Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Bull Cancer ; 108(10S): S92-S95, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34920812

RESUMEN

Treatment of hematological malignancies by autologous T cells expressing a chimeric antigen receptor (CAR) is a breakthrough in the field of cancer immunotherapy. As CAR-T cells are entering advanced phases of clinical development, there is a need to develop universal, ready-to-use products using immune cells from healthy donors, to reduce time to treatment, improve response rate and finally reduce the cost of production. Mucosal-associated invariant T cells (MAIT) are unconventional T cells which recognize microbial-derived riboflavin derivatives presented by the conserved MR1 molecule and are endowed with potent effector functions. Because they are not selected by classical MHC/peptide complexes and express a semi-invariant T cell receptor, MAIT cells do not mediate alloreactivity, prompting their use as a new source of universal effector cells for allogeneic CAR-T cell therapy without the need to inactivate their endogenous TCR. We produced CD19-CAR MAIT cells as proof-of-concept allowing subsequent head-to-head comparison with currently used CD19-CAR T cells. We demonstrated their anti-tumor efficacy in vitro and their capacity to engraft without mediating GVHD in preclinical immunodeficient mouse models. Universal, off-the-shelf CAR-MAIT cells could provide a suitable alternative to current autologous CAR-T cells to treat patients regardless of HLA disparity, without production delay, enabling a cost-effective manufacturing model for large-scale clinical application.


Asunto(s)
Neoplasias Hematológicas/terapia , Inmunoterapia Adoptiva/métodos , Células T Invariantes Asociadas a Mucosa/trasplante , Receptores Quiméricos de Antígenos/inmunología , Animales , Antígenos CD19/inmunología , Análisis Costo-Beneficio , Neoplasias Hematológicas/inmunología , Ratones , Ratones SCID , Células T Invariantes Asociadas a Mucosa/citología , Células T Invariantes Asociadas a Mucosa/inmunología , Prueba de Estudio Conceptual
2.
Front Immunol ; 12: 782775, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790207

RESUMEN

Chimeric antigen receptor T (CAR-T) cell therapy has exhibited a substantial clinical response in hematological malignancies, including B-cell leukemia, lymphoma, and multiple myeloma. Therefore, the feasibility of using CAR-T cells to treat solid tumors is actively evaluated. Currently, multiple basic research projects and clinical trials are being conducted to treat lung cancer with CAR-T cell therapy. Although numerous advances in CAR-T cell therapy have been made in hematological tumors, the technology still entails considerable challenges in treating lung cancer, such as on-target, of-tumor toxicity, paucity of tumor-specific antigen targets, T cell exhaustion in the tumor microenvironment, and low infiltration level of immune cells into solid tumor niches, which are even more complicated than their application in hematological tumors. Thus, progress in the scientific understanding of tumor immunology and improvements in the manufacture of cell products are advancing the clinical translation of these important cellular immunotherapies. This review focused on the latest research progress of CAR-T cell therapy in lung cancer treatment and for the first time, demonstrated the underlying challenges and future engineering strategies for the clinical application of CAR-T cell therapy against lung cancer.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/terapia , Animales , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor , Técnicas de Cultivo de Célula , Ensayos Clínicos como Asunto , Terapia Combinada/métodos , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Humanos , Inmunomodulación , Inmunoterapia Adoptiva/efectos adversos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/mortalidad , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento
3.
Cell Rep Med ; 2(10): 100422, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34755134

RESUMEN

Provoked by sterile/nonsterile insults, prolonged monocyte mobilization and uncontrolled monocyte/macrophage activation can pose imminent or impending harm to the affected organs. Curiously, folate receptor beta (FRß), with subnanomolar affinity for the vitamin folic acid (FA), is upregulated during immune activation in hematopoietic cells of the myeloid lineage. This phenomenon has inspired a strong interest in exploring FRß-directed diagnostics/therapeutics. Previously, we have reported that FA-targeted aminopterin (AMT) therapy can modulate macrophage function and effectively treat animal models of inflammation. Our current investigation of a lead compound (EC2319) leads to discovery of a highly FR-specific mechanism of action independent of the root causes against inflammatory monocytes. We further show that EC2319 suppresses interleukin-6/interleukin-1ß release by FRß+ monocytes in a triple co-culture leukemic model of cytokine release syndrome with anti-CD19 chimeric antigen receptor T cells. Because of its chemical stability and metabolically activated linker, EC2319 demonstrates favorable pharmacokinetic characteristics and cross-species translatability to support future pre-clinical and clinical development.


Asunto(s)
Aminopterina/farmacología , Síndrome de Liberación de Citoquinas/prevención & control , Receptor 2 de Folato/genética , Antagonistas del Ácido Fólico/farmacología , Ácido Fólico/metabolismo , Macrófagos/efectos de los fármacos , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Células CHO , Cricetulus , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Femenino , Receptor 1 de Folato/antagonistas & inhibidores , Receptor 1 de Folato/genética , Receptor 1 de Folato/inmunología , Receptor 2 de Folato/antagonistas & inhibidores , Receptor 2 de Folato/inmunología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Modelos Biológicos , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/patología , Células RAW 264.7 , Ratas , Ratas Endogámicas Lew , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
4.
Front Immunol ; 12: 718686, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34386015

RESUMEN

In recent years, chimeric antigen receptor T cells (CAR-T cells) have been faced with the problems of weak proliferation and poor persistence in the treatment of some malignancies. Researchers have been trying to perfect the function of CAR-T by genetically modifying its structure. In addition to the participation of T cell receptor (TCR) and costimulatory signals, immune cytokines also exert a decisive role in the activation and proliferation of T cells. Therefore, genetic engineering strategies were used to generate cytokines to enhance tumor killing function of CAR-T cells. When CAR-T cells are in contact with target tumor tissue, the proliferation ability and persistence of T cells can be improved by structurally or inductively releasing immunoregulatory molecules to the tumor region. There are a large number of CAR-T cells studies on gene-edited cytokines, and the most common cytokines involved are interleukins (IL-7, IL-12, IL-15, IL-18, IL-21, IL-23). Methods for the construction of gene-edited interleukin CAR-T cells include co-expression of single interleukin, two interleukin, interleukin combined with other cytokines, interleukin receptors, interleukin subunits, and fusion inverted cytokine receptors (ICR). Preclinical and clinical trials have yielded positive results, and many more are under way. By reading a large number of literatures, we summarized the functional characteristics of some members of the interleukin family related to tumor immunotherapy, and described the research status of gene-edited interleukin CAR-T cells in the treatment of malignant tumors. The objective is to explore the optimized strategy of gene edited interleukin-CAR-T cell function.


Asunto(s)
Edición Génica , Inmunoterapia Adoptiva , Interleucinas/genética , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Ensayos Clínicos como Asunto , Citocinas/genética , Citocinas/metabolismo , Manejo de la Enfermedad , Evaluación Preclínica de Medicamentos , Edición Génica/métodos , Humanos , Inmunidad , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/tendencias , Interleucinas/metabolismo , Familia de Multigenes , Neoplasias/etiología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Resultado del Tratamiento
5.
Front Immunol ; 12: 693105, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34177953

RESUMEN

Regulatory T cells are essential players of peripheral tolerance and suppression of inflammatory immune responses. Type 1 regulatory T (Tr1) cells are FoxP3- regulatory T cells induced in the periphery under tolerogenic conditions. Tr1 cells are identified as LAG3+CD49b+ mature CD4+ T cells that promote peripheral tolerance through secretion of IL-10 and TGF-ß in addition to exerting perforin- and granzyme B-mediated cytotoxicity against myeloid cells. After the initial challenges of isolation were overcome by surface marker identification, ex vivo expansion of antigen-specific Tr1 cells in the presence of tolerogenic dendritic cells (DCs) and IL-10 paved the way for their use in clinical trials. With one Tr1-enriched cell therapy product already in a Phase I clinical trial in the context of allogeneic hematopoietic stem cell transplantation (allo-HSCT), Tr1 cell therapy demonstrates promising results so far in terms of efficacy and safety. In the current review, we identify developments in phenotypic and molecular characterization of Tr1 cells and discuss the potential of engineered Tr1-like cells for clinical applications of Tr1 cell therapies. More than 3 decades after their initial discovery, Tr1 cell therapy is now being used to prevent graft versus host disease (GvHD) in allo-HSCT and will be an alternative to immunosuppression to promote graft tolerance in solid organ transplantation in the near future.


Asunto(s)
Alergia e Inmunología , Investigación Biomédica , Linfocitos T Reguladores/inmunología , Animales , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Inmunoterapia Adoptiva , Fenotipo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante
6.
Int J Oncol ; 59(1)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34132378

RESUMEN

Anaplastic large cell lymphoma (ALCL) is a rare and highly invasive non­Hodgkin's lymphoma. In the past few decades, traditional chemotherapy regimens, such as as the cyclophosphamide, vincristine, doxorubicin and prednisone regimen, have been recommended for first­line treatment. In order to improve the survival of patients, dose­intensive chemotherapy and hematopoietic stem cell transplantation have been deeply studied and some progress has been made. Recently, with the accumulation of clinical cases and the development of clinical trials, as well improvements to our in­depth understanding of the biological behavior of ALCL, the signaling pathways and the immunotherapy involved, research on this topic is in full swing. The emergence of several targeted drugs and immunotherapies, including anaplastic lymphoma kinase inhibitors, brentuximab vedotin, mTOR inhibitors, programmed cell death protein 1/programmed death ligand 1 inhibitors and chimeric antigen receptor­T cell therapy, seems to provide new opportunities for certain patients with ALCL. The present review focuses on the current use of traditional therapy and the treatment prospects of these new drugs in ALCL.


Asunto(s)
Linfoma Anaplásico de Células Grandes/terapia , Quimioterapia , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoterapia Adoptiva , Linfoma Anaplásico de Células Grandes/inmunología , Receptores Quiméricos de Antígenos/inmunología , Transducción de Señal/efectos de los fármacos
7.
Int J Mol Sci ; 22(11)2021 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-34072732

RESUMEN

CAR-T (chimeric antigen receptor T) cells have emerged as a milestone in the treatment of patients with refractory B-cell neoplasms. However, despite having unprecedented efficacy against hematological malignancies, the treatment is far from flawless. Its greatest drawbacks arise from a challenging and expensive production process, strict patient eligibility criteria and serious toxicity profile. One possible solution, supported by robust research, is the replacement of T lymphocytes with NK cells for CAR expression. NK cells seem to be an attractive vehicle for CAR expression as they can be derived from multiple sources and safely infused regardless of donor-patient matching, which greatly reduces the cost of the treatment. CAR-NK cells are known to be effective against hematological malignancies, and a growing number of preclinical findings indicate that they have activity against non-hematological neoplasms. Here, we present a thorough overview of the current state of knowledge regarding the use of CAR-NK cells in treating various solid tumors.


Asunto(s)
Inmunoterapia Adoptiva , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Técnicas de Cultivo de Célula , Ensayos Clínicos como Asunto , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Humanos , Células Asesinas Naturales/metabolismo , Neoplasias/diagnóstico , Neoplasias/etiología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Resultado del Tratamiento
8.
ACS Synth Biol ; 10(5): 1176-1183, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33856201

RESUMEN

Various antibody-redirected immunotherapeutic approaches, including antibody-drug conjugates (ADCs), bispecific antibodies (bsAbs), and chimeric antigen receptor-T (CAR-T) cells, have been devised to produce specific activity against various cancer types. Using genetically encoded unnatural amino acids, we generated a homogeneous Her2-targeted ADC, a T cell-redirected bsAb, and a FITC-modified antibody capable of redirecting anti-FITC CAR-T (switchable CAR-T; sCAR-T) cells to target different Her2-expressing breast cancers. sCAR-T cells showed activity against Her2-expressing tumor cells comparable to that of conventional anti-Her2 CAR-T cells and superior to that of ADC- and bsAb-based approaches. To prevent antigen escape, we designed bispecific sCAR-T cells targeting both the Her2 receptor and IGF1R, which showed an overall improved activity against cancer cells with low Her2 expression. This study increases our understanding of various explored cancer therapeutics and underscores the efficient application of sCAR-T cells as a promising therapeutic option for breast cancer patients with low or heterogeneous antigen expression.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Neoplasias de la Mama/metabolismo , Inmunoconjugados/inmunología , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 1/inmunología , Receptor IGF Tipo 1/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Aminoácidos/genética , Deriva y Cambio Antigénico/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Femenino , Fluoresceína-5-Isotiocianato , Humanos , Inmunoterapia Adoptiva/métodos , Terapia Molecular Dirigida/métodos
9.
Cancer Discov ; 11(9): 2186-2199, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33820778

RESUMEN

The adoptive transfer of chimeric antigen receptor (CAR) T cells represents a breakthrough in clinical oncology, yet both between- and within-patient differences in autologously derived T cells are a major contributor to therapy failure. To interrogate the molecular determinants of clinical CAR T-cell persistence, we extensively characterized the premanufacture T cells of 71 patients with B-cell malignancies on trial to receive anti-CD19 CAR T-cell therapy. We performed RNA-sequencing analysis on sorted T-cell subsets from all 71 patients, followed by paired Cellular Indexing of Transcriptomes and Epitopes (CITE) sequencing and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) on T cells from six of these patients. We found that chronic IFN signaling regulated by IRF7 was associated with poor CAR T-cell persistence across T-cell subsets, and that the TCF7 regulon not only associates with the favorable naïve T-cell state, but is maintained in effector T cells among patients with long-term CAR T-cell persistence. These findings provide key insights into the underlying molecular determinants of clinical CAR T-cell function. SIGNIFICANCE: To improve clinical outcomes for CAR T-cell therapy, there is a need to understand the molecular determinants of CAR T-cell persistence. These data represent the largest clinically annotated molecular atlas in CAR T-cell therapy to date, and significantly advance our understanding of the mechanisms underlying therapeutic efficacy.This article is highlighted in the In This Issue feature, p. 2113.


Asunto(s)
Inmunoterapia Adoptiva , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/trasplante , Adolescente , Niño , Supervivencia sin Enfermedad , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/mortalidad , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Philadelphia , Linfocitos T/inmunología
10.
Front Immunol ; 12: 640082, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746981

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy has emerged as one of the major breakthroughs in cancer immunotherapy in the last decade. Outstanding results in hematological malignancies and encouraging pre-clinical anti-tumor activity against a wide range of solid tumors have made CAR T cells one of the most promising fields for cancer therapies. CAR T cell therapy is currently being investigated in solid tumors including glioblastoma (GBM), a tumor for which survival has only modestly improved over the past decades. CAR T cells targeting EGFRvIII, Her2, or IL-13Rα2 have been tested in GBM, but the first clinical trials have shown modest results, potentially due to GBM heterogeneity and to the presence of an immunosuppressive microenvironment. Until now, the use of autologous T cells to manufacture CAR products has been the norm, but this approach has several disadvantages regarding production time, cost, manufacturing delay and dependence on functional fitness of patient T cells, often reduced by the disease or previous therapies. Universal "off-the-shelf," or allogeneic, CAR T cells is an alternative that can potentially overcome these issues, and allow for multiple modifications and CAR combinations to target multiple tumor antigens and avoid tumor escape. Advances in genome editing tools, especially via CRISPR/Cas9, might allow overcoming the two main limitations of allogeneic CAR T cells product, i.e., graft-vs.-host disease and host allorejection. Here, we will discuss how allogeneic CAR T cells could allow for multivalent approaches and alteration of the tumor microenvironment, potentially allowing the development of next generation therapies for the treatment of patients with GBM.


Asunto(s)
Células Alogénicas/inmunología , Glioblastoma/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Humanos , Microambiente Tumoral/inmunología
11.
Cancer Discov ; 11(3): 560-574, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33563662

RESUMEN

Adoptive cell therapy (ACT) for cancer shows tremendous potential; however, several challenges preclude its widespread use. These include poor T-cell function in hostile tumor microenvironments, a lack of tumor-specific target antigens, and the high cost and poor scalability of cell therapy manufacturing. Creative genome-editing strategies are beginning to emerge to address each of these limitations, which has initiated the next generation of cell therapy products now entering clinical trials. CRISPR is at the forefront of this revolution, offering a simple and versatile platform for genetic engineering. This review provides a comprehensive overview of CRISPR applications that have advanced ACT. SIGNIFICANCE: The clinical impact of ACT for cancer can be expanded by implementing specific genetic modifications that enhance the potency, safety, and scalability of cellular products. Here we provide a detailed description of such genetic modifications, highlighting avenues to enhance the therapeutic efficacy and accessibility of ACT for cancer. Furthermore, we review high-throughput CRISPR genetic screens that have unveiled novel targets for cell therapy enhancement.


Asunto(s)
Sistemas CRISPR-Cas , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Edición Génica/métodos , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Ensayos Clínicos como Asunto , Terapia Combinada , Manejo de la Enfermedad , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Terapia Genética , Humanos , Inmunoterapia Adoptiva/efectos adversos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento
12.
Clin Drug Investig ; 41(3): 201-210, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33598857

RESUMEN

BACKGROUND AND OBJECTIVE: Registrational trials for ciltacabtagene autoleucel [cilta-cel]) and idecabtagene vicleucel [ide-cel] chimeric antigen receptor T-cell (CAR-T) therapies were single-arm studies conducted with relapse refractory multiple myeloma (MM) patients who were triple-class-exposed (TCE) or triple-class-refractory (TCR). It is critical for researchers conducting comparative effectiveness research (CER) to carefully consider the most appropriate data sources and comparable patient populations. The aim of this study was to identify potential data sources and populations for comparing to single-arm CAR-T trials CARTITUDE-1 (cilta-cel) and KarMMa (ide-cel). METHODS: A 2-part global systematic literature search produced a review of (1) clinical trials of National Comprehensive Cancer Network (NCCN) guideline preferred regimens in previously treated MM, and (2) real-world data cohorts of TCE or TCR populations, published between 1/1/2015 and 12/10/2020, with sample sizes of > 50 patients and reporting survival-related outcomes. Implications on CER and accepted best practices are discussed. RESULTS: Nine clinical trials of NCCN preferred regimens were identified along with five real-world data-based publications. No clinical trials evaluated patients with TCE or TCR MM. Among the real-world data-based publications, two evaluated patients exclusively with TCR MM, two analyzed a mixed population of patients with TCE or TCR MM, and one publication assessed patients exclusively with TCE MM. Real-world data treatment patterns were heterogeneous. CONCLUSION: Current NCCN preferred regimens were not specifically studied in TCE or TCR MM patients, although some studies do include a proportion of these types of patients. Therefore, appropriate matching of populations using either real-world data or patient level clinical trial data is critical to putting trials of novel CAR-Ts (i.e., CARTITUDE-1 or KarMMa) into appropriate comparative context.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Mieloma Múltiple/terapia , Receptores Quiméricos de Antígenos/inmunología , Investigación sobre la Eficacia Comparativa , Humanos , Almacenamiento y Recuperación de la Información
13.
Int J Hematol ; 114(5): 572-579, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32705572

RESUMEN

The use of allogeneic, pluripotent stem cell-derived immune cells for cancer immunotherapy has been the subject of recent research, including clinical trials. The use of pluripotent stem cells as the source for allogeneic immune cells facilitates stringent quality control of the final product, regarding efficacy, safety, and producibility. In this review, we have described the characteristics of natural killer (NK) cells from multiple cell sources, including pluripotent stem cells, the chimeric antigen receptor (CAR)-modification method and strategy for these NK cells, and the current and planned clinical trials of CAR-modified induced pluripotent stem cell-derived NK cells.


Asunto(s)
Expresión Génica , Inmunoterapia Adoptiva , Células Madre Pluripotentes Inducidas/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias/terapia , Receptores Quiméricos de Antígenos/genética , Animales , Antígenos de Neoplasias/inmunología , Estudios Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Células Asesinas Naturales/citología , Neoplasias/etiología , Pronóstico , Receptores Quiméricos de Antígenos/inmunología , Resultado del Tratamiento
14.
Blood Rev ; 45: 100695, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32402724

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy is currently approved for treatment of refractory B-cell malignancies. Response rates in these diseases are impressive by historical standards, but most patients do not have a durable response and there remains room for improvement. To date, CAR T cell activity has been even more limited in solid malignancies. These limitations are thought to be due to several pathways of resistance to CAR T cells, including cell-intrinsic mechanisms and the immunosuppressive tumor microenvironment. In this review, we discuss current experimental strategies that combine small molecules and monoclonal antibodies with CAR T cells to overcome these resistance mechanisms. We describe the biological rationale, pre-clinical data and clinical trials in progress that test the efficacy and safety of these combinations.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Ensayos Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Humanos , Inmunomodulación , Inmunoterapia Adoptiva/métodos , Neoplasias/etiología , Neoplasias/patología , Receptores de Antígenos de Linfocitos T/genética , Resultado del Tratamiento , Microambiente Tumoral/inmunología
15.
Leukemia ; 35(3): 752-763, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32632095

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy remains limited to select centers that can carefully monitor adverse events. To broaden use of CAR T cells in community clinics and in a frontline setting, we developed a novel CD8+ CAR T-cell product, Descartes-08, with predictable pharmacokinetics for treatment of multiple myeloma. Descartes-08 is engineered by mRNA transfection to express anti-BCMA CAR for a defined length of time. Descartes-08 expresses anti-BCMA CAR for 1 week, limiting risk of uncontrolled proliferation; produce inflammatory cytokines in response to myeloma target cells; and are highly cytolytic against myeloma cells regardless of the presence of myeloma-protecting bone marrow stromal cells, exogenous a proliferation-inducing ligand, or drug resistance including IMiDs. The magnitude of cytolysis correlates with anti-BCMA CAR expression duration, indicating a temporal limit in activity. In the mouse model of aggressive disseminated human myeloma, Descartes-08 induces BCMA CAR-specific myeloma growth inhibition and significantly prolongs host survival (p < 0.0001). These preclinical data, coupled with an ongoing clinical trial of Descartes-08 in relapsed/refractory myeloma (NCT03448978) showing preliminary durable responses and a favorable therapeutic index, have provided the framework for a recently initiated trial of an optimized/humanized version of Descartes-08 (i.e., Descartes-11) in newly diagnosed myeloma patients with residual disease after induction therapy.


Asunto(s)
Antígeno de Maduración de Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva/métodos , Mieloma Múltiple/terapia , ARN Mensajero/genética , Receptores Quiméricos de Antígenos/inmunología , Animales , Apoptosis , Antígeno de Maduración de Linfocitos B/genética , Proliferación Celular , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Leukemia ; 35(6): 1563-1570, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33077866

RESUMEN

Safety and efficacy of allogeneic anti-CD19 chimeric antigen receptor T cells (CAR-T cells) in persons with CD19-positive B-cell acute lymphoblastic leukemia (B-ALL) relapsing after an allotransplant remain unclear. Forty-three subjects with B-ALL relapsing post allotransplant received CAR-T cells were analyzed. 34 (79%; 95% confidence interval [CI]: 66, 92%) achieved complete histological remission (CR). Cytokine release syndrome (CRS) occurred in 38 (88%; 78, 98%) and was ≥grade-3 in 7. Two subjects died from multiorgan failure and CRS. Nine subjects (21%; 8, 34%) developed ≤grade-2 immune effector cell-associated neurotoxicity syndrome (ICANS). Two subjects developed ≤grade-2 acute graft-versus-host disease (GvHD). 1-year event-free survival (EFS) and survival was 43% (25, 62%). In 32 subjects with a complete histological remission without a second transplant, 1-year cumulative incidence of relapse was 41% (25, 62%) and 1-year EFS and survival, 59% (37, 81%). Therapy of B-ALL subjects relapsing post transplant with donor-derived CAR-T cells is safe and effective but associated with a high rate of CRS. Outcomes seem comparable to those achieved with alternative therapies but data from a randomized trial are lacking.


Asunto(s)
Antígenos CD19/metabolismo , Trasplante de Células Madre Hematopoyéticas/mortalidad , Inmunoterapia Adoptiva/métodos , Recurrencia Local de Neoplasia/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Pronóstico , Receptores Quiméricos de Antígenos/inmunología , Estudios Retrospectivos , Tasa de Supervivencia , Donantes de Tejidos , Trasplante Homólogo , Adulto Joven
17.
Int Rev Immunol ; 39(5): 223-232, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32546036

RESUMEN

One of the cutting edge techniques for treating cancer is the use of the patient's immune system to prevail cancerous disease. The versatility of the chimeric antigen receptor (CAR) T-cell approach in conjugation with promising treatments in haematological cancer has led to countless cases of research literature for the treatment of solid cancer. A systematic search of online databases as well as gray literature and reference lists of retrieved studies were carried out up to March 2019 to identify experimental animal studies that investigated the antigens targeted by CAR T-cell for pancreatic cancer treatment. Studies were evaluated for methodological quality using the SYstematic Review Center for Laboratory Animal Experimentation bias risk tool (SYRCLE's ROB tool). Pooled cytotoxicity ratio/percentage and 95% confidence intervals were calculated using the inverse-variance method while random-effects meta-analysis was used, taking into account conceptual heterogeneity. Heterogeneity was assessed with the Cochran Q statistic and quantified with the I2 statistic using Stata 13.0. Of the 485 identified studies, 56 were reviewed in-depth with 16 preclinical animal studies eligible for inclusion in the systematic review and 11 studies included in our meta-analysis. CAR immunotherapy significantly increased the cytotoxicity assay (percentage: 65%; 95% CI: 46%, 82%). There were no evidence for significant heterogeneity across studies [P = 0.38 (Q statistics), I2 = 7.14%] and for publication bias. The quality assessment of included studies revealed that the evidence was moderate to low quality and none of studies was judged as having a low risk of bias across all domains. CAR T-cell therapy is effective for pancreatic cancer treatment in preclinical animal studies. Further high-quality studies are needed to confirm our finding and a standard approach of this type of studies is necessary according to our assessment.


Asunto(s)
Biomarcadores de Tumor , Inmunoterapia Adoptiva/métodos , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Inmunoterapia Adoptiva/efectos adversos , Linfocitos T/metabolismo
18.
Scand J Immunol ; 92(4): e12917, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32557659

RESUMEN

Ovarian Cancer (OC) is currently difficult to cure, mainly due to its late detection and the advanced state of the disease at the time of diagnosis. Therefore, conventional treatments such as debulking surgery and combination chemotherapy are rarely able to control progression of the tumour, and relapses are frequent. Alternative therapies are currently being evaluated, including immunotherapy and advanced T cell-based therapy. In the present review, we will focus on a description of those Chimeric Antigen Receptors (CARs) that have been validated in the laboratory or are being tested in the clinic. Numerous target antigens have been defined due to the identification of OC biomarkers, and many are being used as CAR targets. We provide an exhaustive list of these constructs and their current status. Despite being innovative and efficient, the OC-specific CARs face a barrier to their clinical efficacy: the tumour microenvironment (TME). Indeed, effector cells expressing CARs have been shown to be severely inhibited, rendering the CAR T cells useless once at the tumour site. Herein, we give a thorough description of the highly immunosuppressive OC TME and present recent studies and innovations that have enabled CAR T cells to counteract this negative environment and to destroy tumours.


Asunto(s)
Carcinoma Epitelial de Ovario/inmunología , Neoplasias Ováricas/inmunología , Receptores Quiméricos de Antígenos/inmunología , Microambiente Tumoral/inmunología , Animales , Antígenos de Neoplasias/inmunología , Carcinoma Epitelial de Ovario/terapia , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Neoplasias Ováricas/terapia , Receptores Quiméricos de Antígenos/uso terapéutico , Escape del Tumor/inmunología
19.
Drug Resist Updat ; 51: 100702, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32371296

RESUMEN

The tyrosine kinase inhibitor (TKI) sorafenib continues to be the anchor drug in the treatment of advanced stage hepatocellular carcinoma (HCC). Other TKIs as well as immune checkpoint inhibitors (ICIs) have also been approved, however the response rates remain poor and heterogeneous among HCC patients, largely due to antitumor drug resistance. Studies aimed at identifying novel biomarkers and developing new strategies to improve the response to current treatment and to overcome drug resistance, are urgently needed. Germline or somatic mutations, neoantigens, and an immunotolerogenic state against constant inflammatory stimuli in the liver, are crucial for the anti-tumor response. A pharmacogenetic approach has been attempted considering germline polymorphisms in genes encoding for proteins involved in drug-targeted pathways. Single gene and comprehensive multi-gene somatic profiling approaches have been adopted in HCC to identify tumor sensitivity scores and immunogenic profiles that can be exploited for new biomarkers and innovative therapeutic approaches. However, the high genomic heterogeneity of tumors and lack of molecularly targeted agents, hamper the discovery of specific molecular markers of resistance to therapy. Adoptive cell therapy with chimeric antigen receptor redirected T (CAR-T) cells targeting specific tumor-associated antigens (TAAs) was proposed recently. The specificity of the chosen TAA, an efficient homing of CAR-T cells to the tumor site, and the ability of CAR-T cells to survive in the tumor microenvironment are central factors in the success of CAR-T therapy. The current review describes the principal systemic treatments for HCC and the molecular evidence regarding potential predictive host and somatic genetic markers, as well as the emerging strategy of liquid biopsy for disease monitoring. Novel immunotherapeutic approaches for HCC treatment, including the use of ICIs and CAR-T, as well as strategies to overcome drug resistance, are discussed.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Inmunoterapia Adoptiva/métodos , Neoplasias Hepáticas/tratamiento farmacológico , Receptores Quiméricos de Antígenos/uso terapéutico , Sorafenib/uso terapéutico , Inhibidores de la Angiogénesis/farmacología , Antígenos de Neoplasias/efectos de los fármacos , Antígenos de Neoplasias/inmunología , Antineoplásicos/inmunología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Citocinas/inmunología , Resistencia a Antineoplásicos/inmunología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Estadificación de Neoplasias , Farmacogenética , Inhibidores de Proteínas Quinasas/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Radioterapia Adyuvante/métodos , Receptores Quiméricos de Antígenos/inmunología , Sorafenib/inmunología , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología
20.
J Immunol Res ; 2020: 8459496, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32411806

RESUMEN

NK cells are lymphocytes with antitumor properties and can directly lyse tumor cells in a non-MHC-restricted manner. However, the tumor microenvironment affects the immune function of NK cells, which leads to immune evasion. This may be related to the pathogenesis of some diseases. Therefore, great efforts have been made to improve the immunotherapy effect of natural killer cells. NK cells from different sources can meet different clinical needs, in order to minimize the inhibition of NK cells and maximize the response potential of NK cells, for example, modification of NK cells can increase the number of NK cells in tumor target area, change the direction of NK cells, and improve their targeting ability to malignant cells. Checkpoint blocking is also a promising strategy for NK cells to kill tumor cells. Combination therapy is another strategy for improving antitumor ability, especially in combination with oncolytic viruses and nanomaterials. In this paper, the mechanisms affecting the activity of NK cells were reviewed, and the therapeutic potential of different basic NK cell strategies in tumor therapy was focused on. The main strategies for improving the immune function of NK cells were described, and some new strategies were proposed.


Asunto(s)
Inmunoterapia/métodos , Células Asesinas Naturales/trasplante , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/administración & dosificación , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Evasión Inmune/efectos de los fármacos , Memoria Inmunológica , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de la radiación , Magnetoterapia , Ratones , Nanomedicina/métodos , Nanopartículas/administración & dosificación , Neoplasias/inmunología , Virus Oncolíticos/inmunología , Receptores Quiméricos de Antígenos/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA