1
|
Testa U, Castelli G, Pelosi E. Emerging Role of Chimeric Antigen Receptor-Natural Killer Cells for the Treatment of Hematologic Malignancies. Cancers (Basel) 2025; 17:1454. [PMID: 40361380 PMCID: PMC12071031 DOI: 10.3390/cancers17091454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
The clinical use of T lymphocytes engineered with chimeric antigen receptors (CARs) has revolutionized the treatment of patients with refractory or relapsed hematological malignancies. CAR natural killer (CAR-NK) cells are NK cells engineered with CARs to specifically target cell antigens expressed on the membrane of tumor cells. CAR-NK cells could offer some advantages with respect to CAR-T cells, related to their specific and innate anti-tumor activity, availability as an "off the shelf" cellular therapy, reduced costs, and improved safety. Promising efficacy of CAR-Nk cell therapy was observed in clinical trials based on the treatment of some hematological malignancies. However, to date, the clinical experience of CAR-NK cell therapy has been preliminary, with the evaluation of only a limited number of patients. Furthermore, CAR-NK cell therapy has been limited by the short persistence of these cells and by the suboptimal cytotoxic activity of some CAR-NK preparations. Therefore, studies based on the enrollment of a number of patients is required to carefully assess and confirm the safety and the efficacy of CAR-NK cell therapy in hematological malignancies and to compare their efficacy with respect to allogeneic CAR-T cells.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, 00135 Rome, Italy; (G.C.); (E.P.)
| | | | | |
Collapse
|
2
|
Guo P, Zhong L, Wang T, Luo W, Zhou A, Cao D. NK cell-based immunotherapy for hepatocellular carcinoma: Challenges and opportunities. Scand J Immunol 2025; 101:e13433. [PMID: 39934640 DOI: 10.1111/sji.13433] [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: 09/07/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most challenging malignancies globally, characterized by significant heterogeneity, late-stage diagnosis, and resistance to treatment. In recent years, the advent of immune-checkpoint blockades (ICBs) and targeted immune cell therapies has marked a substantial advancement in HCC treatment. However, the clinical efficacy of these existing therapies is still limited, highlighting the urgent need for new breakthroughs. Natural killer (NK) cells, a subset of the innate lymphoid cell family, have shown unique advantages in the anti-tumour response. With increasing evidence suggesting the crucial role of dysfunctional NK cells in the pathogenesis and progression of HCC, considerable efforts have been directed toward exploring NK cells as a potential therapeutic target for HCC. In this review, we will provide an overview of the role of NK cells in normal liver immunity and in HCC, followed by a detailed discussion of various NK cell-based immunotherapies and their potential applications in HCC treatment.
Collapse
Affiliation(s)
- Pei Guo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Liyuan Zhong
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tao Wang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weijia Luo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Aiqiang Zhou
- Guangzhou Hospital of Integrated Chinese and Western Medicine, Guangzhou, Guangdong, P.R China
| | - Deliang Cao
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
3
|
Diorio C, Teachey DT, Grupp SA. Allogeneic chimeric antigen receptor cell therapies for cancer: progress made and remaining roadblocks. Nat Rev Clin Oncol 2025; 22:10-27. [PMID: 39548270 DOI: 10.1038/s41571-024-00959-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 11/17/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are revolutionizing cancer therapy, particularly for haematological malignancies, conferring durable and sometimes curative responses in patients with advanced-stage disease. The CAR T cell products currently approved for clinical use are all autologous and are often effective; however, in patients who are lymphopenic and/or heavily pretreated with chemotherapy, autologous T cells can be difficult to harvest in sufficient numbers or have functional impairments that might ultimately render them less efficacious. Moreover, autologous products take several weeks to produce, and each product can be used in only one patient. By contrast, allogeneic CAR T cells can be produced for many patients using T cells from a single healthy donor, can be optimized for safety and efficacy, can be instantly available for 'off-the-shelf' use and, therefore, might also be more cost-effective. Despite these potential advantages, the development of allogeneic CAR T cells has lagged behind that of autologous products, owing to the additional challenges such as avoiding graft-versus-host disease and host-mediated graft rejection. Over the past few years, the development of advanced genome-editing techniques has facilitated the generation of novel allogeneic CAR T cell products. Furthermore, CAR cell products derived from other cell types such as induced pluripotent stem cells and natural killer cells are being investigated for clinical use. In this Review, we discuss the potential of allogeneic CAR cell products to expand life-saving immunotherapy to a much broader population of patients in the coming years, the progress made to date and strategies to overcome remaining hurdles.
Collapse
Affiliation(s)
- Caroline Diorio
- Division of Oncology and Center for Childhood Cancer Research, Department of Paediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Susan S. and Stephen P. Kelly Center for Cancer Immunotherapy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David T Teachey
- Division of Oncology and Center for Childhood Cancer Research, Department of Paediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Susan S. and Stephen P. Kelly Center for Cancer Immunotherapy, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephan A Grupp
- Division of Oncology and Center for Childhood Cancer Research, Department of Paediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Susan S. and Stephen P. Kelly Center for Cancer Immunotherapy, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Lajevardi MS, Ashrafpour M, Mubarak SMH, Rafieyan B, Kiani A, Noori E, Roayaei Ardakani M, Montazeri M, Kouhi Esfahani N, Asadimanesh N, Khalili S, Payandeh Z. Dual roles of extracellular vesicles in acute lymphoblastic leukemia: implications for disease progression and theranostic strategies. Med Oncol 2024; 42:11. [PMID: 39572459 PMCID: PMC11582151 DOI: 10.1007/s12032-024-02547-7] [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: 07/11/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024]
Abstract
Acute Lymphoblastic Leukemia (ALL) is a heterogeneous blood cancer characterized by the uncontrolled growth of immature lymphoid cells due to dysregulated signaling pathways. It is the most common pediatric cancer, with high cure rates in children, but significantly lower survival rates in adults. Current theranostic strategies, including chemotherapy, immunotherapy, and nanomedicine, aim to improve detection and treatment precision but are limited by side effects, drug resistance, high costs, and stability issues. Notably, extracellular vesicles (EVs) offer a promising alternative, addressing these limitations through their natural biocompatibility and targeted delivery capabilities. EVs play a dual role in ALL: they contribute to leukemia progression by promoting tumor growth, immune suppression, and drug resistance via the transfer of oncogenic molecules, while also serving as valuable non-invasive biomarkers due to their specific miRNA and protein content. Their ability to deliver therapeutic agents directly to leukemic cells, combined with their stability and low immunogenicity, makes EVs a compelling tool for improving ALL treatments. Indeed, by targeting the molecular pathways influenced by EVs or leveraging them for drug delivery, innovative therapeutic strategies can be developed to enhance treatment outcomes and reduce side effects. Thus, EVs represent a promising frontier for advancing theranostic strategies in ALL, offering new opportunities to improve diagnosis and treatment while overcoming the limitations of traditional therapies. This review will explore the dual roles of EVs in ALL, addressing their contributions to disease progression and their potential as therapeutic agents and biomarkers for early diagnosis and targeted therapies.
Collapse
Affiliation(s)
- Mahya Sadat Lajevardi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Shaden M H Mubarak
- Department of Clinical Laboratory Science, Faculty of Pharmacy, University of Kufa, Kufa, Iraq
| | - Behnoosh Rafieyan
- School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Arash Kiani
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Effat Noori
- Department of Biotechnology, Faculty of Medicine, Shahed University, Tehran, Iran
| | | | - Maryam Montazeri
- Razi Clinical Researches Development, Mazandaran University of Medical Science, Sari, Iran
| | - Niloofar Kouhi Esfahani
- Faculty of Medicine, People's Friendship University of Russia (Rudn University), Moscow, Russia
| | - Naghmeh Asadimanesh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, 1678815811, Iran.
| | - Zahra Payandeh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41346, Gothenburg, Sweden.
| |
Collapse
|
7
|
Cheng W, Kang K, Zhao A, Wu Y. Dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 in lung cancer. J Hematol Oncol 2024; 17:54. [PMID: 39068460 PMCID: PMC11283714 DOI: 10.1186/s13045-024-01581-2] [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: 06/06/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer immunotherapies, represented by immune checkpoint inhibitors (ICIs), have reshaped the treatment paradigm for both advanced non-small cell lung cancer and small cell lung cancer. Programmed death receptor-1/programmed death receptor ligand-1 (PD-1/PD-L1) and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) are some of the most common and promising targets in ICIs. Compared to ICI monotherapy, which occasionally demonstrates treatment resistance and limited efficacy, the dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 operates at different stages of T cell activation with synergistically enhancing immune responses against cancer cells. This emerging dual therapy heralds a new direction for cancer immunotherapy, which, however, may increase the risk of drug-related adverse reactions while improving efficacy. Previous clinical trials have explored combination therapy strategy of anti-PD-1/PD-L1 and anti-CTLA-4 agents in lung cancer, yet its efficacy remains to be unclear with the inevitable incidence of immune-related adverse events. The recent advent of bispecific antibodies has made this sort of dual targeting more feasible, aiming to alleviate toxicity without compromising efficacy. Thus, this review highlights the role of dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 in treating lung cancer, and further elucidates its pre-clinical mechanisms and current advancements in clinical trials. Besides, we also provide novel insights into the potential combinations of dual blockade therapies with other strategies to optimize the future treatment mode for lung cancer.
Collapse
Affiliation(s)
- Weishi Cheng
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Kai Kang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Yijun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
8
|
Graham LV, Khakoo SI, Blunt MD. NK Cells in the Lymph Nodes and Their Role in Anti-Tumour Immunity. Biomedicines 2024; 12:1667. [PMID: 39200132 PMCID: PMC11351147 DOI: 10.3390/biomedicines12081667] [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: 06/12/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
The lymph nodes are vital to enable adaptive immune responses to infection. Natural killer (NK) cells are cytotoxic lymphocytes that directly kill cancer cells and modulate the activation of other immune cells during anti-tumour immune response. NK cells in the lymph nodes are involved in the regulation of T-cell and B-cell populations and the clearance of viral infections. In solid tumours, lymph nodes are a frequent site of metastasis and immune cell priming, whilst in haematological malignancies, tumour cells can proliferate in the lymph nodes. Thus, lymph nodes are an important site in anti-tumour immunity and therapy resistance. It is therefore crucial to identify strategies to increase recruitment and overcome suppression of NK cells in the lymph node microenvironment to improve tumour clearance. In this review, we summarise the literature interrogating NK cell phenotype and function in the lymph nodes in the context of infection and cancer and evaluate both current and potential strategies to mobilise and activate NK cells within the lymph nodes of cancer patients.
Collapse
Affiliation(s)
| | | | - Matthew D. Blunt
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
9
|
Cantoni C, Falco M, Vitale M, Pietra G, Munari E, Pende D, Mingari MC, Sivori S, Moretta L. Human NK cells and cancer. Oncoimmunology 2024; 13:2378520. [PMID: 39022338 PMCID: PMC11253890 DOI: 10.1080/2162402x.2024.2378520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
The long story of NK cells started about 50 y ago with the first demonstration of a natural cytotoxic activity within an undefined subset of circulating leukocytes, has involved an ever-growing number of researchers, fascinated by the apparently easy-to-reach aim of getting a "universal anti-tumor immune tool". In fact, in spite of the impressive progress obtained in the first decades, these cells proved far more complex than expected and, paradoxically, the accumulating findings have continuously moved forward the attainment of a complete control of their function for immunotherapy. The refined studies of these latter years have indicated that NK cells can epigenetically calibrate their functional potential, in response to specific environmental contexts, giving rise to extraordinarily variegated subpopulations, comprehensive of memory-like cells, tissue-resident cells, or cells in various differentiation stages, or distinct functional states. In addition, NK cells can adapt their activity in response to a complex body of signals, spanning from the interaction with either suppressive or stimulating cells (myeloid-derived suppressor cells or dendritic cells, respectively) to the engagement of various receptors (specific for immune checkpoints, cytokines, tumor/viral ligands, or mediating antibody-dependent cell-mediated cytotoxicity). According to this picture, the idea of an easy and generalized exploitation of NK cells is changing, and the way is opening toward new carefully designed, combined and personalized therapeutic strategies, also based on the use of genetically modified NK cells and stimuli capable of strengthening and redirecting their effector functions against cancer.
Collapse
Affiliation(s)
- Claudia Cantoni
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Michela Falco
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Massimo Vitale
- UO Pathology and Experimental Immunology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Gabriella Pietra
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- UO Pathology and Experimental Immunology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Enrico Munari
- Pathology Unit, Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Daniela Pende
- UO Pathology and Experimental Immunology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- UO Pathology and Experimental Immunology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| |
Collapse
|
10
|
Zang J, Mei Y, Zhu S, Yin S, Feng N, Ci T, Lyu Y. Natural Killer-Based Therapy: A Prospective Thought for Cancer Treatment Related to Diversified Drug Delivery Pathways. Pharmaceutics 2024; 16:939. [PMID: 39065636 PMCID: PMC11279587 DOI: 10.3390/pharmaceutics16070939] [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: 06/08/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Immunotherapy has been a research hotspot due to its low side effects, long-lasting efficacy, and wide anti-tumor spectrum. Recently, NK cell-based immunotherapy has gained broad attention for its unique immunological character of tumor identification and eradication and low risk of graft-versus-host disease and cytokine storm. With the cooperation of a drug delivery system (DDS), NK cells activate tumoricidal activity by adjusting the balance of the activating and inhibitory signals on their surface after drug-loaded DDS administration. Moreover, NK cells or NK-derived exosomes can also be applied as drug carriers for distinct modification to promote NK activation and exert anti-tumor effects. In this review, we first introduce the source and classification of NK cells and describe the common activating and inhibitory receptors on their surface. Then, we summarize the strategies for activating NK cells in vivo through various DDSs. Finally, the application prospects of NK cells in tumor immunotherapy are also discussed.
Collapse
Affiliation(s)
- Jing Zang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Z.); (N.F.)
| | - Yijun Mei
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China;
| | - Shiguo Zhu
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
| | - Shaoping Yin
- School of Pharmacy, Jiangsu Provincial Engineering Research Center of Traditional Chinese Medicine External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Nianping Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Z.); (N.F.)
| | - Tianyuan Ci
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Z.); (N.F.)
| | - Yaqi Lyu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Z.); (N.F.)
| |
Collapse
|
11
|
Locatelli F, del Bufalo F, Quintarelli C. Allogeneic chimeric antigen receptor T cells for children with relapsed/refractory B-cell precursor acute lymphoblastic leukemia. Haematologica 2024; 109:1689-1699. [PMID: 38832424 PMCID: PMC11141659 DOI: 10.3324/haematol.2023.284604] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 02/01/2024] [Indexed: 06/05/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as a breakthrough cancer therapy over the past decade. Remarkable outcomes in B-cell lymphoproliferative disorders and multiple myeloma have been reported in both pivotal trials and real-word studies. Traditionally, the use of a patient's own (autologous) T cells to manufacture CAR products has been the standard practice. Nevertheless, this approach has some drawbacks, including manufacturing delays, dependence on the functional fitness of the patient's T cells, which can be compromised by both the disease and prior therapies, and contamination of the product with blasts. A promising alternative is offered by the development of allogeneic CAR-cell products. This approach has the potential to yield more efficient drug products and enables the use of effector cells with negligible alloreactive potential and a significant CAR-independent antitumor activity through their innate receptors (i.e., natural killer cells, γδ T cells and cytokine induced killer cells). In addition, recent advances in genome editing tools offer the potential to overcome the primary challenges associated with allogeneic CAR T-cell products, namely graft-versus-host disease and host allo-rejection, generating universal, off-the-shelf products. In this review, we summarize the current pre-clinical and clinical approaches based on allogeneic CAR T cells, as well as on alternative effector cells, which represent exciting opportunities for multivalent approaches and optimized antitumor activity.
Collapse
Affiliation(s)
- Franco Locatelli
- Department of Hematology/Oncology, Cell and Gene Therapy – IRCCS, Bambino Gesù Children’s Hospital, Rome
- Catholic University of the Sacred Heart, Department of Life Sciences and Public Health, Rome
| | - Francesca del Bufalo
- Department of Hematology/Oncology, Cell and Gene Therapy – IRCCS, Bambino Gesù Children’s Hospital, Rome
| | - Concetta Quintarelli
- Department of Hematology/Oncology, Cell and Gene Therapy – IRCCS, Bambino Gesù Children’s Hospital, Rome
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
12
|
Hassan SH, Alshahrani MY, Saleh RO, Mohammed BA, Kumar A, Almalki SG, Alkhafaji AT, Ghildiyal P, Al-Tameemi AR, Elawady A. A new vision of the efficacy of both CAR-NK and CAR-T cells in treating cancers and autoimmune diseases. Med Oncol 2024; 41:127. [PMID: 38656354 DOI: 10.1007/s12032-024-02362-0] [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: 02/21/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024]
Abstract
Chimeric Antigen Receptor (CAR) based therapies are becoming increasingly important in treating patients. CAR-T cells have been shown to be highly effective in the treatment of hematological malignancies. However, harmful therapeutic barriers have been identified, such as the potential for graft-versus-host disease (GVHD), neurotoxicity, and cytokine release syndrome (CRS). As a result, CAR NK-cell therapy is expected to be a new therapeutic option. NK cells act as cytotoxic lymphocytes, supporting the innate immune response against autoimmune diseases and cancer cells by precisely detecting and eliminating malignant cells. Genetic modification of these cells provides a dual approach to the treatment of AD and cancer. It can be used through both CAR-independent and CAR-dependent mechanisms. The use of CAR-based cell therapies has been successful in treating cancer patients, leading to further investigation of this innovative treatment for alternative diseases, including AD. The complementary roles of CAR T and CAR NK cells have stimulated exploration in this area. Our study examines the latest research on the therapeutic effectiveness of these cells in treating both cancer and ADs.
Collapse
Affiliation(s)
- Salim Hussein Hassan
- Community Health Department, Technical Institute of Karbala, AL-Furat Al-Awsat Technical University, Najaf, Iraq.
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named After the First President of Russia Boris Yeltsin, Ekaterinburg, 620002, Russia
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, 11952, Majmaah, Saudi Arabia
| | | | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Ahmed Elawady
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
13
|
Gao F, You X, Yang L, Zou X, Sui B. Boosting immune responses in lung tumor immune microenvironment: A comprehensive review of strategies and adjuvants. Int Rev Immunol 2024; 43:280-308. [PMID: 38525925 DOI: 10.1080/08830185.2024.2333275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
The immune system has a substantial impact on the growth and expansion of lung malignancies. Immune cells are encompassed by a stroma comprising an extracellular matrix (ECM) and different cells like stromal cells, which are known as the tumor immune microenvironment (TIME). TME is marked by the presence of immunosuppressive factors, which inhibit the function of immune cells and expand tumor growth. In recent years, numerous strategies and adjuvants have been developed to extend immune responses in the TIME, to improve the efficacy of immunotherapy. In this comprehensive review, we outline the present knowledge of immune evasion mechanisms in lung TIME, explain the biology of immune cells and diverse effectors on these components, and discuss various approaches for overcoming suppressive barriers. We highlight the potential of novel adjuvants, including toll-like receptor (TLR) agonists, cytokines, phytochemicals, nanocarriers, and oncolytic viruses, for enhancing immune responses in the TME. Ultimately, we provide a summary of ongoing clinical trials investigating these strategies and adjuvants in lung cancer patients. This review also provides a broad overview of the current state-of-the-art in boosting immune responses in the TIME and highlights the potential of these approaches for improving outcomes in lung cancer patients.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Xiaoqing You
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Oncology, Da Qing Long Nan Hospital, Daqing, Heilongjiang Province, China
| | - Xiangni Zou
- Department of Nursing, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Bowen Sui
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
14
|
Ouyang P, Wang L, Wu J, Tian Y, Chen C, Li D, Yao Z, Chen R, Xiang G, Gong J, Bao Z. Overcoming cold tumors: a combination strategy of immune checkpoint inhibitors. Front Immunol 2024; 15:1344272. [PMID: 38545114 PMCID: PMC10965539 DOI: 10.3389/fimmu.2024.1344272] [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: 11/25/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024] Open
Abstract
Immune Checkpoint Inhibitors (ICIs) therapy has advanced significantly in treating malignant tumors, though most 'cold' tumors show no response. This resistance mainly arises from the varied immune evasion mechanisms. Hence, understanding the transformation from 'cold' to 'hot' tumors is essential in developing effective cancer treatments. Furthermore, tumor immune profiling is critical, requiring a range of diagnostic techniques and biomarkers for evaluation. The success of immunotherapy relies on T cells' ability to recognize and eliminate tumor cells. In 'cold' tumors, the absence of T cell infiltration leads to the ineffectiveness of ICI therapy. Addressing these challenges, especially the impairment in T cell activation and homing, is crucial to enhance ICI therapy's efficacy. Concurrently, strategies to convert 'cold' tumors into 'hot' ones, including boosting T cell infiltration and adoptive therapies such as T cell-recruiting bispecific antibodies and Chimeric Antigen Receptor (CAR) T cells, are under extensive exploration. Thus, identifying key factors that impact tumor T cell infiltration is vital for creating effective treatments targeting 'cold' tumors.
Collapse
Affiliation(s)
- Peng Ouyang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Lijuan Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jianlong Wu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yao Tian
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Caiyun Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Dengsheng Li
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zengxi Yao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Ruichang Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Guoan Xiang
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Jin Gong
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhen Bao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Zhang Y, Zhang C, He M, Xing W, Hou R, Zhang H. Co-expression of IL-21-Enhanced NKG2D CAR-NK cell therapy for lung cancer. BMC Cancer 2024; 24:119. [PMID: 38263004 PMCID: PMC10807083 DOI: 10.1186/s12885-023-11806-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 12/28/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Adoptive cell therapy has achieved great success in treating hematological malignancies. However, the production of chimeric antigen receptor T (CAR-T) cell therapy still faces various difficulties. Natural killer (NK)-92 is a continuously expandable cell line and provides a promising alternative for patient's own immune cells. METHODS We established CAR-NK cells by co-expressing natural killer group 2 member D (NKG2D) and IL-21, and evaluated the efficacy of NKG2D-IL-21 CAR-NK cells in treating lung cancer in vitro and in vivo. RESULTS Our data suggested that the expression of IL-21 effectively increased the cytotoxicity of NKG2D CAR-NK cells against lung cancer cells in a dose-dependent manner and suppressed tumor growth in vitro and in vivo. In addition, the proliferation of NKG2D-IL-21 CAR-NK cells were enhanced while the apoptosis and exhaustion of these cells were suppressed. Mechanistically, IL-21-mediated NKG2D CAR-NK cells function by activating AKT signaling pathway. CONCLUSION Our findings provide a novel option for treating lung cancer using NKG2D-IL-21 CAR-NK cell therapy.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oncology, Shenyang 242 Hospital, 110034, Shenyang, China
| | - Cong Zhang
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, 610072, Chengdu, China
| | - Minghong He
- Department of Respiratory and Critical Care Medicine, Yidu Central Hospital of Weifang, 262500, Weifang, China
| | - Weipeng Xing
- Geneis Beijing Co., Ltd., 100102, Beijing, China
| | - Rui Hou
- Geneis Beijing Co., Ltd., 100102, Beijing, China.
| | - Haijin Zhang
- Department of Respiratory and Critical Care Medicine, Yidu Central Hospital of Weifang, 262500, Weifang, China.
| |
Collapse
|
16
|
Wang Y, Jin S, Zhuang Q, Liu N, Chen R, Adam SA, Jin J, Sun J. Chimeric antigen receptor natural killer cells: a promising antitumor immunotherapy. MedComm (Beijing) 2023; 4:e422. [PMID: 38045827 PMCID: PMC10691297 DOI: 10.1002/mco2.422] [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: 05/09/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 12/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have been successfully used in adoptive cell therapy for malignancies. However, some obstacles, including side effects such as graft-versus-host disease and cytokine release syndrome, therapy resistance, limited sources, as well as high cost, limited the application of CAR T cells. Recently, CAR natural killer (NK) cells have been pursued as the effector cells for adoptive immunotherapy for their attractive merits of strong intrinsic antitumor activity and relatively mild side effects. Additionally, CAR NK cells can be available from various sources and do not require strict human leukocyte antigen matching, which suggests them as promising "off-the-shelf" products for clinical application. Although the use of CAR NK cells is restrained by the limited proliferation and impaired efficiency within the immunosuppressive tumor microenvironment, further investigation in optimizing CAR structure and combination therapies will overcome these challenges. This review will summarize the advancement of CAR NK cells, CAR NK cell manufacture, the clinical outcomes of CAR NK therapy, the challenges in the field, and prospective solutions. Besides, we will discuss the emerging application of other immune cells for CAR engineering. Collectively, this comprehensive review will provide a valuable and informative summary of current progress and evaluate challenges and future opportunities of CAR NK cells in tumor treatment.
Collapse
Affiliation(s)
- Yan Wang
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Shengjie Jin
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Qiqi Zhuang
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Na Liu
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Department of OncologyAffiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifangShandongChina
| | - Ruyi Chen
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Sofia Abdulkadir Adam
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Jie Jin
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Zhejiang University Cancer CenterHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouZhejiangChina
| | - Jie Sun
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouZhejiangChina
| |
Collapse
|
17
|
Hojjatipour T, Sharifzadeh Z, Maali A, Azad M. Chimeric antigen receptor-natural killer cells: a promising sword against insidious tumor cells. Hum Cell 2023; 36:1843-1864. [PMID: 37477869 DOI: 10.1007/s13577-023-00948-w] [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: 04/01/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023]
Abstract
Natural killer (NK) cells are a critical component of innate immunity, particularly in initial cancer recognition and inhibition of additional tumor growth or metastasis propagation. NK cells recognize transformed cells without prior sensitization via stimulatory receptors and rapidly eradicate them. However, the protective tumor microenvironment facilitates tumor escaping via induction of an exhaustion state in immune cells, including NK cells. Hence, genetic manipulation of NK cells for specific identification of tumor-associated antigens or a more robust response against tumor cells is a promising strategy for NK cells' tumoricidal augmentation. Regarding the remarkable achievement of engineered CAR-T cells in treating hematologic malignancies, there is evolving interest in CAR-NK cell recruitment in cancer immunotherapy. Innate functionality of NK cells, higher safety, superior in vivo maintenance, and the off-the-shelf potential move CAR-NK-based therapy superior to CAR-T cells treatment. In this review, we have comprehensively discussed the recent genetic manipulations of CAR-NK cell manufacturing regarding different domains of CAR constructs and their following delivery systems into diverse sources of NK cells. Then highlight the preclinical and clinical investigations of CAR-NK cells and examine the current challenges and prospects as an optimistic remedy in cancer immunotherapy.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Department of Hematology and Blood Transfusion, Students Research Center, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciecnes, Qazvin, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, 3419759811, Iran.
| |
Collapse
|
18
|
Zhang Y, Zhou W, Yang J, Yang J, Wang W. Chimeric antigen receptor engineered natural killer cells for cancer therapy. Exp Hematol Oncol 2023; 12:70. [PMID: 37563648 PMCID: PMC10413722 DOI: 10.1186/s40164-023-00431-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
Natural killer (NK) cells, a unique component of the innate immune system, are inherent killers of stressed and transformed cells. Based on their potent capacity to kill cancer cells and good tolerance of healthy cells, NK cells have been successfully employed in adoptive cell therapy to treat cancer patients. In recent years, the clinical success of chimeric antigen receptor (CAR)-T cells has proven the vast potential of gene-manipulated immune cells as the main force to fight cancer. Following the lessons learned from mature gene-transfer technologies and advanced strategies in CAR-T therapy, NK cells have been rapidly explored as a promising candidate for CAR-based therapy. An exponentially growing number of studies have employed multiple sources of CAR-NK cells to target a wide range of cancer-related antigens, showing remarkable outcomes and encouraging safety profiles. Clinical trials of CAR-NK cells have also shown their impressive therapeutic efficacy in the treatment of hematological tumors, but CAR-NK cell therapy for solid tumors is still in the initial stages. In this review, we present the favorable profile of NK cells as a potential platform for CAR-based engineering and then summarize the outcomes and strategies of CAR-NK therapies in up-to-date preclinical and clinical investigations. Finally, we evaluate the challenges remaining in CAR-NK therapy and describe existing strategies that can assist us in devising future prospective solutions.
Collapse
Affiliation(s)
- Yalan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Jiangping Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jinrong Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China
- Hematology Research Laboratory, Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
19
|
Shin MH, Oh E, Kim Y, Nam DH, Jeon SY, Yu JH, Minn D. Recent Advances in CAR-Based Solid Tumor Immunotherapy. Cells 2023; 12:1606. [PMID: 37371075 DOI: 10.3390/cells12121606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Adoptive cell therapy using chimeric antigen receptor (CAR) technology is one of the most advanced engineering platforms for cancer immunotherapy. CAR-T cells have shown remarkable efficacy in the treatment of hematological malignancies. However, their limitations in solid tumors include an immunosuppressive tumor microenvironment (TME), insufficient tumor infiltration, toxicity, and the absence of tumor-specific antigens. Although recent advances in CAR-T cell design-such as the incorporation of co-stimulatory domains and the development of armored CAR-T cells-have shown promising results in treating solid tumors, there are still challenges that need to be addressed. To overcome these limitations, other immune cells, such as natural killer (NK) cells and macrophages (M), have been developed as attractive options for efficient cancer immunotherapy of solid tumors. CAR-NK cells exhibit substantial clinical improvements with "off-the-shelf" availability and low toxicity. CAR-M cells have promising therapeutic potential because macrophages can infiltrate the TME of solid tumors. Here, we review the recent advances and future perspectives associated with engineered immune cell-based cancer immunotherapies for solid tumors. We also summarize ongoing clinical trials investigating the safety and efficacy of engineered immune cells, such as CAR-T, CAR-NK, and CAR-M, for targeting solid tumors.
Collapse
Affiliation(s)
- Min Hwa Shin
- Immune Research Institute, Seegene Medical Foundation, Seoul 04805, Republic of Korea
| | - Eunha Oh
- Immune Research Institute, Seegene Medical Foundation, Seoul 04805, Republic of Korea
| | - Yunjeong Kim
- Immune Research Institute, Seegene Medical Foundation, Seoul 04805, Republic of Korea
| | - Dae-Hwan Nam
- Immune Research Institute, Seegene Medical Foundation, Seoul 04805, Republic of Korea
| | - So Young Jeon
- Immune Research Institute, Seegene Medical Foundation, Seoul 04805, Republic of Korea
| | - Jin Hyuk Yu
- Immune Research Institute, Seegene Medical Foundation, Seoul 04805, Republic of Korea
| | - Dohsik Minn
- Immune Research Institute, Seegene Medical Foundation, Seoul 04805, Republic of Korea
- Department of Diagnostic Immunology, Seegene Medical Foundation, Seoul 04805, Republic of Korea
| |
Collapse
|
20
|
Berrien-Elliott MM, Jacobs MT, Fehniger TA. Allogeneic natural killer cell therapy. Blood 2023; 141:856-868. [PMID: 36416736 PMCID: PMC10023727 DOI: 10.1182/blood.2022016200] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Interest in adoptive cell therapy for treating cancer is exploding owing to early clinical successes of autologous chimeric antigen receptor (CAR) T lymphocyte therapy. However, limitations using T cells and autologous cell products are apparent as they (1) take weeks to generate, (2) utilize a 1:1 donor-to-patient model, (3) are expensive, and (4) are prone to heterogeneity and manufacturing failures. CAR T cells are also associated with significant toxicities, including cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and prolonged cytopenias. To overcome these issues, natural killer (NK) cells are being explored as an alternative cell source for allogeneic cell therapies. NK cells have an inherent ability to recognize cancers, mediate immune functions of killing and communication, and do not induce graft-versus-host disease, cytokine release syndrome, or immune effector cell-associated neurotoxicity syndrome. NK cells can be obtained from blood or cord blood or be derived from hematopoietic stem and progenitor cells or induced pluripotent stem cells, and can be expanded and cryopreserved for off-the-shelf availability. The first wave of point-of-care NK cell therapies led to the current allogeneic NK cell products being investigated in clinical trials with promising preliminary results. Basic advances in NK cell biology and cellular engineering have led to new translational strategies to block inhibition, enhance and broaden target cell recognition, optimize functional persistence, and provide stealth from patients' immunity. This review details NK cell biology, as well as NK cell product manufacturing, engineering, and combination therapies explored in the clinic leading to the next generation of potent, off-the-shelf cellular therapies for blood cancers.
Collapse
Affiliation(s)
| | - Miriam T. Jacobs
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Todd A. Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
21
|
He B, Mai Q, Pang Y, Deng S, He Y, Xue R, Xu N, Zhou H, Liu X, Xuan L, Li C, Liu Q. Cytokines induced memory-like NK cells engineered to express CD19 CAR exhibit enhanced responses against B cell malignancies. Front Immunol 2023; 14:1130442. [PMID: 37207215 PMCID: PMC10191231 DOI: 10.3389/fimmu.2023.1130442] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
CD19 chimeric antigen receptor (CAR) engineered NK cells have been used for treating patients with relapsed and/or refractory B cell malignancies and show encouraging outcomes and safety profile. However, the poor persistence of NK cells remains a major challenge for CAR NK cell therapy. Memory-like NK cells (MLNK) induced by IL-12, IL-15, and IL-18 have shown enhanced and prolonged responses to tumor re-stimulation, making them an attractive candidate for adoptive cellular immunotherapy. Here, we show efficient and stable gene delivery of CD19 CAR to memory-like NK cells using retroviral vectors with transduction efficiency comparable to those achieved with conventional NK cells. Analysis of surface molecules revealed a distinct phenotypic profile in CAR engineered memory-like NK cells (CAR MLNK), as evidenced by increased expression of CD94 and downregulation of NKp30 as well as KIR2DL1. Compared to conventional CAR NK cells, CAR MLNK cells exhibited significantly increased IFN-γ production and degranulation in response to CD19+ target cells, resulting in enhanced cytotoxic activity against CD19+ leukemia cells and lymphoma cells. Furthermore, memory properties induced by IL-12/-15/-18 improved the in vivo persistence of CAR MLNK cells and significantly suppressed tumor growth in a exnograft mouse model of lymphoma, leading to prolonged survival of CD19+ tumor-bearing mouse. Altogether, our data indicate that CD19 CAR engineered memory-like NK cells exhibited superior persistence and antitumor activity against CD19+ tumors, which might be an attractive approach for treating patient with relapse or refractory B cell malignancies.
Collapse
Affiliation(s)
- Bailin He
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiusui Mai
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yunyi Pang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Shikai Deng
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Rongtao Xue
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoli Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- *Correspondence: Qifa Liu, ; Chengyao Li,
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Qifa Liu, ; Chengyao Li,
| |
Collapse
|
22
|
El Marsafy S, Larghero J. Cancer Cell De-Differentiation: Plasticity-Driven Stratagem For Tumor Metastasis and Recurrence. Curr Stem Cell Res Ther 2023; 18:54-61. [PMID: 35676837 DOI: 10.2174/1574888x17666220608101852] [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: 02/14/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022]
Abstract
Tumor recurrence is a colossal challenge in clinical oncology. This multifactorial problem is attributed to the emergence of additional genetic mutations and the presence of dormant cancer cells. However, the plasticity of non-stem cancer cells and the acquisition of cancer stem cell (CSC) functionality is another contributing factor to tumor recurrence. Herein, I focus attention on the mechanisms that fuel cancer cell de-differentiation and the interplay between intra-cellular regulators and tumor microenvironment (TME) landscape that promotes cancer cell stemness. Our understanding of the mechanisms underlying tumor cell de-differentiation is crucial for developing innovative therapeutic strategies that prevent cancer from ever recurring.
Collapse
|
23
|
Gribkova IV. CAR NK-сells for the treatment of hematological malignancies: A review. JOURNAL OF MODERN ONCOLOGY 2022. [DOI: 10.26442/18151434.2022.3.201699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hematological malignant neoplasms include more than a hundred different subtypes and account for about 4.8% of all neoplastic diseases in Russia. Despite significant advances in diagnosis and treatment, many of them remain incurable. In recent years, cell-based therapy appears to be a promising approach to the treatment of these incurable hematologic malignancies, showing striking results in various clinical trials. The most studied and advanced cell therapy is the therapy with T-lymphocytes modified with chimeric antigen receptors (CAR). However, although the US Food and Drug Administration has approved CAR T cells for the treatment of B-cell lymphoma and acute lymphoblastic leukemia, significant problems remain in terms of production, cost, and serious side effects. An alternative to the use of T cells can be the use of innate immune cells, in particular natural killer cells (NK), which have a high antitumor potential. Recent studies have shown the antitumor efficacy of a therapy that uses genetically modified natural killer cells CAR NK cells. The purpose of this review was to describe and systematize the experience of using CAR NK cells for the treatment of hematological neoplasms. The review presents the advantages and disadvantages of this method, as well as the problems that still have to be solved for its widespread introduction into clinical practice.
Collapse
|
24
|
Current and Future Perspectives for Chimeric Antigen Receptor T Cells Development in Poland. Biomedicines 2022; 10:biomedicines10112912. [PMID: 36428480 PMCID: PMC9687915 DOI: 10.3390/biomedicines10112912] [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: 10/06/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cells are genetically modified autologous T cells that have revolutionized the treatment of relapsing and refractory haematological malignancies. In this review we present molecular pathways involved in the activation of CAR-T cells, describe in details the structures of receptors and the biological activity of CAR-T cells currently approved for clinical practice in the European Union, and explain the functional differences between them. Finally, we present the potential for the development of CAR-T cells in Poland, as well as indicate the possible directions of future research in this area, including novel modifications and applications of CAR-T cells and CAR-natural killer (NK) cells.
Collapse
|
25
|
Caruso S, De Angelis B, Del Bufalo F, Ciccone R, Donsante S, Volpe G, Manni S, Guercio M, Pezzella M, Iaffaldano L, Silvestris DA, Sinibaldi M, Di Cecca S, Pitisci A, Velardi E, Merli P, Algeri M, Lodi M, Paganelli V, Serafini M, Riminucci M, Locatelli F, Quintarelli C. Safe and effective off-the-shelf immunotherapy based on CAR.CD123-NK cells for the treatment of acute myeloid leukaemia. J Hematol Oncol 2022; 15:163. [PMID: 36335396 PMCID: PMC9636687 DOI: 10.1186/s13045-022-01376-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/04/2022] [Indexed: 11/08/2022] Open
Abstract
Background Paediatric acute myeloid leukaemia (AML) is characterized by poor outcomes in patients with relapsed/refractory disease, despite the improvements in intensive standard therapy. The leukaemic cells of paediatric AML patients show high expression of the CD123 antigen, and this finding provides the biological basis to target CD123 with the chimeric antigen receptor (CAR). However, CAR.CD123 therapy in AML is hampered by on-target off-tumour toxicity and a long “vein-to-vein” time.
Methods We developed an off-the-shelf product based on allogeneic natural killer (NK) cells derived from the peripheral blood of healthy donors and engineered them to express a second-generation CAR targeting CD123 (CAR.CD123). Results CAR.CD123-NK cells showed significant anti-leukaemia activity not only in vitro against CD123+ AML cell lines and CD123+ primary blasts but also in two animal models of human AML-bearing immune-deficient mice. Data on anti-leukaemia activity were also corroborated by the quantification of inflammatory cytokines, namely granzyme B (Granz B), interferon gamma (IFN-γ) and tumour necrosis factor alpha (TNF-α), both in vitro and in the plasma of mice treated with CAR.CD123-NK cells.
To evaluate and compare the on-target off-tumour effects of CAR.CD123-T and NK cells, we engrafted human haematopoietic cells (hHCs) in an immune-deficient mouse model. All mice infused with CAR.CD123-T cells died by Day 5, developing toxicity against primary human bone marrow (BM) cells with a decreased number of total hCD45+ cells and, in particular, of hCD34+CD38− stem cells. In contrast, treatment with CAR.CD123-NK cells was not associated with toxicity, and all mice were alive at the end of the experiments. Finally, in a mouse model engrafted with human endothelial tissues, we demonstrated that CAR.CD123-NK cells were characterized by negligible endothelial toxicity when compared to CAR.CD123-T cells.
Conclusions Our data indicate the feasibility of an innovative off-the-shelf therapeutic strategy based on CAR.CD123-NK cells, characterized by remarkable efficacy and an improved safety profile compared to CAR.CD123-T cells. These findings open a novel intriguing scenario not only for the treatment of refractory/resistant AML patients but also to further investigate the use of CAR-NK cells in other cancers characterized by highly difficult targeting with the most conventional T effector cells.
Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01376-3.
Collapse
Affiliation(s)
- Simona Caruso
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Biagio De Angelis
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Francesca Del Bufalo
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Roselia Ciccone
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Samantha Donsante
- grid.7841.aDepartment of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Gabriele Volpe
- grid.414125.70000 0001 0727 6809Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Simona Manni
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Marika Guercio
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Michele Pezzella
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Laura Iaffaldano
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Domenico Alessandro Silvestris
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Matilde Sinibaldi
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Stefano Di Cecca
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Angela Pitisci
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Enrico Velardi
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Pietro Merli
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Mattia Algeri
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Mariachiara Lodi
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Valeria Paganelli
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Marta Serafini
- grid.7563.70000 0001 2174 1754Department of Pediatrics, Tettamanti Research Center, Fondazione MBBM/San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Mara Riminucci
- grid.7841.aDepartment of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Franco Locatelli
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy ,grid.8142.f0000 0001 0941 3192Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Concetta Quintarelli
- grid.414125.70000 0001 0727 6809Department of Oncology-Haematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy ,grid.4691.a0000 0001 0790 385XDepartment of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
26
|
Castiello L, Santodonato L, Napolitano M, Carlei D, Montefiore E, Monque DM, D’Agostino G, Aricò E. Chimeric Antigen Receptor Immunotherapy for Solid Tumors: Choosing the Right Ingredients for the Perfect Recipe. Cancers (Basel) 2022; 14:5351. [PMID: 36358770 PMCID: PMC9655484 DOI: 10.3390/cancers14215351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 10/21/2023] Open
Abstract
Chimeric antigen receptor T cell therapies are revolutionizing the clinical practice of hematological tumors, whereas minimal progresses have been achieved in the solid tumor arena. Multiple reasons have been ascribed to this slower pace: The higher heterogeneity, the hurdles of defining reliable tumor antigens to target, and the broad repertoire of immune escape strategies developed by solid tumors are considered among the major ones. Currently, several CAR therapies are being investigated in preclinical and early clinical trials against solid tumors differing in the type of construct, the cells that are engineered, and the additional signals included with the CAR constructs to overcome solid tumor barriers. Additionally, novel approaches in development aim at overcoming some of the limitations that emerged with the approved therapies, such as large-scale manufacturing, duration of manufacturing, and logistical issues. In this review, we analyze the advantages and challenges of the different approaches under development, balancing the scientific evidences supporting specific choices with the manufacturing and regulatory issues that are essential for their further clinical development.
Collapse
Affiliation(s)
- Luciano Castiello
- Cell Factory FaBioCell, Core Facilities, Italian National Institute of Health, 00161 Rome, Italy
| | - Laura Santodonato
- Cell Factory FaBioCell, Core Facilities, Italian National Institute of Health, 00161 Rome, Italy
| | - Mariarosaria Napolitano
- Research Coordination and Support Service, Italian National Institute of Health, 00161 Rome, Italy
| | - Davide Carlei
- Cell Factory FaBioCell, Core Facilities, Italian National Institute of Health, 00161 Rome, Italy
| | - Enrica Montefiore
- Cell Factory FaBioCell, Core Facilities, Italian National Institute of Health, 00161 Rome, Italy
| | - Domenica Maria Monque
- Cell Factory FaBioCell, Core Facilities, Italian National Institute of Health, 00161 Rome, Italy
| | - Giuseppina D’Agostino
- Cell Factory FaBioCell, Core Facilities, Italian National Institute of Health, 00161 Rome, Italy
| | - Eleonora Aricò
- Cell Factory FaBioCell, Core Facilities, Italian National Institute of Health, 00161 Rome, Italy
| |
Collapse
|
27
|
Li H, Song W, Li Z, Zhang M. Preclinical and clinical studies of CAR-NK-cell therapies for malignancies. Front Immunol 2022; 13:992232. [PMID: 36353643 PMCID: PMC9637940 DOI: 10.3389/fimmu.2022.992232] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 12/01/2022] Open
Abstract
The development of chimeric antigen receptor T (CAR-T) cell therapy, a specific type of immunotherapy, in recent decades was a fantastic breakthrough for the treatment of hematological malignancies. However, difficulties in collecting normal T cells from patients and the time cost of manufacturing CAR-T cells have limited the application of CAR-T-cell therapy. In addition, the termination of related clinical trials on universal CAR-T cell therapy has made further research more difficult. Natural killer (NK) cells have drawn great attention in recent years. Chimeric antigen receptor-NK (CAR-NK) cell therapy is a promising strategy in the treatment of malignant tumors because of its lack of potential for causing graft-versus-host disease (GVHD). In this review, we will address the advances in and achievements of CAR-NK cell therapy.
Collapse
Affiliation(s)
- Hongwen Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenting Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Mingzhi Zhang,
| |
Collapse
|
28
|
Yaping W, Zhe W, Zhuling C, Ruolei L, Pengyu F, Lili G, Cheng J, Bo Z, Liuyin L, Guangdong H, Yaoling W, Niuniu H, Rui L. The soldiers needed to be awakened: Tumor-infiltrating immune cells. Front Genet 2022; 13:988703. [PMID: 36246629 PMCID: PMC9558824 DOI: 10.3389/fgene.2022.988703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
In the tumor microenvironment, tumor-infiltrating immune cells (TIICs) are a key component. Different types of TIICs play distinct roles. CD8+ T cells and natural killer (NK) cells could secrete soluble factors to hinder tumor cell growth, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) release inhibitory factors to promote tumor growth and progression. In the meantime, a growing body of evidence illustrates that the balance between pro- and anti-tumor responses of TIICs is associated with the prognosis in the tumor microenvironment. Therefore, in order to boost anti-tumor response and improve the clinical outcome of tumor patients, a variety of anti-tumor strategies for targeting TIICs based on their respective functions have been developed and obtained good treatment benefits, including mainly immune checkpoint blockade (ICB), adoptive cell therapies (ACT), chimeric antigen receptor (CAR) T cells, and various monoclonal antibodies. In recent years, the tumor-specific features of immune cells are further investigated by various methods, such as using single-cell RNA sequencing (scRNA-seq), and the results indicate that these cells have diverse phenotypes in different types of tumors and emerge inconsistent therapeutic responses. Hence, we concluded the recent advances in tumor-infiltrating immune cells, including functions, prognostic values, and various immunotherapy strategies for each immune cell in different tumors.
Collapse
Affiliation(s)
- Wang Yaping
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Zhe
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Chu Zhuling
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
| | - Li Ruolei
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Fan Pengyu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Guo Lili
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ji Cheng
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhang Bo
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Liu Liuyin
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hou Guangdong
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Yaoling
- Department of Geriatrics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hou Niuniu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| | - Ling Rui
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| |
Collapse
|
29
|
Zhao Y, Bai Y, Shen M, Li Y. Therapeutic strategies for gastric cancer targeting immune cells: Future directions. Front Immunol 2022; 13:992762. [PMID: 36225938 PMCID: PMC9549957 DOI: 10.3389/fimmu.2022.992762] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer (GC) is a malignancy with a high incidence and mortality, and the emergence of immunotherapy has brought survival benefits to GC patients. Compared with traditional therapy, immunotherapy has the advantages of durable response, long-term survival benefits, and lower toxicity. Therefore, targeted immune cells are the most promising therapeutic strategy in the field of oncology. In this review, we introduce the role and significance of each immune cell in the tumor microenvironment of GC and summarize the current landscape of immunotherapy in GC, which includes immune checkpoint inhibitors, adoptive cell therapy (ACT), dendritic cell (DC) vaccines, reduction of M2 tumor-associated macrophages (M2 TAMs), N2 tumor-associated neutrophils (N2 TANs), myeloid-derived suppressor cells (MDSCs), effector regulatory T cells (eTregs), and regulatory B cells (Bregs) in the tumor microenvironment and reprogram TAMs and TANs into tumor killer cells. The most widely used immunotherapy strategies are the immune checkpoint inhibitor programmed cell death 1/programmed death-ligand 1 (PD-1/PD-L1) antibody, cytotoxic T lymphocyte–associated protein 4 (CTLA-4) antibody, and chimeric antigen receptor T (CAR-T) in ACT, and these therapeutic strategies have significant anti-tumor efficacy in solid tumors and hematological tumors. Targeting other immune cells provides a new direction for the immunotherapy of GC despite the relatively weak clinical data, which have been confirmed to restore or enhance anti-tumor immune function in preclinical studies and some treatment strategies have entered the clinical trial stage, and it is expected that more and more effective immune cell–based therapeutic methods will be developed and applied.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuansong Bai
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meili Shen
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Yapeng Li, ; Meili Shen,
| | - Yapeng Li
- The National and Local Joint Engineering Laboratory for Synthesis Technology of High Performance Polymer, College of Chemistry, Jilin University, Changchun, China
- *Correspondence: Yapeng Li, ; Meili Shen,
| |
Collapse
|
30
|
Ragoonanan D, Sheikh IN, Gupta S, Khazal SJ, Tewari P, Petropoulos D, Li S, Mahadeo KM. The Evolution of Chimeric Antigen Receptor T-Cell Therapy in Children, Adolescents and Young Adults with Acute Lymphoblastic Leukemia. Biomedicines 2022; 10:biomedicines10092286. [PMID: 36140387 PMCID: PMC9496125 DOI: 10.3390/biomedicines10092286] [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: 10/31/2021] [Revised: 08/05/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor T-cell (CAR T) therapy is a revolutionary treatment for pediatric, adolescent and young adult patients (AYA) with relapsed/refractory B-cell acute lymphoblastic leukemia. While the landscape of immunotherapy continues to rapidly evolve, widespread use of CAR T therapy is limited and many questions remain regarding the durability of CAR T therapy, methods to avoid CAR T therapy resistance and the role of consolidative stem cell transplant. Modified strategies to develop effective and persistent CAR T cells at lower costs and decreased toxicities are warranted. In this review we present current indications, limitations and future directions of CAR T therapy for ALL in the pediatric and AYA population.
Collapse
Affiliation(s)
- Dristhi Ragoonanan
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (D.R.); (I.N.S.)
| | - Irtiza N. Sheikh
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (D.R.); (I.N.S.)
| | - Sumit Gupta
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sajad J. Khazal
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Priti Tewari
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Demetrios Petropoulos
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shulin Li
- Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kris M. Mahadeo
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
31
|
Todorovic Z, Todorovic D, Markovic V, Ladjevac N, Zdravkovic N, Djurdjevic P, Arsenijevic N, Milovanovic M, Arsenijevic A, Milovanovic J. CAR T Cell Therapy for Chronic Lymphocytic Leukemia: Successes and Shortcomings. Curr Oncol 2022; 29:3647-3657. [PMID: 35621683 PMCID: PMC9139644 DOI: 10.3390/curroncol29050293] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor T (CAR T) cell therapy achieved remarkable success in B-cell leukemia and lymphoma which led to its incorporation in treatment protocols for these diseases. CAR T cell therapy for chronic lymphocytic leukemia (CLL) patients showed less success compared to other malignant tumors. In this review, we discuss the published results regarding CAR T cell therapy of CLL, possible mechanisms of failures and expected developments.
Collapse
Affiliation(s)
- Zeljko Todorovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.T.); (N.Z.); (P.D.)
| | - Dusan Todorovic
- Department of Ophthalmology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Vladimir Markovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.L.); (N.A.); (M.M.)
| | - Nevena Ladjevac
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.L.); (N.A.); (M.M.)
| | - Natasa Zdravkovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.T.); (N.Z.); (P.D.)
| | - Predrag Djurdjevic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.T.); (N.Z.); (P.D.)
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.L.); (N.A.); (M.M.)
| | - Marija Milovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.L.); (N.A.); (M.M.)
| | - Aleksandar Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.L.); (N.A.); (M.M.)
| | - Jelena Milovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.L.); (N.A.); (M.M.)
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
32
|
Soldierer M, Bister A, Haist C, Thivakaran A, Cengiz SC, Sendker S, Bartels N, Thomitzek A, Smorra D, Hejazi M, Uhrberg M, Scheckenbach K, Monzel C, Wiek C, Reinhardt D, Niktoreh N, Hanenberg H. Genetic Engineering and Enrichment of Human NK Cells for CAR-Enhanced Immunotherapy of Hematological Malignancies. Front Immunol 2022; 13:847008. [PMID: 35464442 PMCID: PMC9022481 DOI: 10.3389/fimmu.2022.847008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 01/11/2023] Open
Abstract
The great clinical success of chimeric antigen receptor (CAR) T cells has unlocked new levels of immunotherapy for hematological malignancies. Genetically modifying natural killer (NK) cells as alternative CAR immune effector cells is also highly promising, as NK cells can be transplanted across HLA barriers without causing graft-versus-host disease. Therefore, off-the-shelf usage of CAR NK cell products might allow to widely expand the clinical indications and to limit the costs of treatment per patient. However, in contrast to T cells, manufacturing suitable CAR NK cell products is challenging, as standard techniques for genetically engineering NK cells are still being defined. In this study, we have established optimal lentiviral transduction of primary human NK cells by systematically testing different internal promoters for lentiviral CAR vectors and comparing lentiviral pseudotypes and viral entry enhancers. We have additionally modified CAR constructs recognizing standard target antigens for acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) therapy—CD19, CD33, and CD123—to harbor a CD34-derived hinge region that allows efficient detection of transduced NK cells in vitro and in vivo and also facilitates CD34 microbead-assisted selection of CAR NK cell products to >95% purity for potential clinical usage. Importantly, as most leukemic blasts are a priori immunogenic for activated primary human NK cells, we developed an in vitro system that blocks the activating receptors NKG2D, DNAM-1, NKp30, NKp44, NKp46, and NKp80 on these cells and therefore allows systematic testing of the specific killing of CAR NK cells against ALL and AML cell lines and primary AML blasts. Finally, we evaluated in an ALL xenotransplantation model in NOD/SCID-gamma (NSG) mice whether human CD19 CAR NK cells directed against the CD19+ blasts are relying on soluble or membrane-bound IL15 production for NK cell persistence and also in vivo leukemia control. Hence, our study provides important insights into the generation of pure and highly active allogeneic CAR NK cells, thereby advancing adoptive cellular immunotherapy with CAR NK cells for human malignancies further.
Collapse
Affiliation(s)
- Maren Soldierer
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Arthur Bister
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Corinna Haist
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Aniththa Thivakaran
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sevgi Can Cengiz
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephanie Sendker
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nina Bartels
- Department of Experimental Medical Physics, Heinrich Heine University, Düsseldorf, Germany
| | - Antonia Thomitzek
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Denise Smorra
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Maryam Hejazi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Kathrin Scheckenbach
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Cornelia Monzel
- Department of Experimental Medical Physics, Heinrich Heine University, Düsseldorf, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Dirk Reinhardt
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Naghmeh Niktoreh
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Helmut Hanenberg
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
33
|
Tarannum M, Romee R, Shapiro RM. Innovative Strategies to Improve the Clinical Application of NK Cell-Based Immunotherapy. Front Immunol 2022; 13:859177. [PMID: 35401529 PMCID: PMC8990319 DOI: 10.3389/fimmu.2022.859177] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022] Open
Abstract
Natural killer cells constitute a part of the innate immune system that mediates an effective immune response towards virus-infected and malignant cells. In recent years, research has focused on exploring and advancing NK cells as an active immunotherapy platform. Despite major advances, there are several key challenges that need to be addressed for the effective translation of NK cell research to clinical applications. This review highlights some of these challenges and the innovative strategies being developed to overcome them, including in vitro expansion, in vivo persistence, infiltration to the tumor site, and prevention of exhaustion.
Collapse
Affiliation(s)
- Mubin Tarannum
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Rizwan Romee
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Roman M Shapiro
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
34
|
Zhang L, Meng Y, Feng X, Han Z. CAR-NK cells for cancer immunotherapy: from bench to bedside. Biomark Res 2022; 10:12. [PMID: 35303962 PMCID: PMC8932134 DOI: 10.1186/s40364-022-00364-6] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/08/2022] [Indexed: 02/08/2023] Open
Abstract
Natural killer (NK) cells are unique innate immune cells and manifest rapid and potent cytotoxicity for cancer immunotherapy and pathogen removal without the requirement of prior sensitization or recognition of peptide antigens. Distinguish from the T lymphocyte-based cythotherapy with toxic side effects, chimeric antigen receptor-transduced NK (CAR-NK) cells are adequate to simultaneously improve efficacy and control adverse effects including acute cytokine release syndrome (CRS), neurotoxicity and graft-versus-host disease (GVHD). Moreover, considering the inherent properties of NK cells, the CAR-NK cells are “off-the-shelf” product satisfying the clinical demand for large-scale manufacture for cancer immunotherapy attribute to the cytotoxic effect via both NK cell receptor-dependent and CAR-dependent signaling cascades. In this review, we mainly focus on the latest updates of CAR-NK cell-based tactics, together with the opportunities and challenges for cancer immunotherapies, which represent the paradigm for boosting the immune system to enhance antitumor responses and ultimately eliminate malignancies. Collectively, we summarize and highlight the auspicious improvement in CAR-NK cells and will benefit the large-scale preclinical and clinical investigations in adoptive immunotherapy.
Collapse
Affiliation(s)
- Leisheng Zhang
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province & NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China. .,Center for Cellular Therapies, The First Affiliated Hospital of Shandong First Medical University, Ji-nan, 250014, China. .,Key Laboratory of Radiation Technology and Biophysics, Hefei Institute of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Shushan District, Hefei, 230031, Anhui Province, China. .,Institute of Stem Cells, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China. .,Jiangxi Research Center of Stem Cell Engineering, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd., Shangrao, 334000, China. .,Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, 204 Donggangxi Road, Chengguan District, Lanzhou City, 730013, Gansu Province, China.
| | - Yuan Meng
- State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| | - Zhongchao Han
- Institute of Stem Cells, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China. .,Jiangxi Research Center of Stem Cell Engineering, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd., Shangrao, 334000, China. .,State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China. .,Stem Cell Bank of Guizhou Province, Guizhou Health-Biotech Biotechnology Co., Ltd., Guiyang, 550000, China.
| |
Collapse
|
35
|
Morton LT, Wachsmann TLA, Meeuwsen MH, Wouters AK, Remst DFG, van Loenen MM, Falkenburg JHF, Heemskerk MHM. T cell receptor engineering of primary NK cells to therapeutically target tumors and tumor immune evasion. J Immunother Cancer 2022; 10:jitc-2021-003715. [PMID: 35288464 PMCID: PMC8921915 DOI: 10.1136/jitc-2021-003715] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND T cell receptor (TCR)-engineered cells can be powerful tools in the treatment of malignancies. However, tumor resistance by Human Leukocyte antigen (HLA) class I downregulation can negatively impact the success of any TCR-mediated cell therapy. Allogeneic natural killer (NK) cells have demonstrated efficacy and safety against malignancies without inducing graft-versus-host-disease, highlighting the feasibility for an 'off the shelf' cellular therapeutic. Furthermore, primary NK cells can target tumors using a broad array of intrinsic activation mechanisms. In this study, we combined the antitumor effector functions of NK cells with TCR engineering (NK-TCR), creating a novel therapeutic strategy to avoid TCR-associated immune resistance. METHODS BOB1, is a transcription factor highly expressed in all healthy and malignant B cell lineages, including multiple myeloma (MM). Expression of an HLA-B*07:02 restricted BOB1-specifc TCR in peripheral blood-derived NK cells was achieved following a two-step retroviral transduction protocol. NK-TCR was then compared with TCR-negative NK cells and CD8-T cells expressing the same TCR for effector function against HLA-B*07:02+ B-cell derived lymphoblastoid cell lines (B-LCL), B-cell acute lymphoblastic leukemia and MM cell lines in vitro and in vivo. RESULTS Firstly, TCR could be reproducibly expressed in NK cells isolated from the peripheral blood of multiple healthy donors generating pure NK-TCR cell products. Secondly, NK-TCR demonstrated antigen-specific effector functions against malignancies which were previously resistant to NK-mediated lysis and enhanced NK efficacy in vivo using a preclinical xenograft model of MM. Moreover, antigen-specific cytotoxicity and cytokine production of NK-TCR was comparable to CD8 T cells expressing the same TCR. Finally, in a model of HLA-class I loss, tumor cells with B2M KO were lysed by NK-TCR in an NK-mediated manner but were resistant to T-cell based killing. CONCLUSION NK-TCR cell therapy enhances NK cell efficacy against tumors through additional TCR-mediated lysis. Furthermore, the dual efficacy of NK-TCR permits the specific targeting of tumors and the associated TCR-associated immune resistance, making NK-TCR a unique cellular therapeutic.
Collapse
Affiliation(s)
- Laura T Morton
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Miranda H Meeuwsen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne K Wouters
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis F G Remst
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marleen M van Loenen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
36
|
Johnson CDL, Zale NE, Frary ED, Lomakin JA. Feeder-Cell-Free and Serum-Free Expansion of Natural Killer Cells Using Cloudz Microspheres, G-Rex6M, and Human Platelet Lysate. Front Immunol 2022; 13:803380. [PMID: 35320938 PMCID: PMC8934851 DOI: 10.3389/fimmu.2022.803380] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/26/2022] [Indexed: 11/18/2022] Open
Abstract
The versatility of natural killer cells has ignited growing interest in their therapeutic use for cancer and other immunotherapy treatments. However, NK cells compose a small portion of peripheral blood mononuclear cells (5%–20% of PBMCs) and clinical doses require billions of cells. Manufacturing suitable doses of NK cells remains a major challenge for NK immunotherapy. The current standard for expanding NK cells relies on feeder cells and fetal bovine serum to achieve large expansion, but both encounter regulatory concerns. We developed NK Cloudz, a dissolvable polymer-based microsphere platform, as an alternative to a feeder cell approach to expand NK cells. We demonstrated that a combination of NK Cloudz, a G-Rex6M culture vessel, and GMP Human Platelet Lysate expanded NK cells 387 ± 100-fold in 10 days from a PBMC starting population. The NK purity, viability, and cytotoxicity were similar to both a feeder cell protocol and an FBS-based protocol. Additionally, we found no significant differences between FBS and GMP Human Platelet Lysate and concluded that platelet lysate is a good xeno-free alternative to FBS for NK expansion. Overall, we demonstrated a feeder-cell-free and FBS-free protocol that leverages NK Cloudz as a promising step toward a commercial GMP manufacturing method to expand NK cells for therapeutic use.
Collapse
|
37
|
Colomar-Carando N, Gauthier L, Merli P, Loiacono F, Canevali P, Falco M, Galaverna F, Rossi B, Bosco F, Caratini M, Mingari MC, Locatelli F, Vivier E, Meazza R, Pende D. Exploiting Natural Killer Cell Engagers to Control Pediatric B-cell Precursor Acute Lymphoblastic Leukemia. Cancer Immunol Res 2022; 10:291-302. [PMID: 35078821 PMCID: PMC9662914 DOI: 10.1158/2326-6066.cir-21-0843] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/10/2021] [Accepted: 01/24/2022] [Indexed: 01/07/2023]
Abstract
Natural killer (NK) cells represent a promising cell type in antitumor immunotherapy for efficacy and safety, particularly in the treatment of hematologic malignancies. NK cells have been shown to exert antileukemia activity in the context of haploidentical hematopoietic stem cell transplantation (haplo-HSCT). Products have been developed to boost the activation of NK cells only when cross-linked by tumor cells, avoiding any off-target effect. Here, we tested the in vitro effect of different NK-cell engagers (NKCE), which trigger either NKp46 or NKp30 together with CD16A, and target either CD19 or CD20 to induce killing of pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Target cells were NALM-16 and MHH-CALL-4 cell lines and four primary leukemias, while effector cells were resting NK cells derived from healthy donors and pediatric patients with leukemia after αβT/B-depleted haplo-HSCT. The NK cell-resistant MHH-CALL-4 was efficiently killed using all NKCEs. Boosting of NK activity against MHH-CALL-4 was also evident by degranulation and IFNγ production. Because of the lack of CD20 and high expression of CD19 on primary BCP-ALL, we focused on NKCEs targeting CD19. NKp46- and NKp30-based NKCEs displayed similar potency at inducing NK-cell activity, even when challenged with primary BCP-ALL blasts. Their efficacy was shown also using NK cells derived from transplanted patients. NKCE-induced activation against BCP-ALL can override HLA-specific inhibitory interactions, although the strongest response was observed by the alloreactive NK-cell subset. These data support the therapeutic use of NKp46/CD16A/CD19-NKCE to fight refractory/relapsed leukemia in pretransplantation or posttransplantation settings.
Collapse
Affiliation(s)
- Natalia Colomar-Carando
- Laboratory of Immunology, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Experimental Medicine, University of Genoa, Genova, Italy
| | | | - Pietro Merli
- Department of Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Fabrizio Loiacono
- Laboratory of Immunology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paolo Canevali
- Laboratory of Immunology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Michela Falco
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Federica Galaverna
- Department of Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | | | | | | | - Maria Cristina Mingari
- Laboratory of Immunology, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Experimental Medicine, University of Genoa, Genova, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy.,Department of Gynecology/Obstetrics and Pediatrics, Sapienza University, Roma, Italy
| | - Eric Vivier
- Innate Pharma, Marseille, France.,Aix Marseille University, CNRS, INSERM, CIML, Marseille, France.,APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
| | - Raffaella Meazza
- Laboratory of Immunology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Pende
- Laboratory of Immunology, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Corresponding Author: Daniela Pende, Laboratory of Immunology, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genova 16132, Italy. Phone: 39-010-555-8220; E-mail:
| |
Collapse
|
38
|
Gambella M, Carlomagno S, Raiola AM, Giannoni L, Ghiggi C, Setti C, Giordano C, Luchetti S, Serio A, Bo A, Falco M, Della Chiesa M, Angelucci E, Sivori S. CD19-Targeted Immunotherapies for Diffuse Large B-Cell Lymphoma. Front Immunol 2022; 13:837457. [PMID: 35280988 PMCID: PMC8911710 DOI: 10.3389/fimmu.2022.837457] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/07/2022] [Indexed: 12/15/2022] Open
Abstract
Surgical resection, chemotherapy and radiotherapy were, for many years, the only available cancer treatments. Recently, the use of immune checkpoint inhibitors and adoptive cell therapies has emerged as promising alternative. These cancer immunotherapies are aimed to support or harness the patient's immune system to recognize and destroy cancer cells. Preclinical and clinical studies, based on the use of T cells and more recently NK cells genetically modified with chimeric antigen receptors retargeting the adoptive cell therapy towards tumor cells, have already shown remarkable results. In this review, we outline the latest highlights and progress in immunotherapies for the treatment of Diffuse Large B-cell Lymphoma (DLBCL) patients, focusing on CD19-targeted immunotherapies. We also discuss current clinical trials and opportunities of using immunotherapies to treat DLBCL patients.
Collapse
Affiliation(s)
- Massimiliano Gambella
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Simona Carlomagno
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Anna Maria Raiola
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Livia Giannoni
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Ghiggi
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Chiara Giordano
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Silvia Luchetti
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alberto Serio
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alessandra Bo
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michela Falco
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Emanuele Angelucci
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| |
Collapse
|
39
|
Karvouni M, Vidal-Manrique M, Lundqvist A, Alici E. Engineered NK Cells Against Cancer and Their Potential Applications Beyond. Front Immunol 2022; 13:825979. [PMID: 35242135 PMCID: PMC8887605 DOI: 10.3389/fimmu.2022.825979] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/21/2022] Open
Abstract
Cell therapy is an innovative therapeutic concept where viable cells are implanted, infused, or grafted into a patient to treat impaired or malignant tissues. The term was first introduced circa the 19th century and has since resulted in multiple breakthroughs in different fields of medicine, such as neurology, cardiology, and oncology. Lately, cell and gene therapy are merging to provide cell products with additional or enhanced properties. In this context, adoptive transfer of genetically modified cytotoxic lymphocytes has emerged as a novel treatment option for cancer patients. To this day, five cell therapy products have been FDA approved, four of which for CD19-positive malignancies and one for B-cell maturation antigen (BCMA)-positive malignancies. These are personalized immunotherapies where patient T cells are engineered to express chimeric antigen receptors (CARs) with the aim to redirect the cells against tumor-specific antigens. CAR-T cell therapies show impressive objective response rates in clinical trials that, in certain instances, may reach up to 80%. However, the life-threatening side effects associated with T cell toxicity and the manufacturing difficulties of developing personalized therapies hamper their widespread use. Recent literature suggests that Natural Killer (NK) cells, may provide a safer alternative and an 'off-the-shelf' treatment option thanks to their potent antitumor properties and relatively short lifespan. Here, we will discuss the potential of NK cells in CAR-based therapies focusing on the applications of CAR-NK cells in cancer therapy and beyond.
Collapse
Affiliation(s)
- Maria Karvouni
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Marcos Vidal-Manrique
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology‐Pathology, Karolinska Institute, Stockholm, Sweden
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
40
|
Liu M, Huang W, Guo Y, Zhou Y, Zhi C, Chen J, Li J, He J, Lian H, Zhou J, Ye X, Hu Y, Hu H, Liu Z, Huang J, Lin L, Cai M, Wang X, Huang J, Zhang Z, Zhu K, Zhao Q, Cao B. CAR NK-92 cells targeting DLL3 kill effectively small cell lung cancer cells in vitro and in vivo. J Leukoc Biol 2022; 112:901-911. [PMID: 35088475 DOI: 10.1002/jlb.5ma0122-467r] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Small cell lung cancer (SCLC) is characterized by a high relapse rate, drug tolerance, and limited treatment choices. Chimeric antigen receptor (CAR)-modified NK cells represent a promising immunotherapeutic modality for cancer treatment. However, their potential applications have not been explored in SCLC. Delta-like ligand 3 (DLL3) has been reported to be overexpressed in SCLC and may be a rational target for CAR NK immunotherapy. In this study, we developed DLL3-specific NK-92 cells and explored their potential in the treatment of SCLC. A coculture of DLL3+ SCLC cell lines with DLL3-CAR NK-92 cells exhibited significant in vitro cytotoxicity and cytokine production. DLL3-CAR NK-92 cells induced tumor regression in an H446-derived pulmonary metastasis tumor model under a good safety threshold. The potent antitumor activities of DLL3-CAR NK-92 cells were observed in subcutaneous tumor models of SCLC. Moreover, obvious tumor-infiltrated DLL3-CAR NK-92 cells were detected in DLL3+ SCLC xenografts. These findings indicate that DLL3-CAR NK-92 cells might be a potential strategy for the treatment of SCLC.
Collapse
Affiliation(s)
- Manting Liu
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wensou Huang
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yongjian Guo
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yubo Zhou
- Department of Library, Guangzhou Medical University, Guangzhou, China
| | - Cheng Zhi
- Department of Pathology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingwu Chen
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junping Li
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinping He
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hui Lian
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingwen Zhou
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaodie Ye
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuling Hu
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hong Hu
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaoyuan Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingjun Huang
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Liteng Lin
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingyue Cai
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaobin Wang
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingzhen Huang
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhenfeng Zhang
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Bihui Cao
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
41
|
Tang J, Zhu Q, Li Z, Yang J, Lai Y. Natural killer cell-targeted immunotherapy for cancer. Curr Stem Cell Res Ther 2022; 17:513-526. [PMID: 34994316 DOI: 10.2174/1574888x17666220107101722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/12/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Natural killer (NK) cells were initially described in the early 1970s as major histocompatibility complex unrestricted killers due to their ability to spontaneously kill certain tumor cells. In the past decade, the field of NK cell-based treatment has been accelerating exponentially, holding a dominant position in cancer immunotherapy innovation. Generally, research on NK cell-mediated antitumor therapies can be categorized into three areas: choosing the optimal source of allogenic NK cells to yield massively amplified "off-the-shelf" products, improving NK cell cytotoxicity and longevity, and engineering NK cells with the ability of tumor-specific recognition. In this review, we focused on NK cell manufacturing techniques, some auxiliary methods to enhance the therapeutic efficacy of NK cells, chimeric antigen receptor NK cells, and monoclonal antibodies targeting inhibitory receptors, which can significantly augment the antitumor activity of NK cells. Notably, emerging evidence suggests that NK cells are a promising constituent of multipronged therapeutic strategies, strengthening immune responses to cancer.
Collapse
Affiliation(s)
- Jingyi Tang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qi Zhu
- Sichuan Fine Arts Institute, Chongqing, China
| | - Zhaoyang Li
- Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Jiahui Yang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yu Lai
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Shokouhifar A, Firouzi J, Nouri M, Sarab GA, Ebrahimi M. NK cell upraise in the dark world of cancer stem cells. Cancer Cell Int 2021; 21:682. [PMID: 34923966 PMCID: PMC8684645 DOI: 10.1186/s12935-021-02400-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/08/2021] [Indexed: 12/29/2022] Open
Abstract
One of the obstacles in treating different cancers, especially solid tumors, is cancer stem cells (CSCs) with their ability in resistance to chemo/radio therapy. The efforts for finding advanced treatments to overcome these cells have led to the emergence of advanced immune cell-based therapy (AICBT). Today, NK cells have become the center of attention since they have been proved to show an appropriate cytotoxicity against different cancer types as well as the capability of detecting and killing CSCs. Attempts for reaching an off-the-shelf source of NK cells have been made and resulted in the emergence of chimeric antigen receptor natural killer cells (CAR-NK cells). The CAR technology has then been used for generating more cytotoxic and efficient NK cells, which has increased the hope for cancer treatment. Since utilizing this advanced technology to target CSCs have been published in few studies, the present study has focused on discussing the characteristics of CSCs, which are detected and targeted by NK cells, the advantages and restrictions of using CAR-NK cells in CSCs treatment and the probable challenges in this process.
Collapse
Affiliation(s)
- Alireza Shokouhifar
- Department of Molecular Medicine, Genomic Research Center, Birjand University of Medical Sciences, Birjand, Iran.,Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, 16635-148, Tehran, Iran
| | - Javad Firouzi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, 16635-148, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Nouri
- R&D Department, Royan Stem Cell Technology Co., Tehran, Iran
| | - Gholamreza Anani Sarab
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, 16635-148, Tehran, Iran. .,Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, 14155-4364, Tehran, Iran.
| |
Collapse
|
43
|
Guercio M, Manni S, Boffa I, Caruso S, Di Cecca S, Sinibaldi M, Abbaszadeh Z, Camera A, Ciccone R, Polito VA, Ferrandino F, Reddel S, Catanoso ML, Bocceri E, Del Bufalo F, Algeri M, De Angelis B, Quintarelli C, Locatelli F. Inclusion of the Inducible Caspase 9 Suicide Gene in CAR Construct Increases Safety of CAR.CD19 T Cell Therapy in B-Cell Malignancies. Front Immunol 2021; 12:755639. [PMID: 34737753 PMCID: PMC8560965 DOI: 10.3389/fimmu.2021.755639] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/27/2021] [Indexed: 11/14/2022] Open
Abstract
T cells engineered with chimeric antigen receptor (CAR-T cells) are an effective treatment in patients with relapsed/refractory B-cell precursor acute lymphoblastic leukemia or B-cell non-Hodgkin lymphoma. Despite the reported exciting clinical results, the CAR-T cell approach needs efforts to improve the safety profile, limiting the occurrence of adverse events in patients given this treatment. Besides the most common side effects, such as cytokine release syndrome and CAR-T cell–related encephalopathy syndrome, another potential issue involves the inadvertent transduction of leukemia B cells with the CAR construct during the manufacturing process, thus leading to the possibility of a peculiar mechanism of antigen masking and treatment resistance. In this study, we investigated whether the inclusion of the inducible caspase 9 (iC9) suicide gene in the CAR construct design could be an effective safety switch to control malignant CAR+ B cells, ultimately counteracting this serious adverse event. iC9 is a suicide gene able to be activated through binding with an otherwise inert small biomolecule, known as AP1903. The exposure of iC9.CAR.CD19-DAUDI lymphoma and iC9.CAR.CD19-NALM-6 leukemia cells in vitro to 20 nM of AP1903 resulted into the prompt elimination of CAR+ B-leukemia/lymphoma cell lines. The results obtained in the animal model corroborate in vitro data, since iC9.CAR.CD19+ tumor cells were controlled in vivo by the activation of the suicide gene through administration of AP1903. Altogether, our data indicate that the inclusion of the iC9 suicide gene may result in a safe CAR-T cell product, even when manufacturing starts from biological materials characterized by heavy leukemia blast contamination.
Collapse
Affiliation(s)
- Marika Guercio
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Simona Manni
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Iolanda Boffa
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Simona Caruso
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Stefano Di Cecca
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Matilde Sinibaldi
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Zeinab Abbaszadeh
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonio Camera
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Roselia Ciccone
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Vinicia Assunta Polito
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Francesca Ferrandino
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Sofia Reddel
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Maria Luigia Catanoso
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Emilia Bocceri
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Francesca Del Bufalo
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Mattia Algeri
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Biagio De Angelis
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Concetta Quintarelli
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.,Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Franco Locatelli
- Department of Oncology-Haematology and Cell and Gene Therapy, Bambino Gesù Children Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.,Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
44
|
Lu S, Feng Q. CAR-NK cells from engineered pluripotent stem cells: Off-the-shelf therapeutics for all patients. Stem Cells Transl Med 2021; 10 Suppl 2:S10-S17. [PMID: 34724715 PMCID: PMC8560199 DOI: 10.1002/sctm.21-0135] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/04/2021] [Accepted: 05/16/2021] [Indexed: 12/12/2022] Open
Abstract
Clinical success of adoptive cell therapy with chimeric antigen receptor (CAR) T cells for treating hematological malignancies has revolutionized the field of cellular immunotherapy. However, due to the nature of utilizing autologous T cells, affordability and availability are major hurdles, in addition to scientific challenges relating to CAR-T therapy optimization. Natural killer (NK) cell is a specialized immune effector cell type that recognizes and kills targets without human leukocyte antigen (HLA) restriction and prior sensitization. CAR-NK cells do not cause graft vs host disease and can be obtained from unrelated donors as well as pluripotent stem cells (PSC), representing an ideal off-the-shelf therapeutics readily available for patients. Furthermore, unlike cytotoxic T cells, NK cells specifically target and eliminate cancer stem cells, which are the cells causing relapse and metastasis. PSCs can be genetically manipulated and engineered with CARs at the pluripotent stage, which allows the establishment of permanent, stable, and clonal PSC-CAR lines for the manufacture of unlimited homogenous CAR-NK cells. Multiple master PSC-CAR cell banks targeting a variety of antigens for cancer, viral infection, and autoimmune diseases provide inexhaustible cell sources for all patients. Development of a next-generation 3D bioreactor platform for PSC expansion and NK cell production overcomes major barriers related to cost and scalability for CAR-NK product.
Collapse
Affiliation(s)
| | - Qiang Feng
- HebeCell CorporationNatickMassachusettsUSA
| |
Collapse
|
45
|
The Race of CAR Therapies: CAR-NK Cells for Fighting B-Cell Hematological Cancers. Cancers (Basel) 2021; 13:cancers13215418. [PMID: 34771581 PMCID: PMC8582420 DOI: 10.3390/cancers13215418] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Over the last few years, CAR-T cells have arisen as one of the most promising immunotherapies against relapsed or refractory hematological cancers. Despite their good results in clinical trials, there are some limitations to overcome, such as undesirable side-effects or the restraints of an autologous treatment. Therefore, CAR-NK cells have emerged as a good alternative for these kinds of treatments. This review discusses the advantages of CAR-NK cells compared to CAR-T cells, as well as the different sources and strategies in order to obtain these CAR-NK cells. Abstract Acute lymphoblastic leukemia (ALL) and Chronic lymphocytic leukemia (CLL) are the most common leukemias in children and elderly people, respectively. Standard therapies, such as chemotherapy, are only effective in 40% of ALL adult patients with a five-year survival rate and therefore new alternatives need to be used, such as immunotherapy targeting specific receptors of malignant cells. Among all the options, CAR (Chimeric antigen receptor)-based therapy has arisen as a new opportunity for refractory or relapsed hematological cancer patients. CARs were designed to be used along with T lymphocytes, creating CAR-T cells, but they are presenting such encouraging results that they are already in use as drugs. Nonetheless, their side-effects and the fact that it is not possible to infuse an allogenic CAR-T product without causing graft-versus-host-disease, have meant using a different cell source to solve these problems, such as Natural Killer (NK) cells. Although CAR-based treatment is a high-speed race led by CAR-T cells, CAR-NK cells are slowly (but surely) consolidating their position; their demonstrated efficacy and the lack of undesirable side-effects is opening a new door for CAR-based treatments. CAR-NKs are now in the field to stay.
Collapse
|
46
|
Hosseini M, Habibi Z, Hosseini N, Abdoli S, Rezaei N. Preclinical studies of chimeric antigen receptor-modified natural killer cells in cancer immunotherapy: a review. Expert Opin Biol Ther 2021; 22:349-366. [PMID: 34541989 DOI: 10.1080/14712598.2021.1983539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION As one of the most efficacious methods of cancer immunotherapy, chimeric antigen receptor-modified immune cells have recently drawn enormous attention. After the great success achieved with CAR-T-cells in cancer treatment both in preclinical setting and in the clinic, other types of immune cells, including natural killer (NK)-cells and macrophages, have been evaluated for their anti-cancer effects along with their potential superiority against CAR-T-cells, especially in terms of safety. First introduced by Tran et al. almost 26 years ago, CAR-NK-cells are now being considered as efficient immunotherapeutic modalities in various types of cancers, not only in preclinical setting but also in numerous phase I and II clinical studies. AREAS COVERED In this review, we aim to provide a comprehensive survey of the preclinical studies on CAR-NK-cells' development, with an evolutional approach on CAR structures and their associated signaling moieties. Current NK-cell sources and modes of gene transfer are also reviewed. EXPERT OPINION CAR-NK-cells have appeared as safe and effective immunotherapeutic tools in preclinical settings; however, designing CAR structures with an eye on their specific biology, along with choosing the optimal cell source and gene transfer method require further investigation to support clinical studies.
Collapse
Affiliation(s)
- Mina Hosseini
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Habibi
- School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Hosseini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sina Abdoli
- School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Chung S, Revia RA, Zhang M. Iron oxide nanoparticles for immune cell labeling and cancer immunotherapy. NANOSCALE HORIZONS 2021; 6:696-717. [PMID: 34286791 PMCID: PMC8496976 DOI: 10.1039/d1nh00179e] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Cancer immunotherapy is a novel approach to cancer treatment that leverages components of the immune system as opposed to chemotherapeutics or radiation. Cell migration is an integral process in a therapeutic immune response, and the ability to track and image the migration of immune cells in vivo allows for better characterization of the disease and monitoring of the therapeutic outcomes. Iron oxide nanoparticles (IONPs) are promising candidates for use in immunotherapy as they are biocompatible, have flexible surface chemistry, and display magnetic properties that may be used in contrast-enhanced magnetic resonance imaging (MRI). In this review, advances in application of IONPs in cell tracking and cancer immunotherapy are presented. Following a brief overview of the cancer immunity cycle, developments in labeling and tracking various immune cells using IONPs are highlighted. We also discuss factors that influence the effectiveness of IONPs as MRI contrast agents. Finally, we outline different approaches for cancer immunotherapy and highlight current efforts that utilize IONPs to stimulate immune cells to enhance their activity and response to cancer.
Collapse
Affiliation(s)
- Seokhwan Chung
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, USA.
| | | | | |
Collapse
|
48
|
Salvaris R, Fedele PL. Targeted Therapy in Acute Lymphoblastic Leukaemia. J Pers Med 2021; 11:715. [PMID: 34442359 PMCID: PMC8398498 DOI: 10.3390/jpm11080715] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 07/21/2021] [Indexed: 11/26/2022] Open
Abstract
The last decade has seen a significant leap in our understanding of the wide range of genetic lesions underpinning acute lymphoblastic leukaemia (ALL). Next generation sequencing has led to the identification of driver mutations with significant implications on prognosis and has defined entities such as BCR-ABL-like ALL, where targeted therapies such as tyrosine kinase inhibitors (TKIs) and JAK inhibitors may play a role in its treatment. In Philadelphia positive ALL, the introduction of TKIs into frontline treatment regimens has already transformed patient outcomes. In B-ALL, agents targeting surface receptors CD19, CD20 and CD22, including monoclonal antibodies, bispecific T cell engagers, antibody drug conjugates and chimeric antigen receptor (CAR) T cells, have shown significant activity but come with unique toxicities and have implications for how treatment is sequenced. Advances in T-ALL have lagged behind those seen in B-ALL. However, agents such as nelarabine, bortezomib and CAR T cell therapy targeting T cell antigens have been examined with promising results seen. As our understanding of disease biology in ALL grows, as does our ability to target pathways such as apoptosis, through BH3 mimetics, chemokines and epigenetic regulators. This review aims to highlight a range of available and emerging targeted therapeutics in ALL, to explore their mechanisms of action and to discuss the current evidence for their use.
Collapse
Affiliation(s)
- Ross Salvaris
- Department of Clinical Haematology, Monash Health, Clayton 3168, Australia;
- School of Clinical Sciences at Monash Health, Monash University, Clayton 3168, Australia
| | - Pasquale Luke Fedele
- Department of Clinical Haematology, Monash Health, Clayton 3168, Australia;
- School of Clinical Sciences at Monash Health, Monash University, Clayton 3168, Australia
| |
Collapse
|
49
|
Viardot A, Sala E. Investigational immunotherapy targeting CD19 for the treatment of acute lymphoblastic leukemia. Expert Opin Investig Drugs 2021; 30:773-784. [PMID: 33998346 DOI: 10.1080/13543784.2021.1928074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The anti-CD19 immunotherapy for the treatment of B-precursor acute lymphoblastic leukemia (B-ALL) underwent an expansion in the last decade. CD19 is widely expressed on B-ALL and nearly ideal for immunotherapy because of strong 'on target' ─ but manageable 'off target' effects. AREAS COVERED We review the major advances in the field, including data on CD19 monoclonal antibodies, antibody-drug conjugates, bispecific T-cell engaging antibodies and adoptive cellular therapies such as chimeric antigen receptor T cells (CAR-Ts). We discuss novel strategies on approved anti-CD19 immunotherapies. The focus is on experimental anti-CD19 antibodies or CAR-Ts, which might overcome the limitations of toxicity, rapid clearance or resistance. EXPERT OPINION The potential of new anti-CD19 antibodies in ALL is limited. The most promising results were achieved with novel cellular constructs. Bi- or multi-specific CAR-Ts might overcome the immune escape by antigen loss. Modified constructs with lower peak expansion or longer persistence provide better control of the toxicity and might improve the efficacy. Finally, the allogeneic 'off the shelf' constructs from healthy donors avoid the time-consuming preparation and the exhaustion of immune cells.
Collapse
Affiliation(s)
- Andreas Viardot
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Elisa Sala
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
50
|
Marofi F, Saleh MM, Rahman HS, Suksatan W, Al-Gazally ME, Abdelbasset WK, Thangavelu L, Yumashev AV, Hassanzadeh A, Yazdanifar M, Motavalli R, Pathak Y, Naimi A, Baradaran B, Nikoo M, Khiavi FM. CAR-engineered NK cells; a promising therapeutic option for treatment of hematological malignancies. Stem Cell Res Ther 2021; 12:374. [PMID: 34215336 PMCID: PMC8252313 DOI: 10.1186/s13287-021-02462-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
Adoptive cell therapy has received a great deal of interest in the treatment of advanced cancers that are resistant to traditional therapy. The tremendous success of chimeric antigen receptor (CAR)-engineered T (CAR-T) cells in the treatment of cancer, especially hematological cancers, has exposed CAR's potential. However, the toxicity and significant limitations of CAR-T cell immunotherapy prompted research into other immune cells as potential candidates for CAR engineering. NK cells are a major component of the innate immune system, especially for tumor immunosurveillance. They have a higher propensity for immunotherapy in hematologic malignancies because they can detect and eliminate cancerous cells more effectively. In comparison to CAR-T cells, CAR-NK cells can be prepared from allogeneic donors and are safer with a lower chance of cytokine release syndrome and graft-versus-host disease, as well as being a more efficient antitumor activity with high efficiency for off-the-shelf production. Moreover, CAR-NK cells may be modified to target various antigens while also increasing their expansion and survival in vivo. Extensive preclinical research has shown that NK cells can be effectively engineered to express CARs with substantial cytotoxic activity against both hematological and solid tumors, establishing evidence for potential clinical trials of CAR-NK cells. In this review, we discuss recent advances in CAR-NK cell engineering in a variety of hematological malignancies, as well as the main challenges that influence the outcomes of CAR-NK cell-based tumor immunotherapies.
Collapse
Affiliation(s)
- Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marwan Mahmood Saleh
- Department of Biophysics, College of Applied Science, University of Anbar, Ramadi, Iraq
| | - Heshu Sulaiman Rahman
- College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Chaq-Chaq Qularaise, Sulaimaniyah, Iraq
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | | | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA USA
| | - Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yashwant Pathak
- Professor and Associate Dean for Faculty Affairs, Taneja College of Pharmacy, University of South Florida, Tampa, FL USA
- Faculty of Pharmacy, Airlangga University, Surabaya, Indonesia
| | - Adel Naimi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Behzad Baradaran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|