatment, offering hope through their targeted approach. However, the potential adverse effects associated with CAR-T cells, including cytokine release syndrome (CRS), off-tumor toxicity, and neurotoxicity, warrant careful consideration. Recently, CAR-NK cell therapy has emerged as a promising alternative in the landscape of tumor immunotherapy, distinguished by its innate advantages over CAR-T cell modalities. In this review, we will synthesize the latest research and clinical advancements in CAR-NK cell therapies. We will elucidate the therapeutic benefits of employing CAR-NK cells in oncology and critically examine the developmental bottlenecks impeding their broader application. Our discussion aims to provide a comprehensive overview of the current status and future potential of CAR-NK cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Yan Zhong
- Department of Pathology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jingfeng Liu
- Shenzhen Key Laboratory of Immunity and Inflammatory Diseases, Peking University Shenzhen Hospital, Shenzhen, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China.
| |
Collapse
|
40
|
Franks ML, An JH, Leavenworth JW. The Role of Natural Killer Cells in Oncolytic Virotherapy: Friends or Foes? Vaccines (Basel) 2024; 12:721. [PMID: 39066359 PMCID: PMC11281503 DOI: 10.3390/vaccines12070721] [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: 05/31/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Oncolytic virotherapy (OVT) has emerged as a promising cancer immunotherapy, and is capable of potentiating other immunotherapies due to its capacity to increase tumor immunogenicity and to boost host antitumor immunity. Natural killer (NK) cells are a critical cellular component for mediating the antitumor response, but hold a mixed reputation for their role in mediating the therapeutic efficacy of OVT. This review will discuss the pros and cons of how NK cells impact OVT, and how to harness this knowledge for the development of effective strategies that could modulate NK cells to improve OVT-based therapeutic outcomes.
Collapse
Affiliation(s)
- Michael L. Franks
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.L.F.)
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ju-Hyun An
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.L.F.)
| | - Jianmei W. Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.L.F.)
- The O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
41
|
Shah Z, Tian L, Li Z, Jin L, Zhang J, Li Z, Barr T, Tang H, Feng M, Caligiuri MA, Yu J. Human anti-PSCA CAR macrophages possess potent antitumor activity against pancreatic cancer. Cell Stem Cell 2024; 31:803-817.e6. [PMID: 38663406 PMCID: PMC11162318 DOI: 10.1016/j.stem.2024.03.018] [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: 03/10/2023] [Revised: 01/11/2024] [Accepted: 03/28/2024] [Indexed: 05/15/2024]
Abstract
Due to the limitations of autologous chimeric antigen receptor (CAR)-T cells, alternative sources of cellular immunotherapy, including CAR macrophages, are emerging for solid tumors. Human induced pluripotent stem cells (iPSCs) offer an unlimited source for immune cell generation. Here, we develop human iPSC-derived CAR macrophages targeting prostate stem cell antigen (PSCA) (CAR-iMacs), which express membrane-bound interleukin (IL)-15 and truncated epidermal growth factor receptor (EGFR) for immune cell activation and a suicide switch, respectively. These allogeneic CAR-iMacs exhibit strong antitumor activity against human pancreatic solid tumors in vitro and in vivo, leading to reduced tumor burden and improved survival in a pancreatic cancer mouse model. CAR-iMacs appear safe and do not exhibit signs of cytokine release syndrome or other in vivo toxicities. We optimized the cryopreservation of CAR-iMac progenitors that remain functional upon thawing, providing an off-the-shelf, allogeneic cell product that can be developed into CAR-iMacs. Overall, our preclinical data strongly support the potential clinical translation of this human iPSC-derived platform for solid tumors, including pancreatic cancer.
Collapse
Affiliation(s)
- Zahir Shah
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Lei Tian
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Zhixin Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Lewei Jin
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Zhenlong Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Tasha Barr
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Hejun Tang
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Mingye Feng
- Department of Immuno-Oncology, City of Hope, Los Angeles, CA 91010, USA
| | - Michael A Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; City of Hope Comprehensive Cancer Center, Los Angeles, CA 91010, USA.
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Department of Immuno-Oncology, City of Hope, Los Angeles, CA 91010, USA; City of Hope Comprehensive Cancer Center, Los Angeles, CA 91010, USA.
| |
Collapse
|
42
|
Li X, Li W, Xu L, Song Y. Chimeric antigen receptor-immune cells against solid tumors: Structures, mechanisms, recent advances, and future developments. Chin Med J (Engl) 2024; 137:1285-1302. [PMID: 37640679 PMCID: PMC11191032 DOI: 10.1097/cm9.0000000000002818] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Indexed: 08/31/2023] Open
Abstract
ABSTRACT The advent of chimeric antigen receptor (CAR)-T cell immunotherapies has led to breakthroughs in the treatment of hematological malignancies. However, their success in treating solid tumors has been limited. CAR-natural killer (NK) cells have several advantages over CAR-T cells because NK cells can be made from pre-existing cell lines or allogeneic NK cells with a mismatched major histocompatibility complex (MHC), which means they are more likely to become an "off-the-shelf" product. Moreover, they can kill cancer cells via CAR-dependent/independent pathways and have limited toxicity. Macrophages are the most malleable immune cells in the body. These cells can efficiently infiltrate into tumors and are present in large numbers in tumor microenvironments (TMEs). Importantly, CAR-macrophages (CAR-Ms) have recently yielded exciting preclinical results in several solid tumors. Nevertheless, CAR-T, CAR-NK, and CAR-M all have their own advantages and limitations. In this review, we systematically discuss the current status, progress, and the major hurdles of CAR-T cells, CAR-NK cells, and CAR-M as they relate to five aspects: CAR structure, therapeutic mechanisms, the latest research progress, current challenges and solutions, and comparison according to the existing research in order to provide a reasonable option for treating solid tumors in the future.
Collapse
Affiliation(s)
- Xudong Li
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Linping Xu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Yongping Song
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
43
|
Huang M, Liu Y, Yan Q, Peng M, Ge J, Mo Y, Wang Y, Wang F, Zeng Z, Li Y, Fan C, Xiong W. NK cells as powerful therapeutic tool in cancer immunotherapy. Cell Oncol (Dordr) 2024; 47:733-757. [PMID: 38170381 DOI: 10.1007/s13402-023-00909-3] [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] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Natural killer (NK) cells have gained considerable attention and hold great potential for their application in tumor immunotherapy. This is mainly due to their MHC-unrestricted and pan-specific recognition capabilities, as well as their ability to rapidly respond to and eliminate target cells. To artificially generate therapeutic NK cells, various materials can be utilized, such as peripheral blood mononuclear cells (PBMCs), umbilical cord blood (UCB), induced pluripotent stem cells (iPSCs), and NK cell lines. Exploiting the therapeutic potential of NK cells to treat tumors through in vivo and in vitro therapeutic modalities has yielded positive therapeutic results. CONCLUSION This review provides a comprehensive description of NK cell therapeutic approaches for tumors and discusses the current problems associated with these therapeutic approaches and the prospects of NK cell therapy for tumors.
Collapse
Affiliation(s)
- Mao Huang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yixuan Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Miao Peng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Junshang Ge
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Fuyan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Comprehensive Cancer Center, Baylor College of Medicine, Alkek Building, RM N720, Houston, TX, USA
| | - Chunmei Fan
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, 410013, Changsha, Hunan Province, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
44
|
Wang H, Borlongan M, Kaufman HL, Le U, Nauwynck HJ, Rabkin SD, Saha D. Cytokine-armed oncolytic herpes simplex viruses: a game-changer in cancer immunotherapy? J Immunother Cancer 2024; 12:e008025. [PMID: 38821716 PMCID: PMC11149157 DOI: 10.1136/jitc-2023-008025] [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] [Accepted: 05/13/2024] [Indexed: 06/02/2024] Open
Abstract
Cytokines are small proteins that regulate the growth and functional activity of immune cells, and several have been approved for cancer therapy. Oncolytic viruses are agents that mediate antitumor activity by directly killing tumor cells and inducing immune responses. Talimogene laherparepvec is an oncolytic herpes simplex virus type 1 (oHSV), approved for the treatment of recurrent melanoma, and the virus encodes the human cytokine, granulocyte-macrophage colony-stimulating factor (GM-CSF). A significant advantage of oncolytic viruses is the ability to deliver therapeutic payloads to the tumor site that can help drive antitumor immunity. While cytokines are especially interesting as payloads, the optimal cytokine(s) used in oncolytic viruses remains controversial. In this review, we highlight preliminary data with several cytokines and chemokines, including GM-CSF, interleukin 12, FMS-like tyrosine kinase 3 ligand, tumor necrosis factor α, interleukin 2, interleukin 15, interleukin 18, chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 5, chemokine (C-X-C motif) ligand 4, or their combinations, and show how these payloads can further enhance the antitumor immunity of oHSV. A better understanding of cytokine delivery by oHSV can help improve clinical benefit from oncolytic virus immunotherapy in patients with cancer.
Collapse
Affiliation(s)
- Hongbin Wang
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, California, USA
- College of Graduate Studies, California Northstate University, Elk Grove, California, USA
| | - Mia Borlongan
- College of Graduate Studies, California Northstate University, Elk Grove, California, USA
| | - Howard L Kaufman
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Ankyra Therapeutics, Inc, Cambridge, Massachusetts, USA
| | - Uyen Le
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, California, USA
| | - Hans J Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Samuel D Rabkin
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dipongkor Saha
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, California, USA
| |
Collapse
|
45
|
Bugakova AS, Chudakova DA, Myzina MS, Yanysheva EP, Ozerskaya IV, Soboleva AV, Baklaushev VP, Yusubalieva GM. Non-Tumor Cells within the Tumor Microenvironment-The "Eminence Grise" of the Glioblastoma Pathogenesis and Potential Targets for Therapy. Cells 2024; 13:808. [PMID: 38786032 PMCID: PMC11119139 DOI: 10.3390/cells13100808] [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: 04/04/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignancy of the central nervous system in adults. GBM has high levels of therapy failure and its prognosis is usually dismal. The phenotypic heterogeneity of the tumor cells, dynamic complexity of non-tumor cell populations within the GBM tumor microenvironment (TME), and their bi-directional cross-talk contribute to the challenges of current therapeutic approaches. Herein, we discuss the etiology of GBM, and describe several major types of non-tumor cells within its TME, their impact on GBM pathogenesis, and molecular mechanisms of such an impact. We also discuss their value as potential therapeutic targets or prognostic biomarkers, with reference to the most recent works on this subject. We conclude that unless all "key player" populations of non-tumor cells within the TME are considered, no breakthrough in developing treatment for GBM can be achieved.
Collapse
Affiliation(s)
- Aleksandra S. Bugakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Daria A. Chudakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Maria S. Myzina
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Elvira P. Yanysheva
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Iuliia V. Ozerskaya
- Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Alesya V. Soboleva
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
46
|
Vazaios K, van Berkum RE, Calkoen FG, van der Lugt J, Hulleman E. OV Modulators of the Paediatric Brain TIME: Current Status, Combination Strategies, Limitations and Future Directions. Int J Mol Sci 2024; 25:5007. [PMID: 38732225 PMCID: PMC11084613 DOI: 10.3390/ijms25095007] [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: 03/12/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Oncolytic viruses (OVs) are characterised by their preference for infecting and replicating in tumour cells either naturally or after genetic modification, resulting in oncolysis. Furthermore, OVs can elicit both local and systemic anticancer immune responses while specifically infecting and lysing tumour cells. These characteristics render them a promising therapeutic approach for paediatric brain tumours (PBTs). PBTs are frequently marked by a cold tumour immune microenvironment (TIME), which suppresses immunotherapies. Recent preclinical and clinical studies have demonstrated the capability of OVs to induce a proinflammatory immune response, thereby modifying the TIME. In-depth insights into the effect of OVs on different cell types in the TIME may therefore provide a compelling basis for using OVs in combination with other immunotherapy modalities. However, certain limitations persist in our understanding of oncolytic viruses' ability to regulate the TIME to enhance anti-tumour activity. These limitations primarily stem from the translational limitations of model systems, the difficulties associated with tracking reliable markers of efficacy throughout the course of treatment and the role of pre-existing viral immunity. In this review, we describe the different alterations observed in the TIME in PBTs due to OV treatment, combination therapies of OVs with different immunotherapies and the hurdles limiting the development of effective OV therapies while suggesting future directions based on existing evidence.
Collapse
Affiliation(s)
| | | | | | | | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (K.V.); (F.G.C.); (J.v.d.L.)
| |
Collapse
|
47
|
Kong D, Kwon D, Moon B, Kim DH, Kim MJ, Choi J, Kang KS. CD19 CAR-expressing iPSC-derived NK cells effectively enhance migration and cytotoxicity into glioblastoma by targeting to the pericytes in tumor microenvironment. Biomed Pharmacother 2024; 174:116436. [PMID: 38508081 DOI: 10.1016/j.biopha.2024.116436] [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: 10/12/2023] [Revised: 03/07/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
In cancer immunotherapy, chimeric antigen receptors (CARs) targeting specific antigens have become a powerful tool for cell-based therapy. CAR-natural killer (NK) cells offer selective anticancer lysis with reduced off-tumor toxicity compared to CAR-T cells, which is beneficial in the heterogeneous milieu of solid tumors. In the tumor microenvironment (TME) of glioblastoma (GBM), pericytes not only support tumor growth but also contribute to immune evasion, underscoring their potential as therapeutic targets in GBM treatment. Given this context, our study aimed to target the GBM TME, with a special focus on pericytes expressing CD19, to evaluate the potential effectiveness of CD19 CAR-iNK cells against GBM. We performed CD19 CAR transduction in induced pluripotent stem cell-derived NK (iNK) cells. To determine whether CD19 CAR targets the TME pericytes in GBM, we developed GBM-blood vessel assembloids (GBVA) by fusing GBM spheroids with blood vessel organoids. When co-cultured with GBVA, CD19 CAR-iNK cells migrated towards the pericytes surrounding the GBM. Using a microfluidic chip, we demonstrated CD19 CAR-iNK cells' targeted action and cytotoxic effects in a perfusion-like environment. GBVA xenografts recapitulated the TME including human CD19-positive pericytes, thereby enabling the application of an in vivo model for validating the efficacy of CD19 CAR-iNK cells against GBM. Compared to GBM spheroids, the presence of pericytes significantly enhanced CD19 CAR-iNK cell migration towards GBM and reduced proliferation. These results underline the efficacy of CD19 CAR-iNK cells in targeting pericytes within the GBM TME, suggesting their potential therapeutic value for GBM treatment.
Collapse
Affiliation(s)
- Dasom Kong
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Daekee Kwon
- Research Institute in Maru Therapeutics, Seoul 05854, Republic of Korea
| | - Bokyung Moon
- Research Institute in Maru Therapeutics, Seoul 05854, Republic of Korea
| | - Da-Hyun Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Department of Biotechnology, Sungshin Women's University, Seoul 01133, Republic of Korea
| | - Min-Ji Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jungju Choi
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
48
|
Wang Y, Zhu M, Chi H, Liu Y, Yu G. The combination therapy of oncolytic virotherapy. Front Pharmacol 2024; 15:1380313. [PMID: 38725667 PMCID: PMC11079273 DOI: 10.3389/fphar.2024.1380313] [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: 02/01/2024] [Accepted: 04/04/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction: Compared to other cancer immunotherapies, oncolytic viruses possess several advantages, including high killing efficiency, excellent targeting capabilities, minimal adverse reactions, and multiple pathways for tumor destruction. However, the efficacy of oncolytic viruses as a monotherapy often falls short of expectations. Consequently, combining oncolytic viruses with traditional treatments to achieve synergistic effects has emerged as a promising direction for the development of oncolytic virus therapies. Methods: This article provides a comprehensive review of the current progress in preclinical and clinical trials exploring the combination therapies involving oncolytic viruses. Results: Specifically, we discuss the combination of oncolytic viruses with immune checkpoint inhibitors, chemotherapy, targeted therapy, and cellular therapy. Discussion: The aim of this review is to offer valuable insights and references for the further advancement of these combination strategies in clinical applications. Further research is necessary to refine the design of combination therapies and explore novel strategies to maximize the therapeutic benefits offered by oncolytic viruses.
Collapse
Affiliation(s)
- Yue Wang
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Mengying Zhu
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
- Department of Clinical Integration of Traditional Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Huanyu Chi
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
- The Second Clinical College of Dalian Medical University, Dalian, China
| | - Yang Liu
- Department of Ophthalmology, First Hospital of China Medical University, Shenyang, China
| | - Guilin Yu
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
49
|
Xu MY, Zeng N, Liu CQ, Sun JX, An Y, Zhang SH, Xu JZ, Zhong XY, Ma SY, He HD, Hu J, Xia QD, Wang SG. Enhanced cellular therapy: revolutionizing adoptive cellular therapy. Exp Hematol Oncol 2024; 13:47. [PMID: 38664743 PMCID: PMC11046957 DOI: 10.1186/s40164-024-00506-6] [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: 07/13/2023] [Accepted: 03/31/2024] [Indexed: 04/28/2024] Open
Abstract
Enhanced cellular therapy has emerged as a novel concept following the basis of cellular therapy. This treatment modality applied drugs or biotechnology to directly enhance or genetically modify cells to enhance the efficacy of adoptive cellular therapy (ACT). Drugs or biotechnology that enhance the killing ability of immune cells include immune checkpoint inhibitors (ICIs) / antibody drugs, small molecule inhibitors, immunomodulatory factors, proteolysis targeting chimera (PROTAC), oncolytic virus (OV), etc. Firstly, overcoming the inhibitory tumor microenvironment (TME) can enhance the efficacy of ACT, which can be achieved by blocking the immune checkpoint. Secondly, cytokines or cytokine receptors can be expressed by genetic engineering or added directly to adoptive cells to enhance the migration and infiltration of adoptive cells to tumor cells. Moreover, multi-antigen chimeric antigen receptors (CARs) can be designed to enhance the specific recognition of tumor cell-related antigens, and OVs can also stimulate antigen release. In addition to inserting suicide genes into adoptive cells, PROTAC technology can be used as a safety switch or degradation agent of immunosuppressive factors to enhance the safety and efficacy of adoptive cells. This article comprehensively summarizes the mechanism, current situation, and clinical application of enhanced cellular therapy, describing potential improvements to adoptive cellular therapy.
Collapse
Affiliation(s)
- Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Na Zeng
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Si-Han Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Xing-Yu Zhong
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Si-Yang Ma
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Hao-Dong He
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jia Hu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
50
|
Kong JC, Sa’ad MA, Vijayan HM, Ravichandran M, Balakrishnan V, Tham SK, Tye GJ. Chimeric antigen receptor-natural killer cell therapy: current advancements and strategies to overcome challenges. Front Immunol 2024; 15:1384039. [PMID: 38726000 PMCID: PMC11079817 DOI: 10.3389/fimmu.2024.1384039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Chimeric antigen receptor-natural killer (CAR-NK) cell therapy is a novel immunotherapy targeting cancer cells via the generation of chimeric antigen receptors on NK cells which recognize specific cancer antigens. CAR-NK cell therapy is gaining attention nowadays owing to the ability of CAR-NK cells to release potent cytotoxicity against cancer cells without side effects such as cytokine release syndrome (CRS), neurotoxicity and graft-versus-host disease (GvHD). CAR-NK cells do not require antigen priming, thus enabling them to be used as "off-the-shelf" therapy. Nonetheless, CAR-NK cell therapy still possesses several challenges in eliminating cancer cells which reside in hypoxic and immunosuppressive tumor microenvironment. Therefore, this review is envisioned to explore the current advancements and limitations of CAR-NK cell therapy as well as discuss strategies to overcome the challenges faced by CAR-NK cell therapy. This review also aims to dissect the current status of clinical trials on CAR-NK cells and future recommendations for improving the effectiveness and safety of CAR-NK cell therapy.
Collapse
Affiliation(s)
- Jun Chang Kong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Mohammad Auwal Sa’ad
- Celestialab Sdn Bhd, Kuala Lumpur, Malaysia
- Department of Biotechnology, Faculty of Applied Sciences, AIMST University, Bedong, Kedah, Malaysia
| | | | - Manickam Ravichandran
- Department of Biotechnology, Faculty of Applied Sciences, AIMST University, Bedong, Kedah, Malaysia
- MyGenome, ALPS Global Holding, Kuala Lumpur, Malaysia
| | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Seng Kong Tham
- ALPS Medical Centre, ALPS Global Holding, Kuala Lumpur, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| |
Collapse
|