1
|
Ahmad GV, Nouri S, Mohammad Gholian A, Abdollahi E, Ghorbaninezhad F, Tahmasebi S, Eterafi M, Askari MR, Safarzadeh E. Breaking barriers: CAR-NK cell therapy breakthroughs in female-related cancers. Biomed Pharmacother 2025; 187:118071. [PMID: 40253831 DOI: 10.1016/j.biopha.2025.118071] [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: 01/16/2025] [Revised: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025] Open
Abstract
Cancer stands as a leading cause of mortality globally. The main female-related malignancies are breast cancer, with 2.3 million new cases annually, and ovarian cancer, with 300,000 new cases per year worldwide. The current treatments like surgery, chemotherapy, and radiation therapy have presumably had deficiencies in sustaining long-term anti-tumor responses. Cellular immunotherapy, also referred to as adoptive cell therapy, has shown encouraging advances by employing genetically modified immune cells in fighting cancer by engineering chimeric antigen receptors (CARs) mainly on T cells and natural killer (NK) cells. Studies in NK cell therapies involve unmodified NK cells and CAR-NK cell therapies, targeting cancer cells while limiting the destruction of normal cells. CAR-NK cells represent the next generation of therapeutic immune cells that have been shown to eliminate malignancies through CAR-dependent and CAR-independent mechanisms. They also represent possible candidates for "off-the-shelf" therapies due to their advantages, including the ability to target cancer cells independently of the major histocompatibility complex, reduced risk of alloreactivity, and fewer severe toxicities compared to CAR-T cells. To date, there have been no comprehensive review studies examining the therapeutic potential of CAR-NK cell therapy specifically for female-related malignancies, such as breast and ovarian cancers. This review offers a thorough exploration of CAR-NK cell therapy in relation to these cancers and their responses to treatment.
Collapse
Affiliation(s)
- Ghorbani Vanan Ahmad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Samaneh Nouri
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Eileen Abdollahi
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farid Ghorbaninezhad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Safa Tahmasebi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Askari
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
2
|
Lv M, Wang Y, Yuan Z, Zhai L, Iqbal H, Ur-Rehman U, Ning X, Wei H, Xin J, Jin Z, Yi Z, Wang B, Chen W, Xiao R. Decitabine promotes the differentiation of poorly differentiated gastric cancer cells and enhances the sensitivity of NK cell cytotoxicity via TNF-α. Sci Rep 2025; 15:13119. [PMID: 40240368 PMCID: PMC12003911 DOI: 10.1038/s41598-025-95741-0] [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: 09/23/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Poorly differentiated gastric cancer (PDGC) is characterized by high invasiveness, rapid progression, and poor prognosis for patients. Differentiation therapy has long been a promising approach by manipulating the differentiation state of tumor cells to inhibit tumor growth, offering fewer side effects. Decitabine (DAC), is known as an inhibitor of DNA methylation, thus reactivating the transcription of previously methylated silenced genes associated with differentiation to induce a more differentiated state. This study used the differentiation-inducing agents DAC to treat two PDGC cell lines, MKN45 and NUGC4, and explored the impact of DAC on cell proliferation and influence of their sensitivity to Natural Killer cells (NK cells) mediated cytotoxicity. The results demonstrated a significant reduction in cell proliferation, migration, and invasion without affecting cell viability after DAC treatment. Additionally, transcriptomic analysis revealed that DAC-treated PDGC cells upregulated multiple immune-related genes, including the gene encoding for tumor necrosis factor alpha (TNF-α). Co-culture study of NK cells and PDGC cells showed that DAC treatment enhanced the sensitivity of these cancer cells to NK cell-mediated cytotoxicity, and TNF-α played a crucial role in promoting NK cell cytotoxicity. Following the subcutaneous implantation of tumors in nude mice, DAC administration significantly inhibited the growth of PDGC tumors and induced the upregulation of differentiation related genes. In summary, DAC effectively reduces the malignant characteristics of the PDGC cells by promoting their transition towards a higher state of differentiation and enhancing their sensitivity to NK cell-mediated killing, providing new insights for the mechanisms of the antitumor effects of DAC.
Collapse
Affiliation(s)
- Man Lv
- School of Life Science, Tianjin University, Tianjin, 300072, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yue Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Medical College of Tianjin University, Tianjin University, Tianjin, 300072, China
| | - Ziyin Yuan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Lina Zhai
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Haroon Iqbal
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Uzair Ur-Rehman
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xin Ning
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Huiying Wei
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, 310024, China
| | - Jun Xin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Medical College of Tianjin University, Tianjin University, Tianjin, 300072, China
| | - Zihui Jin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Zhou Yi
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Baichuan Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Wangkai Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Run Xiao
- School of Life Science, Tianjin University, Tianjin, 300072, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
3
|
Li W, Liu N, Chen M, Liu D, Liu S. Metformin as an immunomodulatory agent in enhancing head and neck squamous cell carcinoma therapies. Biochim Biophys Acta Rev Cancer 2025; 1880:189262. [PMID: 39827973 DOI: 10.1016/j.bbcan.2025.189262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a significant clinical challenge due to its aggressive behavior and poor prognosis, making the development of novel therapeutics with enhanced efficacy and minimal side effects critical. Metformin, a widely used antidiabetic agent, has recently emerged as a potential adjunctive therapy for HNSCC, exhibiting both direct anti-tumor and immunomodulatory effects. This review comprehensively explores the multifaceted role of metformin in shaping the tumor immune microenvironment within HNSCC. We emphasize its pivotal role in modulating immune cell populations and its potential for synergistic action with immunotherapeutic strategies. Furthermore, we address the current challenges associated with optimizing dosing regimens, identifying predictive biomarkers, and integrating metformin with immunotherapy. By dissecting these aspects, this review aims to pave the way for the development of personalized HNSCC treatment strategies that fully exploit the therapeutic potential of metformin.
Collapse
Affiliation(s)
- Wenting Li
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China
| | - Nanshu Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China
| | - Mingwei Chen
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China
| | - Dongjuan Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China.
| | - Sai Liu
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China.
| |
Collapse
|
4
|
Bindu S, Bibi R, Pradeep R, Sarkar K. The evolving role of B cells in malignancies. Hum Immunol 2025; 86:111301. [PMID: 40132250 DOI: 10.1016/j.humimm.2025.111301] [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: 08/06/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
B cells play diverse roles in different pathological circumstances, such as neoplastic diseases, autoimmune disorders, and neurological maladies. B cells, which are essential elements of the adaptive immune system, demonstrate exceptional functional variety, including the generation of antibodies, the presentation of antigens, and the secretion of cytokines. Within the field of oncology, B cells display a multifaceted nature in the tumor microenvironment, simultaneously manifesting both tumor-promoting and tumor-suppressing characteristics. Studies have found that the existence of tertiary lymphoid structures, which consist of B cells, is linked to better survival rates in different types of cancers. This article examines the involvement of B cells in different types of malignancies, emphasizing their importance in the development of the diseases and their potential as biomarkers. Additionally, the review also examines the crucial role of B cells in autoimmune illnesses and their potential as targets for therapy. The article also analyses the role of B cells in immunization and exploring their potential uses in cancer immunotherapy. This analysis highlights the intricate and occasionally contradictory roles of B cells, underlining the necessity for additional research to clarify their varied actions in various illness scenarios.
Collapse
Affiliation(s)
- Soham Bindu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - Roshni Bibi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - R Pradeep
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India.
| |
Collapse
|
5
|
Sabahi M, Fathi Jouzdani A, Sadeghian Z, Dabbagh Ohadi MA, Sultan H, Salehipour A, Maniakhina L, Rezaei N, Adada B, Mansouri A, Borghei-Razavi H. CAR-engineered NK cells versus CAR T cells in treatment of glioblastoma; strength and flaws. J Neurooncol 2025; 171:495-530. [PMID: 39538038 DOI: 10.1007/s11060-024-04876-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: 09/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive primary brain tumor that carries a grim prognosis. Because of the dearth of treatment options available for treatment of GBM, Chimeric Antigen Receptor (CAR)-engineered T cell and Natural Killer (NK) therapy could provide alternative strategies to address the challenges in GBM treatment. In these approaches, CAR T and NK cells are engineered for cancer-specific immunotherapy by recognizing surface antigens independently of major histocompatibility complex (MHC) molecules. However, the efficacy of CAR T cells is hindered by GBM's downregulation of its targeted antigens. CAR NK cells face similar challenges, but, in contrast, they offer advantages as off-the-shelf allogeneic products, devoid of graft-versus-host disease (GVHD) risk as well as anti-cancer activity beyond CAR specificity, potentially reducing the risk of relapse or resistance. Despite CAR T cell therapies being extensively studied in clinical settings, the use of CAR-modified NK cells in GBM treatment remains largely in the preclinical stage. This review aims to discuss recent advancements in NK cell and CAR T cell therapies for GBM, including methods for introducing CARs into both NK cells and T cells, addressing manufacturing challenges, and providing evidence supporting the efficacy of these approaches from preclinical and early-phase clinical studies. The comprehensive evaluation of CAR-engineered NK cells and CAR T cells seeks to identify the optimal therapeutic approach for GBM, contributing to the development of effective immunotherapies for this devastating disease.
Collapse
Affiliation(s)
- Mohammadmahdi Sabahi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Ali Fathi Jouzdani
- Neurosurgery Research Group (NRG), Hamadan University of Medical Sciences, Hamadan, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zohre Sadeghian
- Department of Pathology & Laboratory Medicine, Cleveland Clinic Florida, Weston, FL, USA
| | | | - Hadi Sultan
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Arash Salehipour
- Neurosurgery Research Group (NRG), Hamadan University of Medical Sciences, Hamadan, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Lana Maniakhina
- Department of Neurosurgery, Geisinger and Geisinger Commonwealth School of Medicine, Wilkes-Barre, PA, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Badih Adada
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
| | - Hamid Borghei-Razavi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| |
Collapse
|
6
|
Kong Y, Li J, Zhao X, Wu Y, Chen L. CAR-T cell therapy: developments, challenges and expanded applications from cancer to autoimmunity. Front Immunol 2025; 15:1519671. [PMID: 39850899 PMCID: PMC11754230 DOI: 10.3389/fimmu.2024.1519671] [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: 10/30/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has rapidly emerged as a groundbreaking approach in cancer treatment, particularly for hematologic malignancies. However, the application of CAR-T cell therapy in solid tumors remains challenging. This review summarized the development of CAR-T technologies, emphasized the challenges and solutions in CAR-T cell therapy for solid tumors. Also, key innovations were discussed including specialized CAR-T, combination therapies and the novel use of CAR-Treg, CAR-NK and CAR-M cells. Besides, CAR-based cell therapy have extended its reach beyond oncology to autoimmune disorders. We reviewed preclinical experiments and clinical trials involving CAR-T, Car-Treg and CAAR-T cell therapies in various autoimmune diseases. By highlighting these cutting-edge developments, this review underscores the transformative potential of CAR technologies in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Yanwei Wu
- School of Medicine, Shanghai University, Shanghai, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
7
|
Yao P, Liu YG, Huang G, Hao L, Wang R. The development and application of chimeric antigen receptor natural killer (CAR-NK) cells for cancer therapy: current state, challenges and emerging therapeutic advances. Exp Hematol Oncol 2024; 13:118. [PMID: 39633491 PMCID: PMC11616395 DOI: 10.1186/s40164-024-00583-7] [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: 05/16/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
Immunotherapy has transformed the landscape of cancer treatment, with chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy emerging as a front runner in addressing some hematological malignancies. Despite its considerable efficacy, the occurrence of severe adverse effects associated with CAR-T cell therapy has limited their scope and prompted the exploration of alternative therapeutic strategies. Natural killer (NK) cells, characterized by both their innate cytotoxicity and ability to lyse target cells without the constraint of peptide specificity conferred by a major histocompatibility complex (MHC), have similarly garnered attention as a viable immunotherapy. As such, another therapeutic approach has recently emerged that seeks to combine the continued success of CAR-T cell therapy with the flexibility of NK cells. Clinical trials involving CAR-engineered NK (CAR-NK) cell therapy have exhibited promising efficacy with fewer deleterious side effects. This review aims to provide a concise overview of the cellular and molecular basis of NK cell biology, facilitating a better understanding of advancements in CAR design and manufacturing. The focus is on current approaches and strategies employed in CAR-NK cell development, exploring at both preclinical and clinical settings. We will reflect upon the achievements, advantages, and challenges intrinsic to CAR-NK cell therapy. Anticipating the maturation of CAR-NK cell therapy technology, we foresee its encouraging prospects for a broader range of cancer patients and other conditions. It is our belief that this CAR-NK progress will bring us closer to making significant strides in the treatment of refractory and recurrent cancers, as well as other immune-mediated disorders.
Collapse
Affiliation(s)
- Pin Yao
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Ya-Guang Liu
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Gang Huang
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Liangchun Hao
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Runan Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
8
|
Arjmand B, Alavi-Moghadam S, Khorsand G, Sarvari M, Arjmand R, Rezaei-Tavirani M, Rajaeinejad M, Mosaed R. Cell-Based Vaccines: Frontiers in Medical Technology for Cancer Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:480-499. [DOI: 10.1007/s40883-024-00338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 01/13/2024] [Accepted: 02/17/2024] [Indexed: 01/03/2025]
|
9
|
Devaraja K, Singh M, Sharan K, Aggarwal S. Coley's Toxin to First Approved Therapeutic Vaccine-A Brief Historical Account in the Progression of Immunobiology-Based Cancer Treatment. Biomedicines 2024; 12:2746. [PMID: 39767654 PMCID: PMC11726767 DOI: 10.3390/biomedicines12122746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Cancer immunobiology is one of the hot topics of discussion amongst researchers today, and immunotherapeutic modalities are among the selected few emerging approaches to cancer treatment that have exhibited a promising outlook. However, immunotherapy is not a new kid on the block; it has been around for centuries. The origin of cancer immunotherapy in modern medicine can be traced back to the initial reports of spontaneous regression of malignant tumors in some patients following an acute febrile infection, at the turn of the twentieth century. This review briefly revisits the historical accounts of immunotherapy, highlighting some of the significant developments in the field of cancer immunobiology, that have been instrumental in bringing back the immunotherapeutic approaches to the forefront of cancer research. Some of the topics covered are: Coley's toxin-the first immunotherapeutic; the genesis of the theory of immune surveillance; the discovery of T lymphocytes and dendritic cells and their roles; the role of tumor antigens; relevance of tumor microenvironment; the anti-tumor (therapeutic) ability of Bacillus Calmette- Guérin; Melacine-the first therapeutic vaccine engineered; theories of immunoediting and immunophenotyping of cancer; and Provenge-the first FDA-approved therapeutic vaccine. In this review, head and neck cancer has been taken as the reference tumor for narrating the progression of cancer immunobiology, particularly for highlighting the advent of immunotherapeutic agents.
Collapse
Affiliation(s)
- K. Devaraja
- Department of Head and Neck Surgery, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Manisha Singh
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Krishna Sharan
- Department of Radiation Oncology, K S Hegde Medical College, Nitte University, Mangalore 574110, India;
| | - Sadhna Aggarwal
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
10
|
Zhu X, Xue J, Jiang H, Xue D. CAR-NK cells for gastrointestinal cancer immunotherapy: from bench to bedside. Mol Cancer 2024; 23:237. [PMID: 39443938 PMCID: PMC11515662 DOI: 10.1186/s12943-024-02151-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Gastrointestinal (GI) cancers represent a significant health burden worldwide. Their incidence continues to increase, and their management remains a clinical challenge. Chimeric antigen receptor (CAR) natural killer (NK) cells have emerged as a promising alternative to CAR-T cells for immunotherapy of GI cancers. Notably, CAR-NK cells offer several advantages, including reduced risk of graft-versus-host disease, lower cytokine release syndrome, and the ability to target cancer cells through both CAR-dependent and natural cytotoxic mechanisms. MAIN BODY This review comprehensively discusses the development and applications of CAR-NK cells in the treatment of GI cancers. We explored various sources of NK cells, CAR design strategies, and the current state of CAR-NK cell therapy for GI cancers, highlighting recent preclinical and clinical trials. Additionally, we addressed existing challenges and propose potential strategies to enhance the efficacy and safety of CAR-NK cell therapy. CONCLUSIONS Our findings highlight the potential of CAR-NK cells to revolutionize GI cancer treatment and pave the way for future clinical applications.
Collapse
Affiliation(s)
- Xingwang Zhu
- Department of Urinary Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China
| | - Jieyun Xue
- China Medical University, Shenyang, Liaoning Province, 110000, P.R. China
| | - Hongzhou Jiang
- Department of Neurosurgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China
| | - Dongwei Xue
- Department of Urinary Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China.
| |
Collapse
|
11
|
Jiang P, Jing S, Sheng G, Jia F. The basic biology of NK cells and its application in tumor immunotherapy. Front Immunol 2024; 15:1420205. [PMID: 39221244 PMCID: PMC11361984 DOI: 10.3389/fimmu.2024.1420205] [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: 04/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Natural Killer (NK) cells play a crucial role as effector cells within the tumor immune microenvironment, capable of identifying and eliminating tumor cells through the expression of diverse activating and inhibitory receptors that recognize tumor-related ligands. Therefore, harnessing NK cells for therapeutic purposes represents a significant adjunct to T cell-based tumor immunotherapy strategies. Presently, NK cell-based tumor immunotherapy strategies encompass various approaches, including adoptive NK cell therapy, cytokine therapy, antibody-based NK cell therapy (enhancing ADCC mediated by NK cells, NK cell engagers, immune checkpoint blockade therapy) and the utilization of nanoparticles and small molecules to modulate NK cell anti-tumor functionality. This article presents a comprehensive overview of the latest advances in NK cell-based anti-tumor immunotherapy, with the aim of offering insights and methodologies for the clinical treatment of cancer patients.
Collapse
Affiliation(s)
- Pan Jiang
- Department of General Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Infectious Diseases, Jingzhou First People’s Hospital, Jingzhou, China
| | - Shaoze Jing
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fajing Jia
- Department of General Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
12
|
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
|
13
|
Bahramloo M, Shahabi SA, Kalarestaghi H, Rafat A, Mazloumi Z, Samimifar A, Asl KD. CAR-NK cell therapy in AML: Current treatment, challenges, and advantage. Biomed Pharmacother 2024; 177:117024. [PMID: 38941897 DOI: 10.1016/j.biopha.2024.117024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024] Open
Abstract
Over the last decade, discovery of novel therapeutic method has been attention by the researchers and has changed the therapeutic perspective of hematological malignancies. Although NK cell play a pivotal role in the elimination of abnormal and cancerous cells, there are evidence that NK cell are disarm in hematological malignancy. Chimeric antigen receptor NK (CAR-NK) cell therapy, which includes the engineering of NK cells to detect tumor-specific antigens and, as a result, clear of cancerous cells, has created various clinical advantage for several human malignancies treatment. In the current review, we summarized NK cell dysfunction and CAR-NK cell based immunotherapy to treat AML patient.
Collapse
Affiliation(s)
- Mohammadmahdi Bahramloo
- Department of Medical Sciences, Student Research Committee, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Sina Alinejad Shahabi
- Department of Medical Sciences, Student Research Committee, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Hossein Kalarestaghi
- Research Laboratory for Embryology and Stem Cell, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Rafat
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zeinab Mazloumi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arian Samimifar
- Department of Medical Sciences, Student Research Committee, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Khadijeh Dizaji Asl
- Department of Histopathology and Anatomy, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran.
| |
Collapse
|
14
|
Liu B, Zhou H, Tan L, Siu KTH, Guan XY. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct Target Ther 2024; 9:175. [PMID: 39013849 PMCID: PMC11252281 DOI: 10.1038/s41392-024-01856-7] [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: 01/23/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 07/18/2024] Open
Abstract
Traditional therapeutic approaches such as chemotherapy and radiation therapy have burdened cancer patients with onerous physical and psychological challenges. Encouragingly, the landscape of tumor treatment has undergone a comprehensive and remarkable transformation. Emerging as fervently pursued modalities are small molecule targeted agents, antibody-drug conjugates (ADCs), cell-based therapies, and gene therapy. These cutting-edge treatment modalities not only afford personalized and precise tumor targeting, but also provide patients with enhanced therapeutic comfort and the potential to impede disease progression. Nonetheless, it is acknowledged that these therapeutic strategies still harbour untapped potential for further advancement. Gaining a comprehensive understanding of the merits and limitations of these treatment modalities holds the promise of offering novel perspectives for clinical practice and foundational research endeavours. In this review, we discussed the different treatment modalities, including small molecule targeted drugs, peptide drugs, antibody drugs, cell therapy, and gene therapy. It will provide a detailed explanation of each method, addressing their status of development, clinical challenges, and potential solutions. The aim is to assist clinicians and researchers in gaining a deeper understanding of these diverse treatment options, enabling them to carry out effective treatment and advance their research more efficiently.
Collapse
Affiliation(s)
- Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Hongyu Zhou
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Licheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Kin To Hugo Siu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China.
| |
Collapse
|
15
|
Yang R, Yang Y, Liu R, Wang Y, Yang R, He A. Advances in CAR-NK cell therapy for hematological malignancies. Front Immunol 2024; 15:1414264. [PMID: 39007146 PMCID: PMC11239349 DOI: 10.3389/fimmu.2024.1414264] [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: 04/08/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has revolutionized the treatment of hematological malignancies, demonstrably improving patient outcomes and prognosis. However, its application has introduced new challenges, such as safety concerns, off-target toxicities, and significant costs. Natural killer (NK) cells are crucial components of the innate immune system, capable of eliminating tumor cells without prior exposure to specific antigens or pre-activation. This inherent advantage complements the limitations of T cells, making CAR-NK cell therapy a promising avenue for hematological tumor immunotherapy. In recent years, preclinical and clinical studies have yielded preliminary evidence supporting the safety and efficacy of CAR-NK cell therapy in hematological malignancies, paving the way for future advancements in immunotherapy. This review aims to succinctly discuss the characteristics, significant therapeutic progress, and potential challenges associated with CAR-NK cell therapy.
Collapse
Affiliation(s)
- Rui Yang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yun Yang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Xi’an Key Laboratory of Hematological Diseases, Xi’an, Shaanxi, China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yiwen Wang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Ruoyu Yang
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Xi’an Key Laboratory of Hematological Diseases, Xi’an, Shaanxi, China
| |
Collapse
|
16
|
Taheri MM, Javan F, Poudineh M, Athari SS. CAR-NKT Cells in Asthma: Use of NKT as a Promising Cell for CAR Therapy. Clin Rev Allergy Immunol 2024; 66:328-362. [PMID: 38995478 DOI: 10.1007/s12016-024-08998-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
NKT cells, unique lymphocytes bridging innate and adaptive immunity, offer significant potential for managing inflammatory disorders like asthma. Activating iNKT induces increasing IFN-γ, TGF-β, IL-2, and IL-10 potentially suppressing allergic asthma. However, their immunomodulatory effects, including granzyme-perforin-mediated cytotoxicity, and expression of TIM-3 and TRAIL warrant careful consideration and targeted approaches. Although CAR-T cell therapy has achieved remarkable success in treating certain cancers, its limitations necessitate exploring alternative approaches. In this context, CAR-NKT cells emerge as a promising approach for overcoming these challenges, potentially achieving safer and more effective immunotherapies. Strategies involve targeting distinct IgE-receptors and their interactions with CAR-NKT cells, potentially disrupting allergen-mast cell/basophil interactions and preventing inflammatory cytokine release. Additionally, targeting immune checkpoints like PDL-2, inducible ICOS, FASL, CTLA-4, and CD137 or dectin-1 for fungal asthma could further modulate immune responses. Furthermore, artificial intelligence and machine learning hold immense promise for revolutionizing NKT cell-based asthma therapy. AI can optimize CAR-NKT cell functionalities, design personalized treatment strategies, and unlock a future of precise and effective care. This review discusses various approaches to enhancing CAR-NKT cell efficacy and longevity, along with the challenges and opportunities they present in the treatment of allergic asthma.
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
| | - Seyyed Shamsadin Athari
- Cancer Gene therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
17
|
Ye Z, Cheng P, Huang Q, Hu J, Huang L, Hu G. Immunocytes interact directly with cancer cells in the tumor microenvironment: one coin with two sides and future perspectives. Front Immunol 2024; 15:1388176. [PMID: 38840908 PMCID: PMC11150710 DOI: 10.3389/fimmu.2024.1388176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
The tumor microenvironment is closely linked to the initiation, promotion, and progression of solid tumors. Among its constitutions, immunologic cells emerge as critical players, facilitating immune evasion and tumor progression. Apart from their indirect impact on anti-tumor immunity, immunocytes directly influence neoplastic cells, either bolstering or impeding tumor advancement. However, current therapeutic modalities aimed at alleviating immunosuppression from regulatory cells on effector immune cell populations may not consistently yield satisfactory results in various solid tumors, such as breast carcinoma, colorectal cancer, etc. Therefore, this review outlines and summarizes the direct, dualistic effects of immunocytes such as T cells, innate lymphoid cells, B cells, eosinophils, and tumor-associated macrophages on tumor cells within the tumor microenvironment. The review also delves into the underlying mechanisms involved and presents the outcomes of clinical trials based on these direct effects, aiming to propose innovative and efficacious therapeutic strategies for addressing solid tumors.
Collapse
Affiliation(s)
- Zhiyi Ye
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital; Shaoxing Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Pu Cheng
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, Anhui Medical University, Hefei, Anhui, China
| | - Jingjing Hu
- School of Medicine, Shaoxing University, Zhejiang, China
| | - Liming Huang
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital; Shaoxing Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Guoming Hu
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, Zhejiang, China
| |
Collapse
|
18
|
Guijarro-Albaladejo B, Marrero-Cepeda C, Rodríguez-Arbolí E, Sierro-Martínez B, Pérez-Simón JA, García-Guerrero E. Chimeric antigen receptor (CAR) modified T Cells in acute myeloid leukemia: limitations and expectations. Front Cell Dev Biol 2024; 12:1376554. [PMID: 38694825 PMCID: PMC11061469 DOI: 10.3389/fcell.2024.1376554] [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: 01/25/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with a poor prognosis despite the advent of novel therapies. Consequently, a major need exists for new therapeutic options, particularly for patients with relapsed/refractory (R/R) AML. In recent years, it has been possible to individualize the treatment of a subgroup of patients, particularly with the emergence of multiple targeted therapies. Nonetheless, a considerable number of patients remain without therapeutic options, and overall prognosis remains poor because of a high rate of disease relapse. In this sense, cellular therapies, especially chimeric antigen receptor (CAR)-T cell therapy, have dramatically shifted the therapeutic options for other hematologic malignancies, such as diffuse large B cell lymphoma and acute lymphoblastic leukemia. In contrast, effectively treating AML with CAR-based immunotherapy poses major biological and clinical challenges, most of them derived from the unmet need to identify target antigens with expression restricted to the AML blast without compromising the viability of the normal hematopoietic stem cell counterpart. Although those limitations have hampered CAR-T cell therapy translation to the clinic, there are several clinical trials where target antigens, such as CD123, CLL-1 or CD33 are being used to treat AML patients showing promising results. Moreover, there are continuing efforts to enhance the specificity and efficacy of CAR-T cell therapy in AML. These endeavors encompass the exploration of novel avenues, including the development of dual CAR-T cells and next-generation CAR-T cells, as well as the utilization of gene editing tools to mitigate off-tumor toxicities. In this review, we will summarize the ongoing clinical studies and the early clinical results reported with CAR-T cells in AML, as well as highlight CAR-T cell limitations and the most recent approaches to overcome these barriers. We will also discuss how and when CAR-T cells should be used in the context of AML.
Collapse
Affiliation(s)
- Beatriz Guijarro-Albaladejo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Cristina Marrero-Cepeda
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Eduardo Rodríguez-Arbolí
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Belén Sierro-Martínez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - José Antonio Pérez-Simón
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Estefanía García-Guerrero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| |
Collapse
|
19
|
Colucci M, Zumerle S, Bressan S, Gianfanti F, Troiani M, Valdata A, D'Ambrosio M, Pasquini E, Varesi A, Cogo F, Mosole S, Dongilli C, Desbats MA, Contu L, Revankdar A, Chen J, Kalathur M, Perciato ML, Basilotta R, Endre L, Schauer S, Othman A, Guccini I, Saponaro M, Maraccani L, Bancaro N, Lai P, Liu L, Pernigoni N, Mele F, Merler S, Trotman LC, Guarda G, Calì B, Montopoli M, Alimonti A. Retinoic acid receptor activation reprograms senescence response and enhances anti-tumor activity of natural killer cells. Cancer Cell 2024; 42:646-661.e9. [PMID: 38428412 PMCID: PMC11003464 DOI: 10.1016/j.ccell.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/19/2023] [Accepted: 02/07/2024] [Indexed: 03/03/2024]
Abstract
Cellular senescence can exert dual effects in tumors, either suppressing or promoting tumor progression. The senescence-associated secretory phenotype (SASP), released by senescent cells, plays a crucial role in this dichotomy. Consequently, the clinical challenge lies in developing therapies that safely enhance senescence in cancer, favoring tumor-suppressive SASP factors over tumor-promoting ones. Here, we identify the retinoic-acid-receptor (RAR) agonist adapalene as an effective pro-senescence compound in prostate cancer (PCa). Reactivation of RARs triggers a robust senescence response and a tumor-suppressive SASP. In preclinical mouse models of PCa, the combination of adapalene and docetaxel promotes a tumor-suppressive SASP that enhances natural killer (NK) cell-mediated tumor clearance more effectively than either agent alone. This approach increases the efficacy of the allogenic infusion of human NK cells in mice injected with human PCa cells, suggesting an alternative therapeutic strategy to stimulate the anti-tumor immune response in "immunologically cold" tumors.
Collapse
Affiliation(s)
- Manuel Colucci
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland; Faculty of Biology and Medicine, University of Lausanne UNIL, CH1011 Lausanne, Switzerland
| | - Sara Zumerle
- Veneto Institute of Molecular Medicine (VIMM) & Department of Medicine, University of Padova, Padova, Italy
| | - Silvia Bressan
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland; Veneto Institute of Molecular Medicine (VIMM) & Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Federico Gianfanti
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland; Veneto Institute of Molecular Medicine (VIMM) & Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Martina Troiani
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland; Bioinformatics Core Unit, Swiss Institute of Bioinformatics, TI, Bellinzona, Switzerland
| | - Aurora Valdata
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Department of Health Sciences and Technology (D-HEST) ETH Zurich, Zurich, CH, Switzerland
| | - Mariantonietta D'Ambrosio
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; MRC London Institute of Medical Sciences (LMS), London, UK
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Angelica Varesi
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Francesca Cogo
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Cristina Dongilli
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Maria Andrea Desbats
- Veneto Institute of Molecular Medicine (VIMM) & Department of Medicine, University of Padova, Padova, Italy
| | - Liliana Contu
- Veneto Institute of Molecular Medicine (VIMM) & Department of Medicine, University of Padova, Padova, Italy
| | - Ajinkya Revankdar
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jingjing Chen
- Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Madhuri Kalathur
- Children's GMP, LLC, St. Jude Children's Research Hospital, 262 Danny Thomas Place Mail Stop 920 Memphis, TN 38105, USA
| | - Maria Luna Perciato
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK
| | - Rossella Basilotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 viale Ferdinando D'Alcontres, Italy
| | - Laczko Endre
- Functional Genomics Center Zurich, ETHZ and University of Zurich, Zurich, CH, Switzerland
| | - Stefan Schauer
- Functional Genomics Center Zurich, ETHZ and University of Zurich, Zurich, CH, Switzerland
| | - Alaa Othman
- Functional Genomics Center Zurich, ETHZ and University of Zurich, Zurich, CH, Switzerland
| | - Ilaria Guccini
- Department of Health Sciences and Technology (D-HEST) ETH Zurich, Zurich, CH, Switzerland
| | - Miriam Saponaro
- Veneto Institute of Molecular Medicine (VIMM) & Department of Medicine, University of Padova, Padova, Italy
| | - Luisa Maraccani
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Veneto Institute of Molecular Medicine (VIMM) & Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Nicolò Bancaro
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Ping Lai
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Lei Liu
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Nicolò Pernigoni
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Federico Mele
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Sara Merler
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine, University of Verona and Verona University and Hospital Trust, Verona, Italy
| | - Lloyd C Trotman
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Greta Guarda
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Bianca Calì
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland
| | - Monica Montopoli
- Veneto Institute of Molecular Medicine (VIMM) & Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), CH6500 Bellinzona, Switzerland; Università della Svizzera Italiana, CH6900 Lugano, Switzerland; Veneto Institute of Molecular Medicine (VIMM) & Department of Medicine, University of Padova, Padova, Italy; Department of Health Sciences and Technology (D-HEST) ETH Zurich, Zurich, CH, Switzerland; Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
| |
Collapse
|
20
|
Khoshtinat Nikkhoi S, Yang G, Owji H, Grizotte-Lake M, Cohen RI, Gil Gonzalez L, Massumi M, Hatefi A. Bispecific immune cell engager enhances the anticancer activity of CD16+ NK cells and macrophages in vitro, and eliminates cancer metastasis in NK humanized NOG mice. J Immunother Cancer 2024; 12:e008295. [PMID: 38490714 PMCID: PMC10946374 DOI: 10.1136/jitc-2023-008295] [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: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND In a prior report, we detailed the isolation and engineering of a bispecific killer cell engager, referred to as BiKE:E5C1. The BiKE:E5C1 exhibits high affinity/specificity for the CD16a activating receptor on natural killer (NK) cells and human epidermal growth factor receptor 2 (HER2) on cancer cells. In vitro studies have demonstrated that BiKE:E5C1 can activate the NK cells and induce the killing of HER2+ ovarian and breast cancer cells, surpassing the performance of the best-in-class monoclonal antibody, Trazimera (trastuzumab). To advance this BiKE technology toward clinical application, the objective of this research was to demonstrate the ability of BiKE:E5C1 to activate CD16+ immune cells such as NK cells and macrophages to kill cancer cells, and eradicate metastatic HER2+ tumors in NK humanized NOG mice. METHODS We assessed BiKE:E5C1's potential to activate CD16-expressing peripheral blood (PB)-NK cells, laNK92 cells, and THP-1-CD16A monocyte-macrophages through flowcytometry and antibody-dependent cell-mediated cytotoxicity/phagocytosis (ADCC) assays. Subsequently, laNK92 cells were selected as effector cells and genetically modified to express the nanoluciferase gene, enabling the monitoring of their viability in NK humanized NOG mice using quantitative bioluminescent imaging (qBLI). To evaluate the functionality of BiKE:E5C1 in vivo, we introduced firefly luciferase-expressing ovarian cancer cells via intraperitoneal injection into hIL-15 and hIL-2 NOG mice, creating a model of ovarian cancer metastasis. Once tumor establishment was confirmed, we treated the mice with laNK92 cells plus BiKE:E5C1 and the response to therapy was assessed using qBLI. RESULTS Our data demonstrate that BiKE:E5C1 activates not only laNK92 cells but also PB-NK cells and macrophages, significantly enhancing their anticancer activities. ADCC assay demonstrated that IgG1 Fc region had no impact on BiKE:E5C1's anticancer activity. In vivo results reveal that both hIL-15 and hIL-2 NOG mouse models support the viability and proliferation of laNK92 cells. Furthermore, it was observed that BiKE:E5C1 activates laNK92 cells in mice, leading to eradication of cancer metastasis in both NK humanized hIL-15 and hIL-2 NOG mouse models. CONCLUSIONS Collectively, our in vivo findings underscore BiKE:E5C1's potential as an immune cell engager capable of activating immune cells for cancer cell elimination, thereby expanding the arsenal of available BiKEs for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Ge Yang
- Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Hajar Owji
- Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | | | - Rick I Cohen
- Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Lazaro Gil Gonzalez
- St Michael's Hospital Keenan Research Centre for Biomedical Science, Toronto, Ontario, Canada
| | - Mohammad Massumi
- Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Arash Hatefi
- Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
21
|
Esmaeilzadeh A, Hadiloo K, Jabbari M, Elahi R. Current progress of chimeric antigen receptor (CAR) T versus CAR NK cell for immunotherapy of solid tumors. Life Sci 2024; 337:122381. [PMID: 38145710 DOI: 10.1016/j.lfs.2023.122381] [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: 10/10/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
Equipping cancer-fighting immune cells with chimeric antigen receptor (CAR) has gained immense attention for cancer treatment. CAR-engineered T cells (CAR T cells) are the first immune-engineered cells that have achieved brilliant results in anti-cancer therapy. Despite promising anti-cancer features, CAR T cells could also cause fatal side effects and have shown inadequate efficacy in some studies. This has led to the introduction of other candidates for CAR transduction, e.g., Natural killer cells (NK cells). Regarding the better safety profile and anti-cancer properties, CAR-armored NK cells (CAR NK cells) could be a beneficial and suitable alternative to CAR T cells. Since introducing these two cells as anti-cancer structures, several studies have investigated their efficacy and safety, and most of them have focused on hematological malignancies. Solid tumors have unique properties that make them more resistant and less curable cancers than hematological malignancies. In this review article, we conduct a comprehensive review of the structure and properties of CAR NK and CAR T cells, compare the recent experience of immunotherapy with CAR T and CAR NK cells in various solid cancers, and overview current challenges and future solutions to battle solid cancers using CARNK cells.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Kaveh Hadiloo
- Student Research Committee, Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Marjan Jabbari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
22
|
Letafati A, Ardekani OS, Naderisemiromi M, Norouzi M, Shafiei M, Nik S, Mozhgani SH. Unraveling the dynamic mechanisms of natural killer cells in viral infections: insights and implications. Virol J 2024; 21:18. [PMID: 38216935 PMCID: PMC10785350 DOI: 10.1186/s12985-024-02287-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/04/2024] [Indexed: 01/14/2024] Open
Abstract
Viruses pose a constant threat to human well-being, necessitating the immune system to develop robust defenses. Natural killer (NK) cells, which play a crucial role in the immune system, have become recognized as vital participants in protecting the body against viral infections. These remarkable innate immune cells possess the unique ability to directly recognize and eliminate infected cells, thereby contributing to the early control and containment of viral pathogens. However, recent research has uncovered an intriguing phenomenon: the alteration of NK cells during viral infections. In addition to their well-established role in antiviral defense, NK cells undergo dynamic changes in their phenotype, function, and regulatory mechanisms upon encountering viral pathogens. These alterations can significantly impact the effectiveness of NK cell responses during viral infections. This review explores the multifaceted role of NK cells in antiviral immunity, highlighting their conventional effector functions as well as the emerging concept of NK cell alteration in the context of viral infections. Understanding the intricate interplay between NK cells and viral infections is crucial for advancing our knowledge of antiviral immune responses and could offer valuable information for the creation of innovative therapeutic approaches to combat viral diseases.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Omid Salahi Ardekani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Mina Naderisemiromi
- Department of Immunology, Faculty of Medicine and Health, The University of Manchester, Manchester, UK
| | - Mehdi Norouzi
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | | | - Soheil Nik
- School of Medicine, Alborz University of Medical Sciences, Karaj, Alborz, Iran
| | - Sayed-Hamidreza Mozhgani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran.
- Department of Microbiology and Virology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
23
|
Hajibabaie F, Abedpoor N, Haghjooy Javanmard S, Hasan A, Sharifi M, Rahimmanesh I, Shariati L, Makvandi P. The molecular perspective on the melanoma and genome engineering of T-cells in targeting therapy. ENVIRONMENTAL RESEARCH 2023; 237:116980. [PMID: 37648188 DOI: 10.1016/j.envres.2023.116980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
Melanoma, an aggressive malignant tumor originating from melanocytes in humans, is on the rise globally, with limited non-surgical treatment options available. Recent advances in understanding the molecular and cellular mechanisms underlying immune escape, tumorigenesis, drug resistance, and cancer metastasis have paved the way for innovative therapeutic strategies. Combination therapy targeting multiple pathways simultaneously has been shown to be promising in treating melanoma, eliciting favorable responses in most melanoma patients. CAR T-cells, engineered to overcome the limitations of human leukocyte antigen (HLA)-dependent tumor cell detection associated with T-cell receptors, offer an alternative approach. By genetically modifying apheresis-collected allogeneic or autologous T-cells to express chimeric antigen receptors, CAR T-cells can appreciate antigens on cell surfaces independently of major histocompatibility complex (MHC), providing a significant cancer cell detection advantage. However, identifying the most effective target antigen is the initial step, as it helps mitigate the risk of toxicity due to "on-target, off-tumor" and establishes a targeted therapeutic strategy. Furthermore, evaluating signaling pathways and critical molecules involved in melanoma pathogenesis remains insufficient. This study emphasizes the novel approaches of CAR T-cell immunoediting and presents new insights into the molecular signaling pathways associated with melanoma.
Collapse
Affiliation(s)
- Fatemeh Hajibabaie
- Department of Biology, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran; Department of Medical Biotechnology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Navid Abedpoor
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran; Department of Medical Biotechnology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, 2713, Qatar; Biomedical Research Center, Qatar University, Doha, 2713, Qatar.
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran; Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| |
Collapse
|
24
|
Huang S, Xing F, Dai Y, Zhang Z, Zhou G, Yang S, Liu YC, Yuan Z, Luo KQ, Ying T, Chu D, Liu TM, Deng CX, Zhao Q. Navigating chimeric antigen receptor-engineered natural killer cells as drug carriers via three-dimensional mapping of the tumor microenvironment. J Control Release 2023; 362:524-535. [PMID: 37673307 DOI: 10.1016/j.jconrel.2023.09.007] [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: 04/13/2023] [Revised: 08/31/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
Chimeric antigen receptor (CAR)-modified natural killer (NK) cells are recognized as promising immunotherapeutic agents for cancer treatment. However, the efficacy and trafficking of CAR-NK cells in solid tumors are hindered by the complex barriers present in the tumor microenvironment (TME). We have developed a novel strategy that utilizes living CAR-NK cells as carriers to deliver anticancer drugs specifically to the tumor site. We also introduce a time-lapse method for evaluating the efficacy and tumor specificity of CAR-NK cells using a two-photon microscope in live mouse models and three-dimensional (3D) tissue slide cultures. Our results demonstrate that CAR-NK cells exhibit enhanced antitumor immunity when combined with photosensitive chemicals in both in vitro and in vivo tumor models. Additionally, we have successfully visualized the trafficking, infiltration, and accumulation of drug-loaded CAR-NK cells in deeply situated TME using non-invasive intravital two-photon microscopy. Our findings highlight that tumor infiltration of CAR-NK cells can be intravitally monitored through the two-photon microscope approach. In conclusion, our study demonstrates the successful integration of CAR-NK cells as drug carriers and paves the way for combined cellular and small-molecule therapies in cancer treatment. Furthermore, our 3D platform offers a valuable tool for assessing the behavior of CAR cells within solid tumors, facilitating the development and optimization of immunotherapeutic strategies with clinical imaging approaches.
Collapse
Affiliation(s)
- Shigao Huang
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Radiation Oncology of Xijing Hospital, Fourth Military Medical University, Xi' an, China
| | - Fuqiang Xing
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China; Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Yeneng Dai
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Zhiming Zhang
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Guangyu Zhou
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Shuo Yang
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Yu-Cheng Liu
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Zhen Yuan
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Kathy Qian Luo
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dafeng Chu
- Geneleap Biotechnology LLC, Woburn, MA, USA.
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| | - Chu-Xia Deng
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| | - Qi Zhao
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
25
|
Zhang M, Lam KP, Xu S. Natural Killer Cell Engagers (NKCEs): a new frontier in cancer immunotherapy. Front Immunol 2023; 14:1207276. [PMID: 37638058 PMCID: PMC10450036 DOI: 10.3389/fimmu.2023.1207276] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Natural Killer (NK) cells are a type of innate lymphoid cells that play a crucial role in immunity by killing virally infected or tumor cells and secreting cytokines and chemokines. NK cell-mediated immunotherapy has emerged as a promising approach for cancer treatment due to its safety and effectiveness. NK cell engagers (NKCEs), such as BiKE (bispecific killer cell engager) or TriKE (trispecific killer cell engager), are a novel class of antibody-based therapeutics that exhibit several advantages over other cancer immunotherapies harnessing NK cells. By bridging NK and tumor cells, NKCEs activate NK cells and lead to tumor cell lysis. A growing number of NKCEs are currently undergoing development, with some already in clinical trials. However, there is a need for more comprehensive studies to determine how the molecular design of NKCEs affects their functionality and manufacturability, which are crucial for their development as off-the-shelf drugs for cancer treatment. In this review, we summarize current knowledge on NKCE development and discuss critical factors required for the production of effective NKCEs.
Collapse
Affiliation(s)
- Minchuan Zhang
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Singapore
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
26
|
Tang Y, Qian C. Research progress in leveraging biomaterials for enhancing NK cell immunotherapy. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:267-278. [PMID: 37476938 PMCID: PMC10409897 DOI: 10.3724/zdxbyxb-2022-0728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/09/2023] [Indexed: 07/22/2023]
Abstract
NK cell immunotherapy is a promising antitumor therapeutic modality after the development of T cell immunotherapy. Structural modification of NK cells with biomaterials may provide a precise, efficient, and low-cost strategy to enhance NK cell immunotherapy. The biomaterial modification of NK cells can be divided into two strategies: surface engineering with biomaterials and intracellular modification. The surface engineering strategies include hydrophobic interaction of lipids, receptor-ligand interaction between membrane proteins, covalent binding to amino acid residues, click reaction and electrostatic interaction. The intracellular modification strategies are based on manipulation by nanotechnology using membranous materials from various sources of NK cells (such as exosome, vesicle and cytomembranes). Finally, the biomaterials-based strategies regulate the recruitment, recognition and cytotoxicity of NK cells in the solid tumor site in situ to boost the activity of NK cells in the tumor. This article reviews the recent research progress in enhancing NK cell therapy based on biomaterial modification, to provide a reference for further researches on engineering NK cell therapy with biomaterials.
Collapse
Affiliation(s)
- Yingqi Tang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, State Key Laboratory of Natural Medicines, Nanjing 210009, China.
| | - Chenggen Qian
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, State Key Laboratory of Natural Medicines, Nanjing 210009, China.
| |
Collapse
|
27
|
Lizana-Vasquez GD, Torres-Lugo M, Dixon R, Powderly JD, Warin RF. The application of autologous cancer immunotherapies in the age of memory-NK cells. Front Immunol 2023; 14:1167666. [PMID: 37205105 PMCID: PMC10185894 DOI: 10.3389/fimmu.2023.1167666] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/17/2023] [Indexed: 05/21/2023] Open
Abstract
Cellular immunotherapy has revolutionized the oncology field, yielding improved results against hematological and solid malignancies. NK cells have become an attractive alternative due to their capacity to activate upon recognition of "stress" or "danger" signals independently of Major Histocompatibility Complex (MHC) engagement, thus making tumor cells a perfect target for NK cell-mediated cancer immunotherapy even as an allogeneic solution. While this allogeneic use is currently favored, the existence of a characterized memory function for NK cells ("memory-like" NK cells) advocates for an autologous approach, that would benefit from the allogeneic setting discoveries, but with added persistence and specificity. Still, both approaches struggle to exert a sustained and high anticancer effect in-vivo due to the immunosuppressive tumor micro-environment and the logistical challenges of cGMP production or clinical deployment. Novel approaches focused on the quality enhancement and the consistent large-scale production of highly activated therapeutic memory-like NK cells have yielded encouraging but still unconclusive results. This review provides an overview of NK biology as it relates to cancer immunotherapy and the challenge presented by solid tumors for therapeutic NKs. After contrasting the autologous and allogeneic NK approaches for solid cancer immunotherapy, this work will present the current scientific focus for the production of highly persistent and cytotoxic memory-like NK cells as well as the current issues with production methods as they apply to stress-sensitive immune cells. In conclusion, autologous NK cells for cancer immunotherapy appears to be a prime alternative for front line therapeutics but to be successful, it will be critical to establish comprehensives infrastructures allowing the production of extremely potent NK cells while constraining costs of production.
Collapse
Affiliation(s)
- Gaby D. Lizana-Vasquez
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
- Cancer Research Clinic, Carolina BioOncology Institute (CBOI), Huntersville, NC, United States
| | - Madeline Torres-Lugo
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| | - R. Brent Dixon
- Cancer Research Clinic, Carolina BioOncology Institute (CBOI), Huntersville, NC, United States
- Human Applications Lab (HAL) - BioCytics, Huntersville, NC, United States
| | - John D. Powderly
- Cancer Research Clinic, Carolina BioOncology Institute (CBOI), Huntersville, NC, United States
- Human Applications Lab (HAL) - BioCytics, Huntersville, NC, United States
| | - Renaud F. Warin
- Cancer Research Clinic, Carolina BioOncology Institute (CBOI), Huntersville, NC, United States
- Human Applications Lab (HAL) - BioCytics, Huntersville, NC, United States
| |
Collapse
|
28
|
Piccinelli S, Romee R, Shapiro RM. The natural killer cell immunotherapy platform: an overview of the landscape of clinical trials in liquid and solid tumors. Semin Hematol 2023; 60:42-51. [PMID: 37080710 DOI: 10.1053/j.seminhematol.2023.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
The translation of natural killer (NK) cells to the treatment of malignant disease has made significant progress in the last few decades. With a variety of available sources and improvements in both in vitro and in vivo NK cell expansion, the NK cell immunotherapy platform has come into its own. The enormous effort continues to further optimize this platform, including ways to enhance NK cell persistence, trafficking to the tumor microenvironment, and cytotoxicity. As this effort bears fruit, it is translated into a plethora of clinical trials in patients with advanced malignancies. The adoptive transfer of NK cells, either as a standalone therapy or in combination with other immunotherapies, has been applied for the treatment of both liquid and solid tumors, with numerous early-phase trials showing promising results. This review aims to summarize the key advantages of NK cell immunotherapy, highlight several of the current approaches being taken for its optimization, and give an overview of the landscape of clinical trials translating this platform into clinic.
Collapse
|