1
|
Jewett A, Memarzadeh S, Kaur K. Commentary: Ovarian Cancer: Path to Effective Treatments. Crit Rev Immunol 2025; 45:93-99. [PMID: 39612280 DOI: 10.1615/critrevimmunol.2024053766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Despite advancements in cancer therapeutics such as checkpoint inhibitors and some targeted therapies, we have not achieved success in effectively treating ovarian cancer, since these therapeutics only benefit a subset of patients, and also provide short-term protection. The use of chemotherapy and radiation therapy can cause depletion and/or lack of immune cells' function. Chimeric antigen receptor T (CAR-T) cell therapy is found to be effective against several blood-based cancers, but limited success was seen against solid tumors. Targeting fewer antigens and significant side effects of therapy decreases the efficacy of CAR-T cells as immunotherapeutic in solid tumors, even though there is a great drive and significant effort to establish these therapies around the world. Bispecific and tri-specific antibodies have recently been advocated as effective cancer therapeutics. However, at present, these also suffer the fate of CAR-Ts since the loss of antigen on tumor cells will render these therapeutics ineffective. At present, we should design therapeutics that may have synergistic effects on killing/treating tumors. The only way we can establish that will be by learning the mechanisms of actions of immune therapeutics. Thus, advancement in the knowledge and effective strategies are required to develop cancer immuno-therapeutics. We have dedicated our efforts to understand the immunobiology of natural killer (NK) cells. One of our most important discoveries was demonstration of targeting of cancer stem-like cells (CSCs)/poorly differentiated tumors exhibiting lower major histocompability complex class I expression by the NK cells. In addition, we showed that supercharged NK (sNK) cells had great ability to target both CSCs/poorly differentiated and well differentiated ovarian tumors, whereas activated primary NK cells only targeted CSCs/poorly differentiated tumors. Therefore, the use of sNK cells in immunotherapy should result in effective elimination of heterogeneous populations of ovarian tumors.
Collapse
Affiliation(s)
- Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA; The Jonsson Comprehensive Cancer Center, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA
| | - Sanaz Memarzadeh
- The Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA; UCLA Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; The VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| |
Collapse
|
2
|
Kalim M, Jing R, Guo W, Xing H, Lu Y. Functional diversity and regulation of IL-9-producing T cells in cancer immunotherapy. Cancer Lett 2024; 606:217306. [PMID: 39426662 DOI: 10.1016/j.canlet.2024.217306] [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: 06/18/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
IL-9-producing T cells (T9) regulate immunological responses that affect various cellular biological processes, though their precise function remains fully understood. Previous studies have linked T9 cells to conditions such as allergic disorders, parasitic infection clearance, and various types of cancers. While the functional heterogeneity of IL-9 and T9 cells in cancer development has been documented, these cells present promising therapeutic opportunities for treating solid tumors. This review highlights the roles of IL-9 and T9 cells in cancer progression and treatment responses, focusing on potential discrepancies in IL-9/IL-9R signaling between murine tumors and cancer patients. Additionally, we discuss the regulation of tumor-specific Th9/Tc9 cell differentiation, the therapeutic potential of these cells, and current strategies to enhance their anti-tumor activities.
Collapse
Affiliation(s)
- Muhammad Kalim
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Rui Jing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Wei Guo
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Hui Xing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Yong Lu
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Kane G, Lusi C, Brassil M, Atukorale P. Engineering approaches for innate immune-mediated tumor microenvironment remodeling. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 21:100406. [PMID: 38213392 PMCID: PMC10777078 DOI: 10.1016/j.iotech.2023.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Cancer immunotherapy offers transformative promise particularly for the treatment of lethal cancers, since a correctly trained immune system can comprehensively orchestrate tumor clearance with no need for continued therapeutic intervention. Historically, the majority of immunotherapies have been T cell-focused and have included immune checkpoint inhibitors, chimeric antigen receptor T cells, and T-cell vaccines. Unfortunately T-cell-focused therapies have failed to achieve optimal efficacy in most solid tumors largely because of a highly immunosuppressed 'cold' or immune-excluded tumor microenvironment (TME). Recently, a rapidly growing treatment paradigm has emerged that focuses on activation of tumor-resident innate antigen-presenting cells, such as dendritic cells and macrophages, which can drive a proinflammatory immune response to remodel the TME from 'cold' or immune-excluded to 'hot'. Early strategies for TME remodeling centered on free cytokines and agonists, but these approaches have faced significant hurdles in both delivery and efficacy. Systemic toxicity from off-target inflammation is a paramount concern in these therapies. To address this critical gap, engineering approaches have provided the opportunity to add 'built-in' capabilities to cytokines, agonists, and other therapeutic agents to mediate improved delivery and efficacy. Such capabilities have included protective encapsulation to shield them from degradation, targeting to direct them with high specificity to tumors, and co-delivery strategies to harness synergistic proinflammatory pathways. Here, we review innate immune-mediated TME remodeling engineering approaches that focus on cytokines, innate immune agonists, immunogenic viruses, and cell-based methods, highlighting emerging preclinical approaches and strategies that are either being tested in clinical trials or already Food and Drug Administration approved.
Collapse
Affiliation(s)
- G.I. Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - C.F. Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - M.L. Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - P.U. Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, USA
| |
Collapse
|
4
|
Dhanyamraju PK. Drug resistance mechanisms in cancers: Execution of pro-survival strategies. J Biomed Res 2024; 38:95-121. [PMID: 38413011 PMCID: PMC11001593 DOI: 10.7555/jbr.37.20230248] [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: 10/13/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 02/29/2024] Open
Abstract
One of the quintessential challenges in cancer treatment is drug resistance. Several mechanisms of drug resistance have been described to date, and new modes of drug resistance continue to be discovered. The phenomenon of cancer drug resistance is now widespread, with approximately 90% of cancer-related deaths associated with drug resistance. Despite significant advances in the drug discovery process, the emergence of innate and acquired mechanisms of drug resistance has impeded the progress in cancer therapy. Therefore, understanding the mechanisms of drug resistance and the various pathways involved is integral to treatment modalities. In the present review, I discuss the different mechanisms of drug resistance in cancer cells, including DNA damage repair, epithelial to mesenchymal transition, inhibition of cell death, alteration of drug targets, inactivation of drugs, deregulation of cellular energetics, immune evasion, tumor-promoting inflammation, genome instability, and other contributing epigenetic factors. Furthermore, I highlight available treatment options and conclude with future directions.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
5
|
Ramezani F, Panahi Meymandi AR, Akbari B, Tamtaji OR, Mirzaei H, Brown CE, Mirzaei HR. Outsmarting trogocytosis to boost CAR NK/T cell therapy. Mol Cancer 2023; 22:183. [PMID: 37974170 PMCID: PMC10652537 DOI: 10.1186/s12943-023-01894-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023] Open
Abstract
Chimeric antigen receptor (CAR) NK and T cell therapy are promising immunotherapeutic approaches for the treatment of cancer. However, the efficacy of CAR NK/T cell therapy is often hindered by various factors, including the phenomenon of trogocytosis, which involves the bidirectional exchange of membrane fragments between cells. In this review, we explore the role of trogocytosis in CAR NK/T cell therapy and highlight potential strategies for its modulation to improve therapeutic efficacy. We provide an in-depth analysis of trogocytosis as it relates to the fate and function of NK and T cells, focusing on its effects on cell activation, cytotoxicity, and antigen presentation. We discuss how trogocytosis can mediate transient antigen loss on cancer cells, thereby negatively affecting the effector function of CAR NK/T cells. Additionally, we address the phenomenon of fratricide and trogocytosis-associated exhaustion, which can limit the persistence and effectiveness of CAR-expressing cells. Furthermore, we explore how trogocytosis can impact CAR NK/T cell functionality, including the acquisition of target molecules and the modulation of signaling pathways. To overcome the negative effects of trogocytosis on cellular immunotherapy, we propose innovative approaches to modulate trogocytosis and augment CAR NK/T cell therapy. These strategies encompass targeting trogocytosis-related molecules, engineering CAR NK/T cells to resist trogocytosis-induced exhaustion and leveraging trogocytosis to enhance the function of CAR-expressing cells. By overcoming the limitations imposed by trogocytosis, it may be possible to unleash the full potential of CAR NK/T therapy against cancer. The knowledge and strategies presented in this review will guide future research and development, leading to improved therapeutic outcomes in the field of immunotherapy.
Collapse
Affiliation(s)
- Faezeh Ramezani
- Division of Medical Biotechnology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Reza Panahi Meymandi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnia Akbari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Reza Tamtaji
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Christine E Brown
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, USA
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
6
|
Narbona J, Hernández-Baraza L, Gordo RG, Sanz L, Lacadena J. Nanobody-Based EGFR-Targeting Immunotoxins for Colorectal Cancer Treatment. Biomolecules 2023; 13:1042. [PMID: 37509078 PMCID: PMC10377705 DOI: 10.3390/biom13071042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/22/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Immunotoxins (ITXs) are chimeric molecules that combine the specificity of a targeting domain, usually derived from an antibody, and the cytotoxic potency of a toxin, leading to the selective death of tumor cells. However, several issues must be addressed and optimized in order to use ITXs as therapeutic tools, such as the selection of a suitable tumor-associated antigen (TAA), high tumor penetration and retention, low kidney elimination, or low immunogenicity of foreign proteins. To this end, we produced and characterized several ITX designs, using a nanobody against EGFR (VHH 7D12) as the targeting domain. First, we generated a nanoITX, combining VHH 7D12 and the fungal ribotoxin α-sarcin (αS) as the toxic moiety (VHHEGFRαS). Then, we incorporated a trimerization domain (TIEXVIII) into the construct, obtaining a trimeric nanoITX (TriVHHEGFRαS). Finally, we designed and characterized a bispecific ITX, combining the VHH 7D12 and the scFv against GPA33 as targeting domains, and a deimmunized (DI) variant of α-sarcin (BsITXαSDI). The results confirm the therapeutic potential of α-sarcin-based nanoITXs. The incorporation of nanobodies as target domains improves their therapeutic use due to their lower molecular size and binding features. The enhanced avidity and toxic load in the trimeric nanoITX and the combination of two different target domains in the bispecific nanoITX allow for increased antitumor effectiveness.
Collapse
Affiliation(s)
- Javier Narbona
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University, 28040 Madrid, Spain
| | - Luisa Hernández-Baraza
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University, 28040 Madrid, Spain
- University Institute of Biomedical and Health Research (IUIBS), Las Palmas University, 35016 Las Palmas de Gran Canaria, Spain
| | - Rubén G Gordo
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University, 28040 Madrid, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute, Hospital Universitario Puerta de Hierro, Majadahonda, 28222 Madrid, Spain
| | - Javier Lacadena
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
7
|
Goodlad JR, Xiao W, Amador C, Cook JR, Happ L, Thakkar D, Dave S, Dogan A, Duffield A, Nejati R, Ott G, Wasik M, Czader M. Phenotypic and genotypic infidelity in B-lineage neoplasms, including transdifferentiation following targeted therapy: Report from the 2021 SH/EAHP Workshop. Am J Clin Pathol 2023:7135991. [PMID: 37085149 DOI: 10.1093/ajcp/aqad035] [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: 12/05/2022] [Accepted: 03/13/2023] [Indexed: 04/23/2023] Open
Abstract
OBJECTIVES Session 2 of the 2021 Society for Hematopathology and European Association for Haematopathology Workshop collected examples of lineage infidelity and transdifferentiation in B-lineage neoplasms, including after targeted therapy. METHODS Twenty cases were submitted. Whole-exome sequencing and genome-wide RNA expression analysis were available on a limited subsample. RESULTS A diagnosis of B-cell acute lymphoblastic leukemia (B-ALL) was rendered on at least 1 biopsy from 13 patients. There was 1 case of acute myeloid leukemia (AML); the remaining 6 cases were mature B-cell neoplasms. Targeted therapy was administered in 7 cases of B-ALL and 4 cases of mature B-cell neoplasms. Six cases of B-ALL underwent lineage switch to AML or mixed-phenotype acute leukemia at relapse, 5 of which had rearranged KMT2A. Changes in maturational state without lineage switch were observed in 2 cases. Examples of de novo aberrant T-cell antigen expression (n = 2) were seen among the mature B-cell lymphoma cohort, and their presence correlated with alterations in tumor cell gene expression patterns. CONCLUSIONS This cohort of cases enabled us to illustrate, discuss, and review current concepts of lineage switch and aberrant antigen expression in a variety of B-cell neoplasms and draw attention to the role targeted therapies may have in predisposing neoplasms to transdifferentiation as well as other, less expected changes in maturational status.
Collapse
Affiliation(s)
- John R Goodlad
- Department of Pathology, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Wenbin Xiao
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, US
| | - Catalina Amador
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, US
| | - James R Cook
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, OH, US
| | | | | | - Sandeep Dave
- Center for Genomic and Computational Biology and Department of Medicine, Duke University School of Medicine, Durham, NC, US
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, US
| | - Amy Duffield
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, US
| | - Reza Nejati
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, US
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Mariusz Wasik
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, US
| | - Magdalena Czader
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, US
| |
Collapse
|
8
|
Sulzmaier FJ, Kern N, Ahn SJ, Polovina A, Ho J, Hussain A, Cyprus G, Macedo C, Pandit R, Crago W, Rowell E, Timmer JC, Eckelman BP. INBRX-120, a CD8α-targeted detuned IL-2 that selectively expands and activates tumoricidal effector cells for safe and durable in vivo responses. J Immunother Cancer 2023; 11:jitc-2022-006116. [PMID: 36653071 PMCID: PMC9853254 DOI: 10.1136/jitc-2022-006116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND As a major driver of lymphocyte proliferation and activation interleukin 2 (IL-2) is a crucial mediator for antitumor responses. Despite promising activity in a subset of patients, wider therapeutic utility of IL-2 (aldesleukin) has been hampered by severe dose-limiting toxicities, the expansion of immunosuppressive regulatory T cells and a poor pharmacokinetic (PK) profile. Recent engineering efforts, including non-α IL-2 variants, have lowered the toxicity profile, but have yet to induce meaningful antitumor activity in a wider patient population. METHODS We engineered INBRX-120, a CD8α-targeted Cisleukin™ molecule consisting of an affinity tuned IL-2 (IL2-x) connected to two high affinity CD8α-specific single domain antibodies via an effector-silenced Fc domain. To show that this large affinity differential enables directed IL-2 cis-signaling exclusively on CD8α-expressing tumoricidal effector cell populations, INBRX-120 effects on target cell expansion, activation and antitumor activity were tested in vitro. In vivo antitumor efficacy was evaluated in syngeneic mouse models alone or in combination with programmed cell death protein-1 (PD-1) blockade. Preclinical safety, as well as pharmacodynamic (PD) and PK profiling was carried out in non-human primates. RESULTS INBRX-120 effectively expanded and enhanced the cytotoxic capacity of CD8 T cells and natural killer cells towards tumor cells without affecting regulatory T cells in vitro and in vivo. In syngeneic mouse models, INBRX-120 surrogate showed safe, potent, and durable antitumor efficacy alone and in combination with PD-1 blockade. In non-human primates, INBRX-120 expanded and activated CD8α-expressing effector cells, showed a favorable PK profile, and was well tolerated up to a dose of 1 mg/kg. CONCLUSIONS Through its unique cis-signaling activity on CD8α-expressing effector cells, INBRX-120 overcomes the major limitations of IL-2-based therapy and effectively harnesses IL-2's potent intrinsic antitumor activity. This novel therapeutic strategy promises safer clinical activity that could induce meaningful antitumor efficacy in a wider set of patients with various cancer indications.
Collapse
Affiliation(s)
| | | | | | | | - Jason Ho
- Inhibrx Inc, La Jolla, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Wang ZH, Li W, Dong H, Han F. Current state of NK cell-mediated immunotherapy in chronic lymphocytic leukemia. Front Oncol 2023; 12:1077436. [PMID: 37078002 PMCID: PMC10107371 DOI: 10.3389/fonc.2022.1077436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) has become one of the most common hematological diseases in western countries, with an annual incidence of 42/100,000. Conventional chemotherapy and targeted therapeutic drugs showed limitations in prognosis or in efficiency in high-risk patients. Immunotherapy represented is one of the most effective therapeutic approaches with the potential of better effect and prognosis. Natural killer (NK) cells are good options for immunotherapy as they can effectively mediate anti-tumor activity of immune system by expressing activating and inhibiting receptors and recognizing specific ligands on various tumor cells. NK cells are critical in the immunotherapy of CLL by enhancing self-mediated antibody-dependent cytotoxicity (ADCC), allogeneic NK cell therapy and chimeric antigen receptor-natural killer (CAR-NK) cell therapy. In this article, we reviewed the features, working mechanisms, and receptors of NK cells, and the available evidence of the advantages and disadvantages of NK cell-based immunotherapies, and put forward future study directions in this field.
Collapse
Affiliation(s)
- Zong-Han Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Li
- Department of General Surgery, Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Hao Dong
- Department of Gastrointestinal Nutrition and Surgical Surgery, The Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Hao Dong, ; Fujun Han,
| | - Fujun Han
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Hao Dong, ; Fujun Han,
| |
Collapse
|
10
|
Faisal SM, Comba A, Varela ML, Argento AE, Brumley E, Abel C, Castro MG, Lowenstein PR. The complex interactions between the cellular and non-cellular components of the brain tumor microenvironmental landscape and their therapeutic implications. Front Oncol 2022; 12:1005069. [PMID: 36276147 PMCID: PMC9583158 DOI: 10.3389/fonc.2022.1005069] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
Glioblastoma (GBM), an aggressive high-grade glial tumor, is resistant to therapy and has a poor prognosis due to its universal recurrence rate. GBM cells interact with the non-cellular components in the tumor microenvironment (TME), facilitating their rapid growth, evolution, and invasion into the normal brain. Herein we discuss the complexity of the interactions between the cellular and non-cellular components of the TME and advances in the field as a whole. While the stroma of non-central nervous system (CNS) tissues is abundant in fibrillary collagens, laminins, and fibronectin, the normal brain extracellular matrix (ECM) predominantly includes proteoglycans, glycoproteins, and glycosaminoglycans, with fibrillary components typically found only in association with the vasculature. However, recent studies have found that in GBMs, the microenvironment evolves into a more complex array of components, with upregulated collagen gene expression and aligned fibrillary ECM networks. The interactions of glioma cells with the ECM and the degradation of matrix barriers are crucial for both single-cell and collective invasion into neighboring brain tissue. ECM-regulated mechanisms also contribute to immune exclusion, resulting in a major challenge to immunotherapy delivery and efficacy. Glioma cells chemically and physically control the function of their environment, co-opting complex signaling networks for their own benefit, resulting in radio- and chemo-resistance, tumor recurrence, and cancer progression. Targeting these interactions is an attractive strategy for overcoming therapy resistance, and we will discuss recent advances in preclinical studies, current clinical trials, and potential future clinical applications. In this review, we also provide a comprehensive discussion of the complexities of the interconnected cellular and non-cellular components of the microenvironmental landscape of brain tumors to guide the development of safe and effective therapeutic strategies against brain cancer.
Collapse
Affiliation(s)
- Syed M. Faisal
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Comba
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria L. Varela
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anna E. Argento
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Emily Brumley
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Clifford Abel
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G. Castro
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R. Lowenstein
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Pedro R. Lowenstein,
| |
Collapse
|
11
|
Keshavarz A, Salehi A, Khosravi S, Shariati Y, Nasrabadi N, Kahrizi MS, Maghsoodi S, Mardi A, Azizi R, Jamali S, Fotovat F. Recent findings on chimeric antigen receptor (CAR)-engineered immune cell therapy in solid tumors and hematological malignancies. Stem Cell Res Ther 2022; 13:482. [PMID: 36153626 PMCID: PMC9509604 DOI: 10.1186/s13287-022-03163-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/12/2022] [Indexed: 11/10/2022] Open
Abstract
Advancements in adoptive cell therapy over the last four decades have revealed various new therapeutic strategies, such as chimeric antigen receptors (CARs), which are dedicated immune cells that are engineered and administered to eliminate cancer cells. In this context, CAR T-cells have shown significant promise in the treatment of hematological malignancies. However, many obstacles limit the efficacy of CAR T-cell therapy in both solid tumors and hematological malignancies. Consequently, CAR-NK and CAR-M cell therapies have recently emerged as novel therapeutic options for addressing the challenges associated with CAR T-cell therapies. Currently, many CAR immune cell trials are underway in various human malignancies around the world to improve antitumor activity and reduce the toxicity of CAR immune cell therapy. This review will describe the comprehensive literature of recent findings on CAR immune cell therapy in a wide range of human malignancies, as well as the challenges that have emerged in recent years.
Collapse
Affiliation(s)
- Ali Keshavarz
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Salehi
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Islamic Azad University,, Isfahan (Khorasgan) Branch, Isfahan, Iran
| | - Setareh Khosravi
- Department of Orthodontics, School of Dentistry, Alborz University of Medical Sciences, Karaj, Iran
| | - Yasaman Shariati
- Department of General Surgery, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Navid Nasrabadi
- Department of Endodontics, School of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Sairan Maghsoodi
- Department of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amirhossein Mardi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramyar Azizi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Jamali
- Department of Endodontics, College of Stomatology, Stomatological Hospital, Xi’an Jiaotong University, Shaanxi, People’s Republic of China
| | - Farnoush Fotovat
- Department of Prosthodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
12
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
13
|
Lindorfer MA, Taylor RP. FcγR-Mediated Trogocytosis 2.0: Revisiting History Gives Rise to a Unifying Hypothesis. Antibodies (Basel) 2022; 11:antib11030045. [PMID: 35892705 PMCID: PMC9326535 DOI: 10.3390/antib11030045] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 12/25/2022] Open
Abstract
There is increasing interest in the clinical implications and immunology of trogocytosis, a process in which the receptors on acceptor cells remove and internalize cognate ligands from donor cells. We have reported that this phenomenon occurs in cancer immunotherapy, in which cells that express FcγR remove and internalize CD20 and bound mAbs from malignant B cells. This process can be generalized to include other reactions including the immune adherence phenomenon and antibody-induced immunosuppression. We discuss in detail FcγR-mediated trogocytosis and the evidence supporting a proposed predominant role for liver sinusoidal endothelial cells via the action of the inhibitory receptor FcγRIIb2. We describe experiments to test the validity of this hypothesis. The elucidation of the details of FcγR-mediated trogocytosis has the potential to allow for the development of novel therapies that can potentially block or enhance this reaction, depending upon whether the process leads to unfavorable or positive biological effects.
Collapse
|
14
|
Anderson GS, Ballester-Beltran J, Giotopoulos G, Guerrero JA, Surget S, Williamson JC, So T, Bloxham D, Aubareda A, Asby R, Walker I, Jenkinson L, Soilleux EJ, Roy JP, Teodósio A, Ficken C, Officer-Jones L, Nasser S, Skerget S, Keats JJ, Greaves P, Tai YT, Anderson KC, MacFarlane M, Thaventhiran JE, Huntly BJ, Lehner PJ, Chapman MA. Unbiased cell surface proteomics identifies SEMA4A as an effective immunotherapy target for myeloma. Blood 2022; 139:2471-2482. [PMID: 35134130 PMCID: PMC11022854 DOI: 10.1182/blood.2021015161] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/04/2022] [Indexed: 11/20/2022] Open
Abstract
The accessibility of cell surface proteins makes them tractable for targeting by cancer immunotherapy, but identifying suitable targets remains challenging. Here we describe plasma membrane profiling of primary human myeloma cells to identify an unprecedented number of cell surface proteins of a primary cancer. We used a novel approach to prioritize immunotherapy targets and identified a cell surface protein not previously implicated in myeloma, semaphorin-4A (SEMA4A). Using knock-down by short-hairpin RNA and CRISPR/nuclease-dead Cas9 (dCas9), we show that expression of SEMA4A is essential for normal myeloma cell growth in vitro, indicating that myeloma cells cannot downregulate the protein to avoid detection. We further show that SEMA4A would not be identified as a myeloma therapeutic target by standard CRISPR/Cas9 knockout screens because of exon skipping. Finally, we potently and selectively targeted SEMA4A with a novel antibody-drug conjugate in vitro and in vivo.
Collapse
Affiliation(s)
- Georgina S.F. Anderson
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | | | - George Giotopoulos
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Wellcome–MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Jose A. Guerrero
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Sylvanie Surget
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | | | - Tsz So
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - David Bloxham
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Anna Aubareda
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Ryan Asby
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Wellcome–MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Ieuan Walker
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Lesley Jenkinson
- CRUK–AstraZeneca Antibody Alliance Laboratory, Cambridge, United Kingdom
| | | | - James P. Roy
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Ana Teodósio
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Catherine Ficken
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | | | - Sara Nasser
- Translational Genomics Research Institute, Phoenix, AZ
| | - Sheri Skerget
- Translational Genomics Research Institute, Phoenix, AZ
| | | | - Peter Greaves
- Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Yu-Tzu Tai
- Dana Farber Cancer Institute, Boston, MA
| | | | - Marion MacFarlane
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
| | | | - Brian J.P. Huntly
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Wellcome–MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Paul J. Lehner
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Michael A. Chapman
- MRC Toxicology Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
15
|
Swamydas M, Murphy EV, Ignatz-Hoover JJ, Malek E, Driscoll JJ. Deciphering mechanisms of immune escape to inform immunotherapeutic strategies in multiple myeloma. J Hematol Oncol 2022; 15:17. [PMID: 35172851 PMCID: PMC8848665 DOI: 10.1186/s13045-022-01234-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/03/2022] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma is an incurable cancer characterized by the uncontrolled growth of malignant plasma cells nurtured within a permissive bone marrow microenvironment. While patients mount numerous adaptive immune responses directed against their disease, emerging data demonstrate that tumor intrinsic and extrinsic mechanisms allow myeloma cells to subvert host immunosurveillance and resist current therapeutic strategies. Myeloma downregulates antigens recognized by cellular immunity and modulates the bone marrow microenvironment to promote uncontrolled tumor proliferation, apoptotic resistance, and further hamper anti-tumor immunity. Additional resistance often develops after an initial clinical response to small molecules, immune-targeting antibodies, immune checkpoint blockade or cellular immunotherapy. Profound quantitative and qualitative dysfunction of numerous immune effector cell types that confer anti-myeloma immunity further supports myelomagenesis, disease progression and the emergence of drug resistance. Identification of tumor intrinsic and extrinsic resistance mechanisms may direct the design of rationally-designed drug combinations that prevent or overcome drug resistance to improve patient survival. Here, we summarize various mechanisms of immune escape as a means to inform novel strategies that may restore and improve host anti-myeloma immunity.
Collapse
Affiliation(s)
| | - Elena V Murphy
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, USA
| | - James J Ignatz-Hoover
- Seidman Cancer Center, University Hospitals, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Hematopoietic and Immune Cancer Biology Program, Cleveland, OH, USA
| | - Ehsan Malek
- Seidman Cancer Center, University Hospitals, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Hematopoietic and Immune Cancer Biology Program, Cleveland, OH, USA
| | - James J Driscoll
- Seidman Cancer Center, University Hospitals, Cleveland, OH, USA. .,Case Comprehensive Cancer Center, Hematopoietic and Immune Cancer Biology Program, Cleveland, OH, USA.
| |
Collapse
|
16
|
Reinert J, Beitzen-Heineke A, Wethmar K, Stelljes M, Fiedler W, Schwartz S. Loss of CD22 expression and expansion of a CD22 dim subpopulation in adults with relapsed/refractory B-lymphoblastic leukaemia after treatment with Inotuzumab-Ozogamicin. Ann Hematol 2021; 100:2727-2732. [PMID: 34331563 PMCID: PMC8510963 DOI: 10.1007/s00277-021-04601-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/04/2021] [Indexed: 12/02/2022]
Abstract
Treatment options for relapsed or refractory B-lymphoblastic leukaemia (r/r B-ALL) are limited and the prognosis of these patients remains dismal, but novel immunotherapeutic options such as the anti-CD22 antibody–drug-conjugate Inotuzumab-Ozogamicin (InO) have improved outcomes in these patients. Flow cytometry is essential to assess antigen-expression prior to treatment initiation of antigen-directed immunotherapies. Here, we present flow cytometric and clinical data of three adult patients with r/r B-ALL who failed treatment with InO associated with reduced or lost antigen-expression. In addition, we present comparative data on two different diagnostic CD22-specific antibody clones that exhibit significant differences in staining intensities.
Collapse
Affiliation(s)
- Jochim Reinert
- Department of Oncology, Haematology and Bone Marrow Transplantation With Section Pneumology, Hubertus Wald Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Antonia Beitzen-Heineke
- Department of Oncology, Haematology and Bone Marrow Transplantation With Section Pneumology, Hubertus Wald Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Wethmar
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Matthias Stelljes
- Department of Medicine A, Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Walter Fiedler
- Department of Oncology, Haematology and Bone Marrow Transplantation With Section Pneumology, Hubertus Wald Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Schwartz
- Department of Haematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität and Humboldt-Universität Zu Berlin, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
17
|
Sportoletti P, De Falco F, Del Papa B, Baldoni S, Guarente V, Marra A, Dorillo E, Rompietti C, Adamo FM, Ruggeri L, Di Ianni M, Rosati E. NK Cells in Chronic Lymphocytic Leukemia and Their Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22136665. [PMID: 34206399 PMCID: PMC8268440 DOI: 10.3390/ijms22136665] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Key features of chronic lymphocytic leukemia (CLL) are defects in the immune system and the ability of leukemic cells to evade immune defenses and induce immunosuppression, resulting in increased susceptibility to infections and disease progression. Several immune effectors are impaired in CLL, including T and natural killer (NK) cells. The role of T cells in defense against CLL and in CLL progression and immunotherapy has been extensively studied. Less is known about the role of NK cells in this leukemia, and data on NK cell alterations in CLL are contrasting. Besides studies showing that NK cells have intrinsic defects in CLL, there is a large body of evidence indicating that NK cell dysfunctions in CLL mainly depend on the escape mechanisms employed by leukemic cells. In keeping, it has been shown that NK cell functions, including antibody-dependent cellular cytotoxicity (ADCC), can be retained and/or restored after adequate stimulation. Therefore, due to their preserved ADCC function and the reversibility of CLL-related dysfunctions, NK cells are an attractive source for novel immunotherapeutic strategies in this disease, including chimeric antigen receptor (CAR) therapy. Recently, satisfying clinical responses have been obtained in CLL patients using cord blood-derived CAR-NK cells, opening new possibilities for further exploring NK cells in the immunotherapy of CLL. However, notwithstanding the promising results of this clinical trial, more evidence is needed to fully understand whether and in which CLL cases NK cell-based immunotherapy may represent a valid, alternative/additional therapeutic option for this leukemia. In this review, we provide an overview of the current knowledge about phenotypic and functional alterations of NK cells in CLL and the mechanisms by which CLL cells circumvent NK cell-mediated immunosurveillance. Additionally, we discuss the potential relevance of using NK cells in CLL immunotherapy.
Collapse
MESH Headings
- Biomarkers
- Cell Communication
- Disease Management
- Disease Susceptibility
- Humans
- Immune System/immunology
- Immune System/metabolism
- Immunotherapy/adverse effects
- Immunotherapy/methods
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Ligands
- Protein Binding
- Receptors, Natural Killer Cell/genetics
- Receptors, Natural Killer Cell/metabolism
- Treatment Outcome
- Tumor Escape/genetics
- Tumor Escape/immunology
Collapse
Affiliation(s)
- Paolo Sportoletti
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Filomena De Falco
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Beatrice Del Papa
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Stefano Baldoni
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Valerio Guarente
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Andrea Marra
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Erica Dorillo
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Chiara Rompietti
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Francesco Maria Adamo
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Loredana Ruggeri
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Mauro Di Ianni
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Department of Oncology and Hematology, Ospedale Civile “Santo Spirito”, ASL Pescara, 65124 Pescara, Italy
| | - Emanuela Rosati
- Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
- Correspondence:
| |
Collapse
|
18
|
Tumor Heterogeneity: A Great Barrier in the Age of Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13040806. [PMID: 33671881 PMCID: PMC7918981 DOI: 10.3390/cancers13040806] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
Throughout the history of oncology research, tumor heterogeneity has been a major hurdle for the successful treatment of cancer. As a result of aberrant changes in the tumor microenvironment such as high mutational burden, hypoxic conditions and abnormal vasculature, several malignant subpopulations often exist within a single tumor mass. Therapeutic intervention can also increase selective pressure towards subpopulations with acquired resistance. This phenomenon is often the cause of relapse in previously responsive patients, drastically changing the expected outcome of therapy. In the case of cancer immunotherapy, tumor heterogeneity is a substantial barrier as acquired resistance often takes the form of antigen escape and immunosuppression. In an effort to combat intrinsic resistance mechanisms, therapies are often combined as a multi-pronged approach to target multiple pathways simultaneously. These multi-therapy regimens have long been a mainstay of clinical oncology with chemotherapy cocktails but are more recently being investigated in the emerging landscape of immunotherapy. Furthermore, as high throughput technology becomes more affordable and accessible, researchers continue to deepen their understanding of the factors that influence tumor heterogeneity and shape the TME over the course of treatment regimens. In this review, we will investigate the factors that give rise to tumor heterogeneity and the impact it has on the field of immunotherapy. We will discuss how tumor heterogeneity causes resistance to various treatments and review the strategies currently being employed to overcome this challenging clinical hurdle. Finally, we will outline areas of research that should be prioritized to gain a better understanding of tumor heterogeneity and develop appropriate solutions.
Collapse
|
19
|
He K, Rad SMAH, Poudel A, McLellan AD. Compact Bidirectional Promoters for Dual-Gene Expression in a Sleeping Beauty Transposon. Int J Mol Sci 2020; 21:ijms21239256. [PMID: 33291599 PMCID: PMC7731152 DOI: 10.3390/ijms21239256] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/26/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Promoter choice is an essential consideration for transgene expression in gene therapy. The expression of multiple genes requires ribosomal entry or skip sites, or the use of multiple promoters. Promoter systems comprised of two separate, divergent promoters may significantly increase the size of genetic cassettes intended for use in gene therapy. However, an alternative approach is to use a single, compact, bidirectional promoter. We identified strong and stable bidirectional activity of the RPBSA synthetic promoter comprised of a fragment of the human Rpl13a promoter, together with additional intron/exon structures. The Rpl13a-based promoter drove long-term bidirectional activity of fluorescent proteins. Similar results were obtained for the EF1-α and LMP2/TAP1 promoters. However, in a lentiviral vector, the divergent bidirectional systems failed to produce sufficient titres to translate into an expression system for dual chimeric antigen receptor (CAR) expression. Although bidirectional promoters show excellent applicability to drive short RNA in Sleeping Beauty transposon systems, their possible use in the lentiviral applications requiring longer and more complex RNA, such as dual-CAR cassettes, is limited.
Collapse
|
20
|
Griggio V, Perutelli F, Salvetti C, Boccellato E, Boccadoro M, Vitale C, Coscia M. Immune Dysfunctions and Immune-Based Therapeutic Interventions in Chronic Lymphocytic Leukemia. Front Immunol 2020; 11:594556. [PMID: 33312177 PMCID: PMC7708380 DOI: 10.3389/fimmu.2020.594556] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/14/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy characterized by a wide range of tumor-induced alterations, which affect both the innate and adaptive arms of the immune response, and accumulate during disease progression. In recent years, the development of targeted therapies, such as the B-cell receptor signaling inhibitors and the Bcl-2 protein inhibitor venetoclax, has dramatically changed the treatment landscape of CLL. Despite their remarkable anti-tumor activity, targeted agents have some limitations, which include the development of drug resistance mechanisms and the inferior efficacy observed in high-risk patients. Therefore, additional treatments are necessary to obtain deeper responses and overcome drug resistance. Allogeneic hematopoietic stem cell transplantation (HSCT), which exploits immune-mediated graft-versus-leukemia effect to eradicate tumor cells, currently represents the only potentially curative therapeutic option for CLL patients. However, due to its potential toxicities, HSCT can be offered only to a restricted number of younger and fit patients. The growing understanding of the complex interplay between tumor cells and the immune system, which is responsible for immune escape mechanisms and tumor progression, has paved the way for the development of novel immune-based strategies. Despite promising preclinical observations, results from pilot clinical studies exploring the safety and efficacy of novel immune-based therapies have been sometimes suboptimal in terms of long-term tumor control. Therefore, further advances to improve their efficacy are needed. In this context, possible approaches include an earlier timing of immunotherapy within the treatment sequencing, as well as the possibility to improve the efficacy of immunotherapeutic agents by administering them in combination with other anti-tumor drugs. In this review, we will provide a comprehensive overview of main immune defects affecting patients with CLL, also describing the complex networks leading to immune evasion and tumor progression. From the therapeutic standpoint, we will go through the evolution of immune-based therapeutic approaches over time, including i) agents with broad immunomodulatory effects, such as immunomodulatory drugs, ii) currently approved and next-generation monoclonal antibodies, and iii) immunotherapeutic strategies aiming at activating or administering immune effector cells specifically targeting leukemic cells (e.g. bi-or tri-specific antibodies, tumor vaccines, chimeric antigen receptor T cells, and checkpoint inhibitors).
Collapse
Affiliation(s)
- Valentina Griggio
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Francesca Perutelli
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Chiara Salvetti
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elia Boccellato
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Mario Boccadoro
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Candida Vitale
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Marta Coscia
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
21
|
Tang JJJ, Sung AP, Guglielmo MJ, Navarrete-Galvan L, Redelman D, Smith-Gagen J, Hudig D. Natural Killer (NK) Cell Expression of CD2 as a Predictor of Serial Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC). Antibodies (Basel) 2020; 9:antib9040054. [PMID: 33081115 PMCID: PMC7709134 DOI: 10.3390/antib9040054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
NK cell ADCC supports monoclonal antibody anti-tumor therapies. We investigated serial ADCC and whether it could be predicted by NK phenotypes, including expression of CD16A, CD2 and perforin. CD16A, the NK receptor for antibodies, has AA158 valine or phenylalanine variants with different affinities for IgG. CD2, a costimulatory protein, associates with CD16A and can augment CD16A-signaling. Pore-forming perforin is essential for rapid NK-mediated killing. NK cells were monitored for their ADCC serial killing frequency (KF). KF is the average number of target cells killed per cell by a cytotoxic cell population. KF comparisons were made at 1:4 CD16pos NK effector:target ratios. ADCC was toward Daudi cells labeled with 51Cr and obinutuzumab anti-CD20 antibody. CD16A genotypes were determined by DNA sequencing. CD2, CD16A, and perforin expression was monitored by flow cytometry. Serial killing KFs varied two-fold among 24 donors and were independent of CD16A genotypes and perforin levels. However, high percentages of CD2pos of the CD16Apos NK cells and high levels of CD16A were associated with high KFs. ROC analysis indicated that the %CD2pos of CD16Apos NK cells can predict KFs. In conclusion, the extent of serial ADCC varies significantly among donors and appears predictable by the CD2posCD16Apos NK phenotype.
Collapse
Affiliation(s)
- Jennifer J.-J. Tang
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Alexander P. Sung
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Michael J. Guglielmo
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Lydia Navarrete-Galvan
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Doug Redelman
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA;
| | - Julie Smith-Gagen
- School of Community Health Sciences, University of Nevada, Reno, 1664 N. Virginia St., Reno, NV 89557, USA;
| | - Dorothy Hudig
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
- Correspondence: ; Tel.: +1-775-784-4430
| |
Collapse
|
22
|
Qin L, Wang S, Dominguez D, Long A, Chen S, Fan J, Ahn J, Skakuj K, Huang Z, Lee A, Mirkin C, Zhang B. Development of Spherical Nucleic Acids for Prostate Cancer Immunotherapy. Front Immunol 2020; 11:1333. [PMID: 32733447 PMCID: PMC7362897 DOI: 10.3389/fimmu.2020.01333] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/26/2020] [Indexed: 01/02/2023] Open
Abstract
Although the strategy of therapeutic vaccination for the treatment of prostate cancer has advanced to and is available in the clinic (Sipuleucel-T), the efficacy of such therapy remains limited. Here, we develop Immunostimulatory Spherical Nucleic Acid (IS-SNA) nanostructures comprised of CpG oligonucleotides as adjuvant and prostate cancer peptide antigens, and evaluate their antitumor efficacy in syngeneic mouse models of prostate cancer. IS-SNAs with the specific structural feature of presenting both antigen and adjuvant CpG on the surface (hybridized model (HM) SNAs) induce stronger cytotoxic T lymphocyte (CTL) mediated antigen-specific killing of target cells than that for IS-SNAs with CpG on the surface and antigen encapsulated within the core (encapsulated model (EM) SNAs). Mechanistically, HM SNAs increase the co-delivery of CpG and antigen to dendritic cells over that for EM SNAs or admixtures of linear CpG and peptide, thereby improving cross-priming of antitumor CD8+ T cells. As a result, vaccination with HM SNAs leads to more effective antitumor immune responses in two prostate cancer models. These data demonstrate the importance of the structural positioning of peptide antigens together with adjuvants within IS-SNAs to the efficacy of IS-SNA-based cancer immunotherapy.
Collapse
Affiliation(s)
- Lei Qin
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Shuya Wang
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL, United States
| | - Donye Dominguez
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Alan Long
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Siqi Chen
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jie Fan
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jihae Ahn
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Kacper Skakuj
- Department of Chemistry, Northwestern University, Evanston, IL, United States
| | - Ziyin Huang
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, United States
| | - Andrew Lee
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
| | - Chad Mirkin
- Department of Chemistry, Northwestern University, Evanston, IL, United States.,The International Institute for Nanotechnology, Northwestern University, Evanston, IL, United States
| | - Bin Zhang
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
23
|
Vito A, El-Sayes N, Mossman K. Hypoxia-Driven Immune Escape in the Tumor Microenvironment. Cells 2020; 9:E992. [PMID: 32316260 PMCID: PMC7227025 DOI: 10.3390/cells9040992] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/05/2020] [Accepted: 04/13/2020] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment is a complex ecosystem comprised of many different cell types, abnormal vasculature and immunosuppressive cytokines. The irregular growth kinetics with which tumors grow leads to increased oxygen consumption and, in turn, hypoxic conditions. Hypoxia has been associated with poor clinical outcome, increased tumor heterogeneity, emergence of resistant clones and evasion of immune detection. Additionally, hypoxia-driven cell death pathways have traditionally been thought of as tolerogenic processes. However, as researchers working in the field of immunotherapy continue to investigate and unveil new types of immunogenic cell death (ICD), it has become clear that, in some instances, hypoxia may actually induce ICD within a tumor. In this review, we will discuss hypoxia-driven immune escape that drives poor prognostic outcomes, the ability of hypoxia to induce ICD and potential therapeutic targets amongst hypoxia pathways.
Collapse
Affiliation(s)
- Alyssa Vito
- Department of Biochemistry and Biomedical Sciences, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (A.V.); (N.E.-S.)
| | - Nader El-Sayes
- Department of Biochemistry and Biomedical Sciences, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (A.V.); (N.E.-S.)
| | - Karen Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
24
|
Shirasu N, Shibaguchi H, Yamada H, Kuroki M, Yasunaga S. Highly versatile cancer photoimmunotherapy using photosensitizer-conjugated avidin and biotin-conjugated targeting antibodies. Cancer Cell Int 2019; 19:299. [PMID: 31787847 PMCID: PMC6858743 DOI: 10.1186/s12935-019-1034-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/12/2019] [Indexed: 01/19/2023] Open
Abstract
Background Photoimmunotherapy (PIT) employing antibody-photosensitizer conjugates is a promising treatment for cancer. However, the fixed antigen specificity severely limits the efficacy and the applicability. Here we describe a universal strategy for PIT of cancer by using a near-infrared (NIR) photosensitizer IRDye700DX-conjugated NeutrAvidin, designated as AvIR, together with various biotinylated antibodies (BioAbs) for cellular targeting. Methods Cytotoxicity of AvIR-mediated PIT was evaluated by fluorescence imaging and cell viability assay. Phototoxic effect on tumorigenicity was assessed by tumorsphere-formation assay and Matrigel invasion assay. Cancer stem cell-like side-population (SP) cells were identified by flow cytometry. Results CHO cells stably expressing carcinoembryonic antigen or EpCAM were pre-labeled with each BioAb for the corresponding antigen, followed by AvIR administration. NIR light irradiation specifically killed the targeted cells, but not off-targets, demonstrating that the AvIR-mediated PIT does work as expected. CSC-like subpopulation of MCF-7 cells (CD24low/CD44high) and SP of HuH-7 cells (CD133+/EpCAM+) were effectively targeted and photokilled by AvIR-PIT with anti-CD44 BioAb or anti-CD133/anti-EpCAM BioAbs, respectively. As results, the neoplastic features of the cell lines were sufficiently suppressed. Cancer-associated fibroblast (CAF)-targeted AvIR-PIT by using anti-fibroblast activation protein BioAb showed an abolishment of CAF-enhanced clonogenicity of MCF-7 cells. Conclusions Collectively, our results demonstrate that AvIR-mediated PIT can greatly broaden the applicable range of target specificity, with feasibility of efficacious and integrative control of CSC and its microenvironment.
Collapse
Affiliation(s)
- Naoto Shirasu
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, 7-45-1 Jonan-ku, Fukuoka, 814-0180 Japan
| | - Hirotomo Shibaguchi
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, 7-45-1 Jonan-ku, Fukuoka, 814-0180 Japan
| | - Hiromi Yamada
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, 7-45-1 Jonan-ku, Fukuoka, 814-0180 Japan
| | - Masahide Kuroki
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, 7-45-1 Jonan-ku, Fukuoka, 814-0180 Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, 7-45-1 Jonan-ku, Fukuoka, 814-0180 Japan
| |
Collapse
|
25
|
Paul MR, Wong V, Aristizabal P, Kuo DJ. Treatment of Recurrent Refractory Pediatric Pre-B Acute Lymphoblastic Leukemia Using Inotuzumab Ozogamicin Monotherapy Resulting in CD22 Antigen Expression Loss as a Mechanism of Therapy Resistance. J Pediatr Hematol Oncol 2019; 41:e546-e549. [PMID: 30807395 PMCID: PMC7216755 DOI: 10.1097/mph.0000000000001440] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Inotuzumab ozogamicin is a novel antibody-drug conjugate that targets CD22, a common antigen on pre-B acute lymphoblastic leukemia cells. OBSERVATIONS A 7-year-old boy with pre-B acute lymphoblastic leukemia in his second relapse was given 2 cycles of inotuzumab ozogamicin. He responded morphologically with a negative bone marrow evaluation. However, he relapsed in cycle 3 of therapy with a loss of CD22 expression on his lymphoblast population. CONCLUSION To our knowledge, this is the first published case of CD22 expression loss as a mechanism of therapy resistance for inotuzumab ozogamicin.
Collapse
Affiliation(s)
- Megan R. Paul
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of California San Diego, Peckham Center for Cancer and Blood Disorders, Rady Children’s Hospital San Diego, San Diego
| | - Victor Wong
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of California San Diego, Peckham Center for Cancer and Blood Disorders, Rady Children’s Hospital San Diego, San Diego
| | - Paula Aristizabal
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of California San Diego, Peckham Center for Cancer and Blood Disorders, Rady Children’s Hospital San Diego, San Diego,Department of Populations Sciences, Disparities, and Community Outreach and Engagement, University of California San Diego, Moores Cancer Center, La Jolla, CA
| | - Dennis J. Kuo
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of California San Diego, Peckham Center for Cancer and Blood Disorders, Rady Children’s Hospital San Diego, San Diego
| |
Collapse
|
26
|
Dunn ZS, Mac J, Wang P. T cell immunotherapy enhanced by designer biomaterials. Biomaterials 2019; 217:119265. [PMID: 31271861 PMCID: PMC6689323 DOI: 10.1016/j.biomaterials.2019.119265] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/21/2022]
Abstract
Cancer immunotherapy has recently burst onto the center stage of cancer treatment and research. T lymphocyte adoptive cellular transfer (ACT), a form of cancer immunotherapy, has spawned unprecedented complete remissions for terminal patients with certain leukemias and lymphomas. Unfortunately, the successes have been overshadowed by the disappointing clinical results of ACT administered to treat solid tumors, in addition to the toxicities associated with the treatment, a lack of efficacy in a significant proportion of the patient population, and cancer relapse following the treatment. Biomaterials hold the promise of addressing these shortcomings. ACT consists of two main stages - T lymphocyte ex vivo expansion followed by reinfusion into the patient - and biomaterials can improve the efficacy of ACT at both stages. In this review, we highlight recent advances in the use of biomaterials for T lymphocyte adoptive cellular cancer immunotherapy and discuss the challenges at each stage.
Collapse
Affiliation(s)
- Zachary S Dunn
- Mork Family Department of of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, United States
| | - John Mac
- Mork Family Department of of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, United States
| | - Pin Wang
- Mork Family Department of of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, United States; Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States; Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
27
|
Nyambura LW, Muñoz AA, le Coutre P, Walden P. HLA class I-restricted T cell epitopes isolated and identified from myeloid leukemia cells. Sci Rep 2019; 9:14029. [PMID: 31575892 PMCID: PMC6773711 DOI: 10.1038/s41598-019-50341-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/11/2019] [Indexed: 12/19/2022] Open
Abstract
Leukemia-associated antigens (LAAs) and HLA-I epitopes published previously have shown promise in inducing leukemia-specific T cell responses. However, the clinical responses are limited, and clinical effectiveness is yet to be achieved. Limitations, among others, being the LAAs themselves, the indirect approach to HLA-I epitope identification by reverse immunology, and the use of single or few LAAs and HLA-I epitopes, which limits the spectrum of inducible tumor-specific T cells. Use of a direct approach to identify naturally processed and presented HLA-I epitopes from LAAs, and higher numbers of antigens for T cell-mediated immunotherapy for leukemia may enhance clinical responses and broaden clinical effectiveness. In a prior study we used immunoaffinity purification of HLA-I peptide complexes from the differentiated myeloid tumor cell lines MUTZ3 and THP1 coupled to high-performance liquid chromatography tandem mass spectrometry (LC-MS/MS). From this we identified in the current study seven new HLA-I epitopes and the corresponding LAAs for myeloid leukemia. In comparison, the myeloid HLA-I epitopes reported here were generally stronger HLA-binders that induce stronger T cell responses than those previously published, and their source LAAs had higher immunogenicity, higher expression levels in myeloid tumors cells compared to normal hemopoietin and other major normal tissues, and more protein interaction partners, and they are targeted by CD8 T cells in CML patients. This study analyses and compares the LAAs and HLA-I epitopes based on various immunotherapeutic targets selection criteria, and highlights new targets for T cell-mediated immunotherapy for leukemia.
Collapse
Affiliation(s)
- Lydon Wainaina Nyambura
- Department of Dermatology, Venerology and Allergology, Clinical Research Group 'Tumor Immunology', Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10098, Berlin, Germany
| | - Alejandro Azorin Muñoz
- Department of Dermatology, Venerology and Allergology, Clinical Research Group 'Tumor Immunology', Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10098, Berlin, Germany
| | - Philipp le Coutre
- Medical Department, Division of Hematology and Oncology, Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10098, Berlin, Germany
| | - Peter Walden
- Department of Dermatology, Venerology and Allergology, Clinical Research Group 'Tumor Immunology', Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10098, Berlin, Germany.
| |
Collapse
|
28
|
Huang SH, McCann CD, Mota TM, Wang C, Lipkin SM, Jones RB. Have Cells Harboring the HIV Reservoir Been Immunoedited? Front Immunol 2019; 10:1842. [PMID: 31447850 PMCID: PMC6691121 DOI: 10.3389/fimmu.2019.01842] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/22/2019] [Indexed: 01/05/2023] Open
Abstract
Immunoediting is an important concept in oncology, delineating the mechanisms through which tumors are selected for resistance to immune-mediated elimination. The recent emergence of immunotherapies, such as checkpoint inhibitors, as pillars of cancer therapy has intensified interest in immunoediting as a constraint limiting the efficacy of these approaches. Immunoediting manifests at a number of levels for different cancers, for example through the establishment of immunosuppressive microenvironments within solid tumors. Of particular interest to the current review, selection also occurs at the cellular level; and recent studies have revealed novel mechanisms by which tumor cells acquire intrinsic resistance to immune recognition and elimination. While the selection of escape mutations in viral epitopes by HIV-specific T cells, which is a hallmark of chronic HIV infection, can be considered a form of immunoediting, few studies have considered the possibility that HIV-infected cells themselves may parallel tumors in having differential intrinsic susceptibilities to immune-mediated elimination. Such selection, on the level of an infected cell, may not play a significant role in untreated HIV, where infection is propagated by high levels of cell-free virus produced by cells that quickly succumb to viral cytopathicity. However, it may play an unappreciated role in individuals treated with effective antiretroviral therapy where viral replication is abrogated. In this context, an "HIV reservoir" persists, comprising long-lived infected cells which undergo extensive and dynamic clonal expansion. The ability of these cells to persist in infected individuals has generally been attributed to viral latency, thought to render them invisible to immune recognition, and/or to their compartmentalization in anatomical sites that are poorly accessible to immune effectors. Recent data from ex vivo studies have led us to propose that reservoir-harboring cells may additionally have been selected for intrinsic resistance to CD8+ T cells, limiting their elimination even in the context of antigen expression. Here, we draw on knowledge from tumor immunoediting to discuss potential mechanisms by which clones of HIV reservoir-harboring cells may resist elimination by CD8+ T cells. The establishment of such parallels may provide a premise for testing therapeutics designed to sensitize tumor cells to immune-mediated elimination as novel approaches aimed at curing HIV infection.
Collapse
Affiliation(s)
- Szu-Han Huang
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Chase D. McCann
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States
| | - Talia M. Mota
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Chao Wang
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Steven M. Lipkin
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - R. Brad Jones
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States
| |
Collapse
|
29
|
Goyal A, Patel S, Goyal K, Morgan EA, Foreman RK. Variable loss of CD30 expression by immunohistochemistry in recurrent cutaneous CD30+ lymphoid neoplasms treated with brentuximab vedotin. J Cutan Pathol 2019; 46:823-829. [PMID: 31286556 DOI: 10.1111/cup.13545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/28/2019] [Accepted: 06/26/2019] [Indexed: 01/08/2023]
Abstract
AIMS Brentuximab vedotin is a monoclonal anti-CD30 antibody-drug conjugate that has been used to treat a variety of CD30+ neoplasms. The phenomenon of antigen loss has been observed in patients treated with the anti-CD20 antibody rituximab. This study seeks to assess for antigen loss in the setting of recurrent CD30+ neoplasms treated with brentuximab vedotin. METHODS We report nine cases of persistent/recurrent cutaneous CD30+ lymphoid neoplasms that demonstrated variable CD30 expression after treatment with brentuximab vedotin. Cases include MF (n = 6), cutaneous T-cell lymphoma, not otherwise specified (n = 1), and anaplastic large cell lymphoma (ALCL), both primary (n = 1) and systemic (n = 1). RESULTS Immunohistochemical staining revealed decreased CD30 expression following brentuximab vedotin therapy in seven of nine cases. In these seven cases, the pre-treatment percent of tumor cells staining for CD30 ranged from 10% to 100% (mean 50.0%, SD 27.8%), compared to 5% to 50% (mean 14.5%, SD 14.8%, P = 0.003) at recurrence. CONCLUSIONS This case series highlights the finding that CD30 positivity can be variable in recurrences after treatment with anti-CD30 antibodies. This serves to raise awareness of the phenomenon of antigen loss after treatment with brentuximab vedotin and underscores the utility of performing multiple biopsies and/or employing molecular diagnostic techniques in patients with recurrent/persistent disease.
Collapse
Affiliation(s)
- Amrita Goyal
- Department of Dermatology, University of Minnesota, Minneapolis, Minnesota
| | - Sanjay Patel
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Kavita Goyal
- Department of Dermatology, University of Minnesota, Minneapolis, Minnesota
| | - Elizabeth A Morgan
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ruth K Foreman
- Dermatopathology Unit, Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
30
|
Minutolo NG, Hollander EE, Powell DJ. The Emergence of Universal Immune Receptor T Cell Therapy for Cancer. Front Oncol 2019; 9:176. [PMID: 30984613 PMCID: PMC6448045 DOI: 10.3389/fonc.2019.00176] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/01/2019] [Indexed: 12/17/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells have shown great success in the treatment of CD19+ hematological malignancies, leading to their recent approval by the FDA as a new cancer treatment modality. However, their broad use is limited since a CAR targets a single tumor associated antigen (TAA), which is not effective against tumors with heterogeneous TAA expression or emerging antigen loss variants. Further, stably engineered CAR T cells can continually and uncontrollably proliferate and activate in response to antigen, potentially causing fatal on-target off-tumor toxicity, cytokine release syndrome, or neurotoxicity without a method of control or elimination. To address these issues, our lab and others have developed various universal immune receptors (UIRs) that allow for targeting of multiple TAAs by T cells expressing a single receptor. UIRs function through the binding of an extracellular adapter domain which acts as a bridge between intracellular T cell signaling domains and a soluble tumor antigen targeting ligand (TL). The dissociation of TAA targeting and T cell signaling confers many advantages over standard CAR therapy, such as dose control of T cell effector function, the ability to simultaneously or sequentially target multiple TAAs, and control of immunologic synapse geometry. There are currently four unique UIR platform types: ADCC-mediating Fc-binding immune receptors, bispecific protein engaging immune receptors, natural binding partner immune receptors, and anti-tag CARs. These UIRs all allow for potential benefits over standard CARs, but also bring unique engineering challenges that will have to be addressed to achieve maximal efficacy and safety in the clinic. Still, UIRs present an exciting new avenue for adoptive T cell transfer therapies and could lead to their expanded use in areas which current CAR therapies have failed. Here we review the development of each UIR platform and their unique functional benefits, and detail the potential hurdles that may need to be overcome for continued clinical translation.
Collapse
Affiliation(s)
- Nicholas G Minutolo
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, PA, United States.,Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, PA, United States.,Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Erin E Hollander
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA, United States.,Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Daniel J Powell
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
31
|
Chuntova P, Downey KM, Hegde B, Almeida ND, Okada H. Genetically Engineered T-Cells for Malignant Glioma: Overcoming the Barriers to Effective Immunotherapy. Front Immunol 2019; 9:3062. [PMID: 30740109 PMCID: PMC6357938 DOI: 10.3389/fimmu.2018.03062] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Malignant gliomas carry a dismal prognosis. Conventional treatment using chemo- and radiotherapy has limited efficacy with adverse events. Therapy with genetically engineered T-cells, such as chimeric antigen receptor (CAR) T-cells, may represent a promising approach to improve patient outcomes owing to their potential ability to attack highly infiltrative tumors in a tumor-specific manner and possible persistence of the adaptive immune response. However, the unique anatomical features of the brain and susceptibility of this organ to irreversible tissue damage have made immunotherapy especially challenging in the setting of glioma. With safety concerns in mind, multiple teams have initiated clinical trials using CAR T-cells in glioma patients. The valuable lessons learnt from those trials highlight critical areas for further improvement: tackling the issues of the antigen presentation and T-cell homing in the brain, immunosuppression in the glioma microenvironment, antigen heterogeneity and off-tumor toxicity, and the adaptation of existing clinical therapies to reflect the intricacies of immune response in the brain. This review summarizes the up-to-date clinical outcomes of CAR T-cell clinical trials in glioma patients and examines the most pressing hurdles limiting the efficacy of these therapies. Furthermore, this review uses these hurdles as a framework upon which to evaluate cutting-edge pre-clinical strategies aiming to overcome those barriers.
Collapse
Affiliation(s)
- Pavlina Chuntova
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Kira M Downey
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Bindu Hegde
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Neil D Almeida
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States.,George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States.,The Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States.,Cancer Immunotherapy Program, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Hammerl D, Rieder D, Martens JWM, Trajanoski Z, Debets R. Adoptive T Cell Therapy: New Avenues Leading to Safe Targets and Powerful Allies. Trends Immunol 2018; 39:921-936. [PMID: 30309702 DOI: 10.1016/j.it.2018.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/12/2018] [Accepted: 09/12/2018] [Indexed: 12/30/2022]
Abstract
Adoptive transfer of TCR-engineered T cells is a potent therapy, able to induce clinical responses in different human malignancies. Nevertheless, treatment toxicities may occur and, in particular for solid tumors, responses may be variable and often not durable. To address these challenges, it is imperative to carefully select target antigens and to immunologically interrogate the corresponding tumors when designing optimal T cell therapies. Here, we review recent advances, covering both omics- and laboratory tools that can enable the selection of optimal T cell epitopes and TCRs as well as the identification of dominant immune evasive mechanisms within tumor tissues. Furthermore, we discuss how these techniques may aid in a rational design of effective combinatorial adoptive T cell therapies.
Collapse
Affiliation(s)
- Dora Hammerl
- Laboratory of Tumor Immunology, Erasmus MC-Cancer Institute, Rotterdam, The Netherlands
| | - Dietmar Rieder
- Division of Bioinformatics, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC-Cancer Institute, Rotterdam, The Netherlands
| | - Zlatko Trajanoski
- Division of Bioinformatics, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Reno Debets
- Laboratory of Tumor Immunology, Erasmus MC-Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
33
|
Abstract
The rapid development of immunomodulatory cancer therapies has led to a concurrent increase in the application of informatics techniques to the analysis of tumors, the tumor microenvironment, and measures of systemic immunity. In this review, the use of tumors to gather genetic and expression data will first be explored. Next, techniques to assess tumor immunity are reviewed, including HLA status, predicted neoantigens, immune microenvironment deconvolution, and T-cell receptor sequencing. Attempts to integrate these data are in early stages of development and are discussed in this review. Finally, we review the application of these informatics strategies to therapy development, with a focus on vaccines, adoptive cell transfer, and checkpoint blockade therapies.
Collapse
Affiliation(s)
- J Hammerbacher
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston
| | - A Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
- Adaptive Biotechnologies, Seattle, USA
| |
Collapse
|