Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 245
Filtrar
1.
J Virol ; 98(6): e0003824, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38767356

RESUMEN

Recent progress on chimeric antigen receptor (CAR)-NK cells has shown promising results in treating CD19-positive lymphoid tumors with minimal toxicities [including graft versus host disease (GvHD) and cytokine release syndrome (CRS) in clinical trials. Nevertheless, the use of CAR-NK cells in combating viral infections has not yet been fully explored. Previous studies have shown that CAR-NK cells expressing S309 single-chain fragment variable (scFv), hereinafter S309-CAR-NK cells, can bind to SARS-CoV-2 wildtype pseudotyped virus (PV) and effectively kill cells expressing wild-type spike protein in vitro. In this study, we further demonstrate that the S309-CAR-NK cells can bind to different SARS-CoV-2 variants, including the B.1.617.2 (Delta), B.1.621 (Mu), and B.1.1.529 (Omicron) variants in vitro. We also show that S309-CAR-NK cells reduce virus loads in the NOD/SCID gamma (NSG) mice expressing the human angiotensin-converting enzyme 2 (hACE2) receptor challenged with SARS-CoV-2 wild-type (strain USA/WA1/2020). Our study demonstrates the potential use of S309-CAR-NK cells for inhibiting infection by SARS-CoV-2 and for the potential treatment of COVID-19 patients unresponsive to otherwise currently available therapeutics. IMPORTANCE: Chimeric antigen receptor (CAR)-NK cells can be "off-the-shelf" products that treat various diseases, including cancer, infections, and autoimmune diseases. In this study, we engineered natural killer (NK) cells to express S309 single-chain fragment variable (scFv), to target the Spike protein of SARS-CoV-2, hereinafter S309-CAR-NK cells. Our study shows that S309-CAR-NK cells are effective against different SARS-CoV-2 variants, including the B.1.617.2 (Delta), B.1.621 (Mu), and B.1.1.529 (Omicron) variants. The S309-CAR-NK cells can (i) directly bind to SARS-CoV-2 pseudotyped virus (PV), (ii) competitively bind to SARS-CoV-2 PV with 293T cells expressing the human angiotensin-converting enzyme 2 (hACE2) receptor (293T-hACE2 cells), (iii) specifically target and lyse A549 cells expressing the spike protein, and (iv) significantly reduce the viral loads of SARS-CoV-2 wild-type (strain USA/WA1/2020) in the lungs of NOD/SCID gamma (NSG) mice expressing hACE2 (hACE2-NSG mice). Altogether, the current study demonstrates the potential use of S309-CAR-NK immunotherapy as an alternative treatment for COVID-19 patients.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Células Asesinas Naturales , Receptores Quiméricos de Antígenos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Carga Viral , Animales , SARS-CoV-2/inmunología , Células Asesinas Naturales/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Ratones , Humanos , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , COVID-19/inmunología , COVID-19/virología , COVID-19/terapia , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/genética , Ratones SCID , Ratones Endogámicos NOD
2.
Trends Immunol ; 43(10): 833-847, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36058806

RESUMEN

Natural killer (NK) cells, a crucial component of the innate immune system, have long been of clinical interest for their antitumor properties. Almost every aspect of NK cell immunity is regulated by interleukin-15 (IL-15), a cytokine in the common γ-chain family. Several current clinical trials are using IL-15 or its analogs to treat various cancers. Moreover, NK cells are being genetically modified to produce membrane-bound or secretory IL-15. Here, we discuss the key role of IL-15 signaling in NK cell immunity and provide an up-to-date overview of IL-15 in NK cell therapy.


Asunto(s)
Interleucina-15 , Neoplasias , Citocinas , Humanos , Inmunoterapia , Células Asesinas Naturales , Neoplasias/terapia
3.
Proc Natl Acad Sci U S A ; 119(25): e2122379119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35696582

RESUMEN

Acute myeloid leukemia (AML) remains a therapeutic challenge, and a paucity of tumor-specific targets has significantly hampered the development of effective immune-based therapies. Recent paradigm-changing studies have shown that natural killer (NK) cells exhibit innate memory upon brief activation with IL-12 and IL-18, leading to cytokine-induced memory-like (CIML) NK cell differentiation. CIML NK cells have enhanced antitumor activity and have shown promising results in early phase clinical trials in patients with relapsed/refractory AML. Here, we show that arming CIML NK cells with a neoepitope-specific chimeric antigen receptor (CAR) significantly enhances their antitumor responses to nucleophosphmin-1 (NPM1)-mutated AML while avoiding off-target toxicity. CIML NK cells differentiated from peripheral blood NK cells were efficiently transduced to express a TCR-like CAR that specifically recognizes a neoepitope derived from the cytosolic oncogenic NPM1-mutated protein presented by HLA-A2. These CAR CIML NK cells displayed enhanced activity against NPM1-mutated AML cell lines and patient-derived leukemic blast cells. CAR CIML NK cells persisted in vivo and significantly improved AML outcomes in xenograft models. Single-cell RNA sequencing and mass cytometry analyses identified up-regulation of cell proliferation, protein folding, immune responses, and major metabolic pathways in CAR-transduced CIML NK cells, resulting in tumor-specific, CAR-dependent activation and function in response to AML target cells. Thus, efficient arming of CIML NK cells with an NPM1-mutation-specific TCR-like CAR substantially improves their innate antitumor responses against an otherwise intracellular mutant protein. These preclinical findings justify evaluating this approach in clinical trials in HLA-A2+ AML patients with NPM1c mutations.


Asunto(s)
Memoria Inmunológica , Células de Memoria Inmunológica , Inmunoterapia Adoptiva , Células Asesinas Naturales , Leucemia Mieloide Aguda , Nucleofosmina , Receptores Quiméricos de Antígenos , Antígeno HLA-A2/inmunología , Humanos , Células de Memoria Inmunológica/inmunología , Células de Memoria Inmunológica/trasplante , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/trasplante , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Mutación , Nucleofosmina/genética , Nucleofosmina/inmunología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología
4.
Mol Cancer ; 23(1): 8, 2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-38195534

RESUMEN

Treatment for renal cell carcinoma (RCC) has improved dramatically over the last decade, shifting from high-dose cytokine therapy in combination with surgical resection of tumors to targeted therapy, immunotherapy, and combination therapies. However, curative treatment, particularly for advanced-stage disease, remains rare. Cell therapy as a "living drug" has achieved hematological malignancy cures with a high response rate, and significant research efforts have been made to facilitate its translation to solid tumors. Herein, we overview the cellular therapies for RCC focusing on allogeneic hematopoietic stem cell transplantation, T cell receptor gene-modified T cells, chimeric antigen receptor (CAR) T cells, CAR natural killer (NK) cells, lymphokine-activated killer (LAK) cells, γδ T cells, and dendritic cell vaccination. We have also included perspectives for using other recent approaches, such as CAR macrophages, dendritic cell-cytokine induced killer cells and regulatory CAR-T cells to shed light on preclinical development of cell therapy and advancing cell therapy into clinic to achieve cures for RCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/terapia , Inmunoterapia , Tratamiento Basado en Trasplante de Células y Tejidos , Terapia Combinada , Neoplasias Renales/terapia
5.
Biol Chem ; 405(7-8): 485-515, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-38766710

RESUMEN

Chimeric antigen receptor (CAR)-T cell therapy has led to remarkable clinical outcomes in the treatment of hematological malignancies. However, challenges remain, such as limited infiltration into solid tumors, inadequate persistence, systemic toxicities, and manufacturing insufficiencies. The use of alternative cell sources for CAR-based therapies, such as natural killer cells (NK), macrophages (MΦ), invariant Natural Killer T (iNKT) cells, γδT cells, neutrophils, and induced pluripotent stem cells (iPSC), has emerged as a promising avenue. By harnessing these cells' inherent cytotoxic mechanisms and incorporating CAR technology, common CAR-T cell-related limitations can be effectively mitigated. We herein present an overview of the tumoricidal mechanisms, CAR designs, and manufacturing processes of CAR-NK cells, CAR-MΦ, CAR-iNKT cells, CAR-γδT cells, CAR-neutrophils, and iPSC-derived CAR-cells, outlining the advantages, limitations, and potential solutions of these therapeutic strategies.


Asunto(s)
Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/inmunología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/inmunología , Inmunoterapia Adoptiva , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/citología , Macrófagos/inmunología , Macrófagos/citología , Macrófagos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Animales , Neoplasias/terapia , Neoplasias/inmunología , Linfocitos T/inmunología , Linfocitos T/citología
6.
Cytotherapy ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38970613

RESUMEN

Chimeric antigen receptor (CAR) engineering of natural killer (NK) cells has shown promising results in early-phase clinical studies. However, advancing CAR-NK cell therapeutic efficacy is imperative. In this study, we investigated the impact of a fourth-generation CD19-targeted CAR (CAR.19) coexpressing IL-27 on NK-92 cells. We observed a significant improvement in NK-92 cell proliferation and cytotoxicity activity against B-cell cancer cell lines, both in vitro and in a xenograft mouse B-cell lymphoma model. Our systematic transcriptome analysis of the activated NK-92 CAR variants further supports the potential of IL-27 in fourth-generation CARs to overcome limitations of NK cell-based targeted tumor therapies by providing essential growth and activation signals. Integrating IL-27 into CAR-NK cells emerges as a promising strategy to enhance their therapeutic potential and elicit robust responses against cancer cells. These findings contribute substantially to the mounting evidence supporting the potential of fourth-generation CAR engineering in advancing NK cell-based immunotherapies.

7.
BMC Cancer ; 24(1): 119, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38263004

RESUMEN

BACKGROUND: Adoptive cell therapy has achieved great success in treating hematological malignancies. However, the production of chimeric antigen receptor T (CAR-T) cell therapy still faces various difficulties. Natural killer (NK)-92 is a continuously expandable cell line and provides a promising alternative for patient's own immune cells. METHODS: We established CAR-NK cells by co-expressing natural killer group 2 member D (NKG2D) and IL-21, and evaluated the efficacy of NKG2D-IL-21 CAR-NK cells in treating lung cancer in vitro and in vivo. RESULTS: Our data suggested that the expression of IL-21 effectively increased the cytotoxicity of NKG2D CAR-NK cells against lung cancer cells in a dose-dependent manner and suppressed tumor growth in vitro and in vivo. In addition, the proliferation of NKG2D-IL-21 CAR-NK cells were enhanced while the apoptosis and exhaustion of these cells were suppressed. Mechanistically, IL-21-mediated NKG2D CAR-NK cells function by activating AKT signaling pathway. CONCLUSION: Our findings provide a novel option for treating lung cancer using NKG2D-IL-21 CAR-NK cell therapy.


Asunto(s)
Interleucinas , Neoplasias Pulmonares , Subfamilia K de Receptores Similares a Lectina de Células NK , Humanos , Inmunoterapia Adoptiva , Tratamiento Basado en Trasplante de Células y Tejidos
8.
Bull Math Biol ; 86(2): 20, 2024 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-38240892

RESUMEN

Chimeric antigen receptor (CAR)-engineered natural killer (NK) cells have recently emerged as a promising and safe alternative to CAR-T cells for targeting solid tumors. In the case of triple-negative breast cancer (TNBC), traditional cancer treatments and common immunotherapies have shown limited effectiveness. However, CAR-NK cells have been successfully employed to target epidermal growth factor receptor (EGFR) on TNBC cells, thereby enhancing the efficacy of immunotherapy. The effectiveness of CAR-NK-based immunotherapy is influenced by various factors, including the vaccination dose, vaccination pattern, and tumor immunosuppressive factors in the microenvironment. To gain insights into the dynamics and effects of CAR-NK-based immunotherapy, we propose a computational model based on experimental data and immunological theories. This model integrates an individual-based model that describes the interplay between the tumor and the immune system, along with an ordinary differential equation model that captures the variation of inflammatory cytokines. Computational results obtained from the proposed model shed light on the conditions necessary for initiating an effective anti-tumor response. Furthermore, global sensitivity analysis highlights the issue of low persistence of CAR-NK cells in vivo, which poses a significant challenge for the successful clinical application of these cells. Leveraging the model, we identify the optimal vaccination time, vaccination dose, and time interval between injections for maximizing therapeutic outcomes.


Asunto(s)
Receptores Quiméricos de Antígenos , Neoplasias de la Mama Triple Negativas , Humanos , Receptores Quiméricos de Antígenos/metabolismo , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/metabolismo , Conceptos Matemáticos , Modelos Biológicos , Células Asesinas Naturales , Simulación por Computador , Microambiente Tumoral
9.
Biochemistry (Mosc) ; 89(5): 765-783, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38880641

RESUMEN

Chimeric antigen receptors (CARs) are genetically engineered receptors that recognize antigens and activate signaling cascades in a cell. Signal recognition and transmission are mediated by the CAR domains derived from different proteins. T cells carrying CARs against tumor-associated antigens have been used in the development of the CAR T cell therapy, a new approach to fighting malignant neoplasms. Despite its high efficacy in the treatment of oncohematological diseases, CAR T cell therapy has a number of disadvantages that could be avoided by using other types of leukocytes as effector cells. CARs can be expressed in a wide range of cells of adaptive and innate immunity with the emergence or improvement of cytotoxic properties. This review discusses the features of CAR function in different types of immune cells, with a particular focus on the results of preclinical and clinical efficacy studies and the safety of potential CAR cell products.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias , Receptores Quiméricos de Antígenos , Linfocitos T , Humanos , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/genética , Inmunoterapia Adoptiva/métodos , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Antígenos de Neoplasias/inmunología
10.
Cryobiology ; 114: 104835, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38070820

RESUMEN

Cryopreservation is a crucial step in the supply process of off-the-shelf chimeric antigen receptor engineered natural killer (CAR-NK) cell products. Concerns have been raised over the clinical application of dimethyl sulfoxide (Me2SO) due to the potential for adverse reactions following infusion and limited cell-specific cytotoxic effects if misapplied. In this study, we developed a Me2SO-free cryopreservation medium specifically tailored for CAR-NK cells to address this limitation. The cryopreservation medium was formulated using human serum albumin (HSA) and glycerol as the base components. Following initial screening of seven clinically-compatible solutions, four with cryoprotective properties were identified. These were combined and optimized into a single formulation: IF-M. The viability, phenotype, and function of CAR-NK cells were evaluated after short-term and long-term cryopreservation to assess the effectiveness of IF-M, with Me2SO serving as the control group. The viability and recovery of CAR-NK cells in the IF-M group were significantly higher than those in the Me2SO group within 90 days of cryopreservation. Moreover, after 1 year of cryopreservation the cytotoxic capacity of CAR-NK cells cryopreserved with IF-M was comparable to that of fresh CAR-NK cells and significantly superior to that of CAR-NK cells cryopreserved in Me2SO. The CD107a expression intensity of CAR-NK cells in IF-M group was significantly higher than that of Me2SO group. No statistical differences were observed in other indicators under different cryopreservation times. These results underscore the robustness of IF-M as a suitable replacement for traditional Me2SO-based cryopreservation medium for the long-term cryopreservation and clinical application of off-the-shelf CAR-NK cells.


Asunto(s)
Criopreservación , Receptores Quiméricos de Antígenos , Humanos , Criopreservación/métodos , Crioprotectores/farmacología , Crioprotectores/metabolismo , Receptores Quiméricos de Antígenos/genética , Dimetilsulfóxido/farmacología , Dimetilsulfóxido/metabolismo , Células Asesinas Naturales , Supervivencia Celular
11.
Int J Mol Sci ; 25(11)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38891957

RESUMEN

Lymphocyte collection by apheresis for CAR-T production usually does not include blood mobilized using granulocyte colony stimulating factor (G-CSF) due to the widespread knowledge that it causes a decrease in the number and functionality of lymphocytes. However, it is used for stem cell transplant, which is a common treatment for hematological malignancies. The growing demand for CAR therapies (CAR-T and NK-CAR), both in research and clinics, makes it necessary to evaluate whether mobilized PBSC products may be potential candidates for use in such therapies. This review collects recent works that experimentally verify the role and functionality of T and NK lymphocytes and the generation of CAR-T from apheresis after G-CSF mobilization. As discussed, T cells do not vary significantly in their phenotype, the ratio of CD4+ and CD8+ remains constant, and the different sub-populations remain stable. In addition, the expansion and proliferation rates are invariant regardless of mobilization with G-CSF as well as the secretion of proinflammatory cytokines and the cytotoxic ability. Therefore, cells mobilized before apheresis are postulated as a new alternative source of T cells for adoptive therapies that will serve to alleviate high demand, increase availability, and take advantage of the substantial number of existing cryopreserved products.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Humanos , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Inmunoterapia Adoptiva/métodos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/inmunología , Movilización de Célula Madre Hematopoyética/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales
12.
Rinsho Ketsueki ; 65(7): 668-675, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-39098018

RESUMEN

Chimeric antigen receptor-transduced autologous T (CAR-T) cell therapy targeting CD19 has revolutionized the treatment of CD19-positive hematological tumors, including acute lymphoblastic leukemia and large B-cell lymphoma. However, despite the high response rate, many problems such as exceedingly high cost, complex logistics, insufficient speed, and manufacturing failures have become apparent. One solution for these problems is to use an allogeneic cell as an effector cell for genetic modification with CAR. Allogeneic, or "off-the-shelf", CAR-expressing immune-effector cells include 1) genome-edited, T-cell receptor (TCR) gene-deleted CAR-T cells generated using healthy adult donor T cells, 2) induced pluripotent stem cell-derived CAR-T cells, and 3) CAR NK cells. NK cells are notorious for their poor ex-vivo expansion and low susceptibility to genetic modification. In this article, I will review the current state and future prospects of allogeneic CAR cell therapies, with special reference to CAR NK cells.


Asunto(s)
Células Asesinas Naturales , Humanos , Células Asesinas Naturales/inmunología , Trasplante Homólogo , Receptores Quiméricos de Antígenos/inmunología , Inmunoterapia Adoptiva/métodos
13.
Chin J Cancer Res ; 36(1): 1-16, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38455373

RESUMEN

Chimeric antigen receptor-natural killer (CAR-NK) cells have emerged as another prominent player in the realm of tumor immunotherapy following CAR-T cells. The unique features of CAR-NK cells make it possible to compensate for deficiencies in CAR-T therapy, such as the complexity of the manufacturing process, clinical adverse events, and solid tumor challenges. To date, CAR-NK products from different allogeneic sources have exhibited remarkable anti-tumor effects on preclinical studies and have gradually been applied in clinical practice. However, each source has advantages and disadvantages. Selecting a suitable source may help maximize CAR-NK cell efficacy and increase the feasibility of clinical transformation. Therefore, this review discusses the development and challenges of CAR-NK cells from different sources to provide a reference for future exploration.

14.
Immunology ; 169(2): 204-218, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36640111

RESUMEN

Although the pre-clinical study of chimeric antigen receptor (CAR)-natural killer (NK) cell was effective against various tumours, immunosuppression mediated by tumour microenvironment hampers their application and several efforts have been explored to improve their effect in combating solid tumours. Glypican 3 (GPC3) is a promising target for hepatocellular carcinoma (HCC), and CAR-T cells targeting GPC3 have been tested in clinical trials. Based on an affinity-enhanced antibody (hYP7) targeting GPC3, we constructed GPC3-CAR-NK cells to explore their potential function in the treatment of HCC. We found that patients with HCC secreted high levels of soluble programmed death-ligand 1 (sPD-L1), which inhibits the function of CAR-NK cells targeting GPC3. In addition, we combined high-affinity sPD-L1 variant (L3C7c-Fc) with GPC3-CAR-NK cells to solve the problem of GPC3-CAR-NK inhibition. Our studies demonstrated that L3C7c-Fc could enhance the therapeutic effect of CAR-NK cells by reversing the suppression of sPD-L1, which provides the experimental evidence for the subsequent development of HCC immunotherapy strategies.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/genética , Glipicanos/genética , Células Asesinas Naturales , Microambiente Tumoral
15.
J Cell Biochem ; 124(8): 1082-1104, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37566723

RESUMEN

Natural killer (NK) cells are considered to be the foremost fighters of our innate immune system against foreign invaders and thus tend to promptly latch onto the virus-infected and tumor/cancerous cells, killing them through phagocytosis. At present, the application of genetically engineered Chimeric antigen receptor (CAR) receptors ensures a guaranteed optimistic response with NK cells and would not allow the affected cells to dodge or escape unchecked. Hence the specificity and uniqueness of CAR-NK cells over CAR-T therapy make them a better immunotherapeutic choice to reduce the load of trafficking of numerous tumor cells near the healthy cell populations in a more intact way than offered by CAR-T immunotherapy. Our review mainly focuses on the preclinical, clinical, and recent advances in clinical research trials and further strategies to achieve an augmented and efficient cure against solid tumors.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/genética , Células Asesinas Naturales , Neoplasias/patología , Inmunoterapia Adoptiva , Inmunoterapia
16.
Mol Cancer ; 22(1): 20, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36717905

RESUMEN

In the last decade, Chimeric Antigen Receptor (CAR)-T cell therapy has emerged as a promising immunotherapeutic approach to fight cancers. This approach consists of genetically engineered immune cells expressing a surface receptor, called CAR, that specifically targets antigens expressed on the surface of tumor cells. In hematological malignancies like leukemias, myeloma, and non-Hodgkin B-cell lymphomas, adoptive CAR-T cell therapy has shown efficacy in treating chemotherapy refractory patients. However, the value of this therapy remains inconclusive in the context of solid tumors and is restrained by several obstacles including limited tumor trafficking and infiltration, the presence of an immunosuppressive tumor microenvironment, as well as adverse events associated with such therapy. Recently, CAR-Natural Killer (CAR-NK) and CAR-macrophages (CAR-M) were introduced as a complement/alternative to CAR-T cell therapy for solid tumors. CAR-NK cells could be a favorable substitute for CAR-T cells since they do not require HLA compatibility and have limited toxicity. Additionally, CAR-NK cells might be generated in large scale from several sources which would suggest them as promising off-the-shelf product. CAR-M immunotherapy with its capabilities of phagocytosis, tumor-antigen presentation, and broad tumor infiltration, is currently being investigated. Here, we discuss the emerging role of CAR-T, CAR-NK, and CAR-M cells in solid tumors. We also highlight the advantages and drawbacks of CAR-NK and CAR-M cells compared to CAR-T cells. Finally, we suggest prospective solutions such as potential combination therapies to enhance the efficacy of CAR-cells immunotherapy.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T , Estudios Prospectivos , Neoplasias/patología , Inmunoterapia Adoptiva/efectos adversos , Tratamiento Basado en Trasplante de Células y Tejidos , Microambiente Tumoral
17.
Mol Cancer ; 22(1): 183, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37974170

RESUMEN

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.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Células Asesinas Naturales , Trogocitosis , Inmunoterapia Adoptiva , Linfocitos T , Receptores Quiméricos de Antígenos/metabolismo , Neoplasias/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos
18.
Curr Issues Mol Biol ; 46(1): 140-152, 2023 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-38248313

RESUMEN

The immune system plays a crucial role in recognizing and eliminating altered tumor cells. However, tumors develop mechanisms to evade the body's natural immune defenses. Therefore, methods for specifically recognizing/targeting tumor cells, for instance, through the activation, directed polarization, and training of immune cells, have been developed based on the body's immune cells. This strategy has been termed cellular immunotherapy. One promising strategy for treating tumor diseases is NK cell-based immunotherapy. NK cells have the ability to recognize and destroy transformed cells without prior activation as well as tumor cells with reduced MHC-I expression. A novel approach in immunotherapy is the use of extracellular vesicles (EVs) derived from NK cells. The main advantages of NK cell-derived EVs are their small size and better tissue penetration into a tumor. The aim of this review is to systematically present existing information on the mechanisms of antitumor immunity and the role of NK cells and extracellular vesicles in cancer immunotherapy. Clinical and preclinical studies utilizing NK cells and extracellular vesicles for anticancer therapy currently underway will provide valuable insights for researchers in the field of cancer.

19.
Curr Issues Mol Biol ; 45(11): 9181-9214, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37998753

RESUMEN

Over 100 innovative in vitro transcribed (IVT)-mRNAs are presently undergoing clinical trials, with a projected substantial impact on the pharmaceutical market in the near future. Τhe idea behind this is that after the successful cellular internalization of IVT-mRNAs, they are subsequently translated into proteins with therapeutic or prophylactic relevance. Simultaneously, cancer immunotherapy employs diverse strategies to mobilize the immune system in the battle against cancer. Therefore, in this review, the fundamental principles of IVT-mRNA to its recruitment in cancer immunotherapy, are discussed and analyzed. More specifically, this review paper focuses on the development of mRNA vaccines, the exploitation of neoantigens, as well as Chimeric Antigen Receptor (CAR) T-Cells, showcasing their clinical applications and the ongoing trials for the development of next-generation immunotherapeutics. Furthermore, this study investigates the synergistic potential of combining the CAR immunotherapy and the IVT-mRNAs by introducing our research group novel, patented delivery method that utilizes the Protein Transduction Domain (PTD) technology to transduce the IVT-mRNAs encoding the CAR of interest into the Natural Killer (NK)-92 cells, highlighting the potential for enhancing the CAR NK cell potency, efficiency, and bioenergetics. While IVT-mRNA technology brings exciting progress to cancer immunotherapy, several challenges and limitations must be acknowledged, such as safety, toxicity, and delivery issues. This comprehensive exploration of IVT-mRNA technology, in line with its applications in cancer therapeutics, offers valuable insights into the opportunities and challenges in the evolving landscape of cancer immunotherapy, setting the stage for future advancements in the field.

20.
Immunol Cell Biol ; 101(8): 705-726, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37282729

RESUMEN

Breast cancer (BC) is the most common cause of cancer death in women. According to the American Cancer Society's yearly cancer statistics, BC constituted almost 15% of all the newly diagnosed cancer cases in 2022 for both sexes. Metastatic disease occurs in 30% of patients with BC. The currently available treatments fail to cure metastatic BC, and the average survival time for patients with metastatic BC is approximately 2 years. Developing a treatment method that terminates cancer stem cells without harming healthy cells is the primary objective of novel therapeutics. Adoptive cell therapy is a branch of cancer immunotherapy that utilizes the immune cells to attack cancer cells. Natural killer (NK) cells are an essential component of innate immunity and are critical in destroying tumor cells without prior stimulation with antigens. With the advent of chimeric antigen receptors (CARs), the autologous or allogeneic use of NK/CAR-NK cell therapy has raised new hopes for treating patients with cancer. Here, we describe recent developments in NK and CAR-NK cell immunotherapy, including the biology and function of NK cells, clinical trials, different sources of NK cells and their future perspectives on BC.


Asunto(s)
Neoplasias de la Mama , Neoplasias , Receptores Quiméricos de Antígenos , Masculino , Humanos , Femenino , Inmunoterapia Adoptiva/métodos , Neoplasias de la Mama/terapia , Células Asesinas Naturales , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda