Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Mol Ther ; 32(6): 1849-1874, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38584391

RESUMEN

The clinical potential of current FDA-approved chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy is encumbered by its autologous nature, which presents notable challenges related to manufacturing complexities, heightened costs, and limitations in patient selection. Therefore, there is a growing demand for off-the-shelf universal cell therapies. In this study, we have generated universal CAR-engineered NKT (UCAR-NKT) cells by integrating iNKT TCR engineering and HLA gene editing on hematopoietic stem cells (HSCs), along with an ex vivo, feeder-free HSC differentiation culture. The UCAR-NKT cells are produced with high yield, purity, and robustness, and they display a stable HLA-ablated phenotype that enables resistance to host cell-mediated allorejection. These UCAR-NKT cells exhibit potent antitumor efficacy to blood cancers and solid tumors, both in vitro and in vivo, employing a multifaceted array of tumor-targeting mechanisms. These cells are further capable of altering the tumor microenvironment by selectively depleting immunosuppressive tumor-associated macrophages and myeloid-derived suppressor cells. In addition, UCAR-NKT cells demonstrate a favorable safety profile with low risks of graft-versus-host disease and cytokine release syndrome. Collectively, these preclinical studies underscore the feasibility and significant therapeutic potential of UCAR-NKT cell products and lay a foundation for their translational and clinical development.


Asunto(s)
Células Madre Hematopoyéticas , Inmunoterapia Adoptiva , Células T Asesinas Naturales , Receptores Quiméricos de Antígenos , Humanos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Animales , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Inmunoterapia Adoptiva/métodos , Ratones , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Edición Génica , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias/terapia , Neoplasias/inmunología , Línea Celular Tumoral , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
2.
J Biomed Sci ; 31(1): 5, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38217016

RESUMEN

Cell-based immunotherapies (CBIs), notably exemplified by chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy, have emerged as groundbreaking approaches for cancer therapy. Nevertheless, akin to various other therapeutic modalities, tumor cells employ counterstrategies to manifest immune evasion, thereby circumventing the impact of CBIs. This phenomenon is facilitated by an intricately immunosuppression entrenched within the tumor microenvironment (TME). Principal mechanisms underpinning tumor immune evasion from CBIs encompass loss of antigens, downregulation of antigen presentation, activation of immune checkpoint pathways, initiation of anti-apoptotic cascades, and induction of immune dysfunction and exhaustion. In this review, we delve into the intrinsic mechanisms underlying the capacity of tumor cells to resist CBIs and proffer prospective stratagems to navigate around these challenges.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/metabolismo , Evasión Inmune , Estudios Prospectivos , Inmunoterapia Adoptiva , Inmunoterapia , Microambiente Tumoral , Neoplasias/terapia
3.
Mol Ther ; 31(3): 631-646, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36463401

RESUMEN

Human mucosal-associated invariant T (MAIT) cells are characterized by their expression of an invariant TCR α chain Vα7.2-Jα33/Jα20/Jα12 paired with a restricted TCR ß chain. MAIT cells recognize microbial peptides presented by the highly conserved MHC class I-like molecule MR1 and bridge the innate and acquired immune systems to mediate augmented immune responses. Upon activation, MAIT cells rapidly proliferate, produce a variety of cytokines and cytotoxic molecules, and trigger efficient antitumor immunity. Administration of a representative MAIT cell ligand 5-OP-RU effectively activates MAIT cells and enhances their antitumor capacity. In this review, we introduce MAIT cell biology and their importance in antitumor immunity, summarize the current development of peripheral blood mononuclear cell-derived and stem cell-derived MAIT cell products for cancer treatment, and discuss the potential of genetic engineering of MAIT cells for off-the-shelf cancer immunotherapy.


Asunto(s)
Células T Invariantes Asociadas a Mucosa , Neoplasias , Humanos , Células T Invariantes Asociadas a Mucosa/metabolismo , Leucocitos Mononucleares/metabolismo , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Inmunoterapia , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/metabolismo
4.
J Transl Med ; 21(1): 686, 2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37784157

RESUMEN

Cancer stem cells (CSCs) have emerged as key contributors to tumor initiation, growth, and metastasis. In addition, CSCs play a significant role in inducing immune evasion, thereby compromising the effectiveness of cancer treatments. The reciprocal communication between CSCs and the tumor microenvironment (TME) is observed, with the TME providing a supportive niche for CSC survival and self-renewal, while CSCs, in turn, influence the polarization and persistence of the TME, promoting an immunosuppressive state. Consequently, these interactions hinder the efficacy of current cancer therapies, necessitating the exploration of novel therapeutic approaches to modulate the TME and target CSCs. In this review, we highlight the intricate strategies employed by CSCs to evade immune surveillance and develop resistance to therapies. Furthermore, we examine the dynamic interplay between CSCs and the TME, shedding light on how this interaction impacts cancer progression. Moreover, we provide an overview of advanced therapeutic strategies that specifically target CSCs and the TME, which hold promise for future clinical and translational studies in cancer treatment.


Asunto(s)
Neoplasias , Humanos , Neoplasias/patología , Inmunoterapia , Transformación Celular Neoplásica , Células Madre Neoplásicas/patología , Microambiente Tumoral
5.
Int J Mol Sci ; 24(4)2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36835495

RESUMEN

Allogeneic cell therapies, defined by genetically mismatched transplantation, have the potential to become a cost-effective solution for cell-based cancer immunotherapy. However, this type of therapy is often accompanied by the development of graft-versus-host disease (GvHD), induced by the mismatched major histocompatibility complex (MHC) between healthy donors and recipients, leading to severe complications and death. To address this issue and increase the potential for allogeneic cell therapies in clinical practice, minimizing GvHD is a crucial challenge. Innate T cells, encompassing subsets of T lymphocytes including mucosal-associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells, and gamma delta T (γδ T) cells, offer a promising solution. These cells express MHC-independent T-cell receptors (TCRs), allowing them to avoid MHC recognition and thus GvHD. This review examines the biology of these three innate T-cell populations, evaluates research on their roles in GvHD modulation and allogeneic stem cell transplantation (allo HSCT), and explores the potential futures for these therapies.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Células T Invariantes Asociadas a Mucosa , Células T Asesinas Naturales , Humanos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Enfermedad Injerto contra Huésped/etiología , Inmunoterapia/efectos adversos
6.
Int J Mol Sci ; 23(14)2022 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-35886891

RESUMEN

Invariant natural killer T (iNKT) cells have the capacity to mount potent anti-tumor reactivity and have therefore become a focus in the development of cell-based immunotherapy. iNKT cells attack tumor cells using multiple mechanisms with a high efficacy; however, their clinical application has been limited because of their low numbers in cancer patients and difficulties in infiltrating solid tumors. In this study, we aimed to overcome these critical limitations by using α-GalCer, a synthetic glycolipid ligand specifically activating iNKT cells, to recruit iNKT to solid tumors. By adoptively transferring human iNKT cells into tumor-bearing humanized NSG mice and administering a single dose of tumor-localized α-GalCer, we demonstrated the rapid recruitment of human iNKT cells into solid tumors in as little as one day and a significantly enhanced tumor killing ability. Using firefly luciferase-labeled iNKT cells, we monitored the tissue biodistribution and pharmacokinetics/pharmacodynamics (PK/PD) of human iNKT cells in tumor-bearing NSG mice. Collectively, these preclinical studies demonstrate the promise of an αGC-driven iNKT cell-based immunotherapy to target solid tumors with higher efficacy and precision.


Asunto(s)
Células T Asesinas Naturales , Neoplasias , Animales , Antígenos CD1d , Galactosilceramidas/farmacología , Humanos , Ratones , Neoplasias/terapia , Distribución Tisular
7.
Mol Biol Rep ; 46(1): 1499-1510, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30554315

RESUMEN

A rapidly growing area of interest in biomedical science involves the reciprocal crosstalk between the sensory nervous and immune systems. Both of these systems are highly integrated, detecting potential environmental harms and restoring homeostasis. Many different cytokines, receptors, neuropeptides, and other proteins are involved in this bidirectional communication that are common to both systems. One such family of proteins includes the transient receptor potential vanilloid (TRPV) proteins. Though much progress has been made in understanding TRPV proteins in the nervous system, their functions in the immune system are not well elucidated. Hence, further understanding their role in the peripheral immune system and as regulators of neuroimmunity is critical for evaluating their potential as therapeutic targets for numerous inflammatory disorders, cancers, and other disease states. Here, we focus on the latest advancements in understanding TRPV1 and TRPV2's roles in the immune system, TRPV1 in neuroimmunity, and TRPV1's potential involvement in anti-tumor therapy.


Asunto(s)
Sistema Nervioso/inmunología , Canales Catiónicos TRPV/metabolismo , Animales , Humanos , Sistema Inmunológico/metabolismo , Modelos Biológicos , Neoplasias/inmunología
8.
Mol Biol Rep ; 45(1): 39-55, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29282598

RESUMEN

Over 10% of genetic diseases are caused by mutations that introduce a premature termination codon in protein-coding mRNA. Nonsense-mediated mRNA decay (NMD) is an essential cellular pathway that degrades these mRNAs to prevent the accumulation of harmful partial protein products. NMD machinery is also increasingly appreciated to play a role in other essential cellular functions, including telomere homeostasis and the regulation of normal mRNA turnover, and is misregulated in numerous cancers. Hence, understanding and designing therapeutics targeting NMD is an important goal in biomedical science. The central regulator of NMD, the Upf1 protein, interacts with translation termination factors and contextual factors to initiate NMD specifically on mRNAs containing PTCs. The molecular details of how these contextual factors affect Upf1 function remain poorly understood. Here, we review plausible models for the NMD pathway and the evidence for the variety of roles NMD machinery may play in different cellular processes.


Asunto(s)
Degradación de ARNm Mediada por Codón sin Sentido/fisiología , ARN Helicasas/metabolismo , ARN Helicasas/fisiología , Transactivadores/metabolismo , Transactivadores/fisiología , Proteínas Portadoras/genética , Codón sin Sentido/genética , Codón sin Sentido/fisiología , Humanos , Mutación , Biosíntesis de Proteínas , Estabilidad del ARN/fisiología , ARN Mensajero/genética , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
9.
Artículo en Inglés | MEDLINE | ID: mdl-38310449

RESUMEN

Chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy has emerged as a revolutionary approach for cancer treatment, especially for hematologic cancers. However, CAR-T therapy has some limitations, including cytokine release syndrome (CRS), immune cellassociated neurologic syndrome (ICANS), and difficulty in targeting solid tumors and delivering allogeneic cell therapy due to graft-versus-host disease (GvHD). Therefore, it is important to explore other cell sources for CAR engineering. Invariant natural killer T (iNKT) cells are a potential target, as they possess powerful antitumor ability and do not recognize mismatched major histocompatibility complexes (MHCs) and protein antigens, thus avoiding the risk of GvHD. CAR-engineered iNKT (CAR-iNKT) cell therapy offers a promising new approach to cancer immunotherapy by overcoming the drawbacks of CAR-T cell therapy while retaining potent antitumor capabilities. This review summarizes the current CAR-iNKT cell products, their functions and phenotypes, and their potential for off-the-shelf cancer immunotherapy.

10.
Nat Biotechnol ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38744947

RESUMEN

Cancer immunotherapy with autologous chimeric antigen receptor (CAR) T cells faces challenges in manufacturing and patient selection that could be avoided by using 'off-the-shelf' products, such as allogeneic CAR natural killer T (AlloCAR-NKT) cells. Previously, we reported a system for differentiating human hematopoietic stem and progenitor cells into AlloCAR-NKT cells, but the use of three-dimensional culture and xenogeneic feeders precluded its clinical application. Here we describe a clinically guided method to differentiate and expand IL-15-enhanced AlloCAR-NKT cells with high yield and purity. We generated AlloCAR-NKT cells targeting seven cancers and, in a multiple myeloma model, demonstrated their antitumor efficacy, expansion and persistence. The cells also selectively depleted immunosuppressive cells in the tumor microenviroment and antagonized tumor immune evasion via triple targeting of CAR, TCR and NK receptors. They exhibited a stable hypoimmunogenic phenotype associated with epigenetic and signaling regulation and did not induce detectable graft versus host disease or cytokine release syndrome. These properties of AlloCAR-NKT cells support their potential for clinical translation.

11.
Front Med (Lausanne) ; 10: 1135468, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37064017

RESUMEN

The realm of cell-based immunotherapy holds untapped potential for the development of next-generation cancer treatment through genetic engineering of chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapies for targeted eradication of cancerous malignancies. Such allogeneic "off-the-shelf" cell products can be advantageously manufactured in large quantities, stored for extended periods, and easily distributed to treat an exponential number of cancer patients. At current, patient risk of graft-versus-host disease (GvHD) and host-versus-graft (HvG) allorejection severely restrict the development of allogeneic CAR-T cell products. To address these limitations, a variety of genetic engineering strategies have been implemented to enhance antitumor efficacy, reduce GvHD and HvG onset, and improve the overall safety profile of T-cell based immunotherapies. In this review, we summarize these genetic engineering strategies and discuss the challenges and prospects these approaches provide to expedite progression of translational and clinical studies for adoption of a universal cell-based cancer immunotherapy.

12.
Cell Stem Cell ; 30(5): 592-610, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36948187

RESUMEN

Advances in cell-based therapy, particularly CAR-T cell therapy, have transformed the treatment of hematological malignancies. Although an important step forward for the field, autologous CAR-T therapies are hindered by high costs, manufacturing challenges, and limited efficacy against solid tumors. With ongoing progress in gene editing and culture techniques, engineered stem cells and their application in cell therapy are poised to address some of these challenges. Here, we review stem cell-based immunotherapy approaches, stem cell sources, gene engineering and manufacturing strategies, therapeutic platforms, and clinical trials, as well as challenges and future directions for the field.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/genética , Inmunoterapia , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Ingeniería Celular
13.
Mol Ther Oncolytics ; 31: 100733, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-37876793

RESUMEN

CRISPR screen technology enables systematic and scalable interrogation of gene function by using the CRISPR-Cas9 system to perturb gene expression. In the field of cancer immunotherapy, this technology has empowered the discovery of genes, biomarkers, and pathways that regulate tumor development and progression, immune reactivity, and the effectiveness of immunotherapeutic interventions. By conducting large-scale genetic screens, researchers have successfully identified novel targets to impede tumor growth, enhance anti-tumor immune responses, and surmount immunosuppression within the tumor microenvironment (TME). Here, we present an overview of CRISPR screens conducted in tumor cells for the purpose of identifying novel therapeutic targets. We also explore the application of CRISPR screens in immune cells to propel the advancement of cell-based therapies, encompassing T cells, natural killer cells, dendritic cells, and macrophages. Furthermore, we outline the crucial components necessary for the successful implementation of immune-specific CRISPR screens and explore potential directions for future research.

14.
ACS Nano ; 17(14): 14152-14160, 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37410702

RESUMEN

Extensive macroscale two-dimensional (2-D) platinum (Pt) nanowire network (NWN) sheets are created through a hierarchical self-assembly process with the aid of biomolecular ligands. The Pt NWN sheet is assembled from the attachment growth of 1.9 nm-sized 0-D nanocrystals into 1-D nanowires featuring a high density of grain boundaries, which then interconnect to form monolayer network structures extending into centimeter-scale size. Further investigation into the formation mechanism reveals that the initial emergence of NWN sheets occurs at the gas/liquid interfaces of the bubbles produced by sodium borohydride (NaBH4) during the synthesis process. Upon the rupture of these bubbles, an exocytosis-like process releases the Pt NWN sheets at the gas/liquid surface, which subsequently merge into a continuous monolayer Pt NWN sheet. The Pt NWN sheets exhibit outstanding oxygen reduction reaction (ORR) activities, with specific and mass activities 12.0 times and 21.2 times greater, respectively, than those of current state-of-the-art commercial Pt/C electrocatalysts.

15.
Sci Adv ; 9(42): eadj0540, 2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37851816

RESUMEN

The current cardiac pacemakers are battery dependent, and the pacing leads are prone to introduce valve damage and infection, plus a complete pacemaker retrieval is needed for battery replacement. Despite the reported wireless bioelectronics to pace the epicardium, open-chest surgery (thoracotomy) is required to implant the device, and the procedure is invasive, requiring prolonged wound healing and health care burden. We hereby demonstrate a fully biocompatible wireless microelectronics with a self-assembled design that can be rolled into a lightweight microtubular pacemaker for intravascular implantation and pacing. The radio frequency was used to transfer energy to the microtubular pacemaker for electrical stimulation. We show that this pacemaker provides effective pacing to restore cardiac contraction from a nonbeating heart and have the capacity to perform overdrive pacing to augment blood circulation in an anesthetized pig model. Thus, this microtubular pacemaker paves the way for the minimally invasive implantation of leadless and battery-free microelectronics.


Asunto(s)
Estimulación Cardíaca Artificial , Marcapaso Artificial , Animales , Porcinos , Estimulación Cardíaca Artificial/métodos , Prótesis e Implantes , Corazón , Estimulación Eléctrica , Diseño de Equipo , Resultado del Tratamiento
16.
iScience ; 26(10): 107952, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37810241

RESUMEN

Ovarian cancer (OC) is highly lethal due to late detection and frequent recurrence. Initial treatments, comprising surgery and chemotherapy, lead to disease remission but are invariably associated with subsequent relapse. The identification of novel therapies and an improved understanding of the molecular and cellular characteristics of OC are urgently needed. Here, we conducted a comprehensive analysis of primary tumor cells and their microenvironment from 16 chemonaive and 10 recurrent OC patient samples. Profiling OC tumor biomarkers allowed for the identification of potential molecular targets for developing immunotherapies, while profiling the microenvironment yielded insights into its cellular composition and property changes between chemonaive and recurrent samples. Notably, we identified CD1d as a biomarker of the OC microenvironment and demonstrated its targeting by invariant natural killer T (iNKT) cells. Overall, our study presents a comprehensive immuno-profiling of OC tumor and microenvironment during disease progression, guiding the development of immunotherapies for OC treatment, especially for recurrent disease.

17.
Nat Commun ; 14(1): 6942, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37938576

RESUMEN

Allogeneic Vγ9Vδ2 (Vδ2) T cells have emerged as attractive candidates for developing cancer therapy due to their established safety in allogeneic contexts and inherent tumor-fighting capabilities. Nonetheless, the limited clinical success of Vδ2 T cell-based treatments may be attributed to donor variability, short-lived persistence, and tumor immune evasion. To address these constraints, we engineer Vδ2 T cells with enhanced attributes. By employing CD16 as a donor selection biomarker, we harness Vδ2 T cells characterized by heightened cytotoxicity and potent antibody-dependent cell-mediated cytotoxicity (ADCC) functionality. RNA sequencing analysis supports the augmented effector potential of Vδ2 T cells derived from CD16 high (CD16Hi) donors. Substantial enhancements are further achieved through CAR and IL-15 engineering methodologies. Preclinical investigations in two ovarian cancer models substantiate the effectiveness and safety of engineered CD16Hi Vδ2 T cells. These cells target tumors through multiple mechanisms, exhibit sustained in vivo persistence, and do not elicit graft-versus-host disease. These findings underscore the promise of engineered CD16Hi Vδ2 T cells as a viable therapeutic option for cancer treatment.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neoplasias Ováricas , Femenino , Humanos , Interleucina-15/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Biomarcadores
18.
Front Immunol ; 13: 999549, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275727

RESUMEN

The immunosuppressive tumor microenvironment (TME) remains one of the most prevailing barriers obstructing the implementation of effective immunotherapy against solid-state cancers. Eminently composed of immunosuppressive tumor associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) among others, the TME attenuates the effects of immune checkpoint blockade and adoptive cell therapies, mandating a novel therapy capable of TME remediation. In this review we explore the potential of three innate-like T cell subsets, invariant natural killer T (iNKT), mucosal-associated invariant T (MAIT) cells, and gamma delta T (γδT) cells, that display an intrinsic anti-TAM/MDSC capacity. Exhibiting both innate and adaptive properties, innate-like T cell types express a subset-specific TCR with distinct recombination, morphology, and target cell recognition, further supplemented by a variety of NK activating receptors. Both NK activating receptor and TCR activation result in effector cell cytotoxicity against targeted immunosuppressive cells for TME remediation. In addition, innate-like T cells showcase moderate levels of tumor cell killing, providing dual antitumor and anti-TAM/MDSC function. This latent antitumor capacity can be further bolstered by chimeric antigen receptor (CAR) engineering for recognition of tumor specific antigens to enhance antitumor targeting. In contrast with established CAR-T cell therapies, adoption of these innate-like cell types provides an enhanced safety profile without the risk of graft versus host disease (GvHD), due to their non-recognition of mismatched major histocompatibility complex (MHC) molecules, for use as widely accessible, allogeneic "off-the-shelf" cancer immunotherapy.


Asunto(s)
Receptores Quiméricos de Antígenos , Microambiente Tumoral , Receptores Quiméricos de Antígenos/metabolismo , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Inmunoterapia Adoptiva
19.
Cells ; 11(9)2022 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-35563889

RESUMEN

Tumor-associated macrophages (TAMs) accumulate in the solid tumor microenvironment (TME) and have been shown to promote tumor growth and dampen antitumor immune responses. TAM-mediated suppression of T-cell antitumor reactivity is considered to be a major obstacle for many immunotherapies, including immune checkpoint blockade and adoptive T/CAR-T-cell therapies. An ex vivo culture system closely mimicking the TME can greatly facilitate the study of cancer immunotherapies. Here, we report the development of a 3D TME-mimicry culture that is comprised of the three major components of a human TME, including human tumor cells, TAMs, and tumor antigen-specific T cells. This TME-mimicry culture can readout the TAM-mediated suppression of T-cell antitumor reactivity, and therefore can be used to study TAM modulation of T-cell-based cancer immunotherapy. As a proof-of-principle, the studies of a PD-1/PD-L1 blockade therapy and a MAO-A blockade therapy were performed and validated.


Asunto(s)
Neoplasias , Microambiente Tumoral , Humanos , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Inmunoterapia Adoptiva , Neoplasias/tratamiento farmacológico , Macrófagos Asociados a Tumores
20.
Cancers (Basel) ; 14(11)2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35681730

RESUMEN

The field of T cell-based and chimeric antigen receptor (CAR)-engineered T (CAR-T) cell-based antitumor immunotherapy has seen substantial developments in the past decade; however, considerable issues, such as graft-versus-host disease (GvHD) and tumor-associated immunosuppression, have proven to be substantial roadblocks to widespread adoption and implementation. Recent developments in innate immune cell-based CAR therapy have opened several doors for the expansion of this therapy, especially as it relates to allogeneic cell sources and solid tumor infiltration. This study establishes in vitro killing assays to examine the TAM-targeting efficacy of MAIT, iNKT, and γδT cells. This study also assesses the antitumor ability of CAR-engineered innate T cells, evaluating their potential adoption for clinical therapies. The in vitro trials presented in this study demonstrate the considerable TAM-killing abilities of all three innate T cell types, and confirm the enhanced antitumor abilities of CAR-engineered innate T cells. The tumor- and TAM-targeting capacity of these innate T cells suggest their potential for antitumor therapy that supplements cytotoxicity with remediation of tumor microenvironment (TME)-immunosuppression.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA