Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Immunol Lett ; 268: 106881, 2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38810886

RESUMO

Wilms' tumor (WT1), a transcription factor highly expressed in various leukemias and solid tumors, is a highly specific intracellular tumor antigen, requiring presentation through complexation with HLA-restricted peptides.. WT1-derived epitopes are able to assemble with MHC-I and thereby be recognized by T cell receptors (TCR). Identification of new targetable epitopes derived from WT1 on solid tumors is a challenge, but meaningful for the development of therapeutics that could in this way target intracellular oncogenic proteins. In this study, we developed and comprehensively describe methods to validate the formation of the complex of WT1126-134 and HLA-A2. Subsequently, we developed an antibody fragment able to recognize the extracellular complex on the surface of cancer cells. The single chain variable fragment (scFv) of an established TCR-mimic antibody, specifically recognizing the WT1-derived peptide presented by the HLA-A2 complex, was expressed, purified, and functionally validated using a T2 cell antigen presentation model. Furthermore, we evaluated the potential of the WT1-derived peptide as a targetable extracellular antigen in multiple solid tumor cell lines. Our study describes methodology for the evaluation of WT1-derived peptides as tumor-specific antigen on solid tumors, and may facilitate the selection of potential candidates for future immunotherapy targeting WT1 epitopes.

2.
Nat Commun ; 15(1): 1909, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429294

RESUMO

Severe heterogeneity within glioblastoma has spurred the notion that disrupting the interplay between multiple elements on immunosuppression is at the core of meaningful anti-tumor responses. T cell immunoreceptor with Ig and ITIM domains (TIGIT) and its glioblastoma-associated antigen, CD155, form a highly immunosuppressive axis in glioblastoma and other solid tumors, yet targeting of TIGIT, a functionally heterogeneous receptor on tumor-infiltrating immune cells, has largely been ineffective as monotherapy, suggesting that disruption of its inhibitory network might be necessary for measurable responses. It is within this context that we show that the usurpation of the TIGIT - CD155 axis via engineered synNotch-mediated activation of induced pluripotent stem cell-derived natural killer (NK) cells promotes transcription factor-mediated activation of a downstream signaling cascade that results in the controlled, localized blockade of CD73 to disrupt purinergic activity otherwise resulting in the production and accumulation of immunosuppressive extracellular adenosine. Such "decoy" receptor engages CD155 binding to TIGIT, but tilts inhibitory TIGIT/CD155 interactions toward activation via downstream synNotch signaling. Usurping activities of TIGIT and CD73 promotes the function of adoptively transferred NK cells into intracranial patient-derived models of glioblastoma and enhances their natural cytolytic functions against this tumor to result in complete tumor eradication. In addition, targeting both receptors, in turn, reprograms the glioblastoma microenvironment via the recruitment of T cells and the downregulation of M2 macrophages. This study demonstrates that TIGIT/CD155 and CD73 are targetable receptor partners in glioblastoma. Our data show that synNotch-engineered pluripotent stem cell-derived NK cells are not only effective mediators of anti-glioblastoma responses within the setting of CD73 and TIGIT/CD155 co-targeting, but represent a powerful allogeneic treatment option for this tumor.


Assuntos
Glioblastoma , Células-Tronco Pluripotentes Induzidas , Células Matadoras Naturais , Humanos , Glioblastoma/terapia , Glioblastoma/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Matadoras Naturais/metabolismo , Receptores Imunológicos/metabolismo , Linfócitos T/metabolismo , Microambiente Tumoral , 5'-Nucleotidase/imunologia , 5'-Nucleotidase/metabolismo
3.
Mol Pharm ; 21(2): 651-660, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38230666

RESUMO

Natural killer (NK) cell-based immunotherapy has benefitted from the multiple strengths that NK cells offer in adoptive transfer settings, not the least of which is their safety and potential for allogeneic use. Such use, however, necessitates the cryopreservation of NK cell-based therapy products to support logistical efforts in deploying these cells in different locations, decentralized from the point of collection or manufacturing. DMSO, the most commonly used cryoprotective agent (CPA), has been effective in protecting immune cells during freezing and thawing, but its ability to induce molecular and genetic changes to immune cells as well as its toxicity has stimulated interest in alternative CPAs. However, replacing DMSO's ability to act intracellularly has been difficult, and the sensitivity of human peripheral blood-derived NK cells to freezing and thawing-induced damage has meant that investigations into the potential of replacing DMSO are lacking. As a first step toward establishing the feasibility of cryopreserving human NK cells with CPAs' alternative to DMSO, we investigate the potential of using noncell-penetrating and cell-penetrating CPAs to recover NK cells post-thaw without DMSO. Here, we find that cryoprotection using cell-penetrating CPAs can retain the viability of human peripheral blood-derived NK cells to a comparable degree to DMSO. In addition, non-DMSO-cryopreserved human NK cells were as cytotoxic as those cryopreserved with DMSO and displayed a comparable level of surface markers of activation. In summary, we present the first example of the potential of developing non-DMSO CPA formulations that could be deployed in future cell therapy regimens.


Assuntos
Criopreservação , Dimetil Sulfóxido , Humanos , Crioprotetores/farmacologia , Congelamento , Células Matadoras Naturais , Sobrevivência Celular
4.
iScience ; 26(12): 108353, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38053639

RESUMO

TIGIT is a receptor on human natural killer (NK) cells. Here, we report that TIGIT does not spontaneously induce inhibition of NK cells in glioblastoma (GBM), but rather acts as a decoy-like receptor, by usurping binding partners and regulating expression of NK activating ligands and receptors. Our data show that in GBM patients, one of the underpinnings of unresponsiveness to TIGIT blockade is that by targeting TIGIT, NK cells do not lose an inhibitory signal, but gains the potential for new interactions with other, shared, TIGIT ligands. Therefore, TIGIT does not define NK cell dysfunction in GBM. Further, in GBM, TIGIT+ NK cells are hyperfunctional. In addition, we discovered that 4-1BB correlates with TIGIT expression, the agonism of which contributes to TIGIT immunotherapy. Overall, our data suggest that in GBM, TIGIT acts as a regulator of a complex network, and provide new clues about its use as an immunotherapeutic target.

5.
Trends Pharmacol Sci ; 44(11): 832-848, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37770314

RESUMO

The adaptation of natural killer (NK) cells to conditions in the microenvironment of tumors is deeply affected by their metabolic activity, itself a result of nutrient availability and the metabolism of the cancer cells themselves. Elevated rates of glycolysis and lipid metabolism in cancers not only lead to the accumulation of immunosuppressive byproducts but also contribute to an environment of elevated concentrations of extracellular metabolites. This results in altered NK cell bioenergetics through changes in transcriptional and translational profiles, ultimately affecting their pharmacology and impairing NK cell responses. However, understanding the metabolic processes that drive alterations in immunological signaling on NK cells remains both difficult and vastly underexplored. We discuss the varied and complex drivers of NK cell metabolism in homeostasis and the tumor microenvironment (TME), challenges associated with their targetability, and unexplored therapeutic opportunities.

6.
Wiad Lek ; 76(12): 2543-2555, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38290016

RESUMO

Marie Sklodowska-Curie Symposia on Cancer Research and Care (MSCS-CRC) promote collaborations between cancer researchers and care providers in the United States, Canada and Central and Eastern European Countries (CEEC), to accelerate the development of new cancer therapies, advance early detection and prevention, increase cancer awareness, and improve cancer care and the quality of life of patients and their families. The third edition of MSCS-CRC, held at Roswell Park Comprehensive Cancer Center, Buffalo, NY, in September 2023, brought together 137 participants from 20 academic institutions in the US, Poland, Ukraine, Lithuania, Croatia and Hungary, together with 16 biotech and pharma entities. The key areas of collaborative opportunity identified during the meeting are a) creating of a database of available collaborative projects in the areas of early-phase clinical trials, preclinical development, and identification of early biomarkers; b) promoting awareness of cancer risks and efforts at cancer prevention; c) laboratory and clinical training; and d) sharing experience in cost-effective delivery of cancer care and improving the quality of life of cancer patients and their families. Examples of ongoing international collaborations in the above areas were discussed. Participation of the representatives of the Warsaw-based Medical Research Agency, National Cancer Institute (NCI) of the United States, National Cancer Research Institutes of Poland and Lithuania, New York State Empire State Development, Ministry of Health of Ukraine and Translational Research Cancer Center Consortium of 13 cancer centers from the US and Canada, facilitated the discussion of available governmental and non-governmental funding initiatives in the above areas.


Assuntos
Pesquisa Biomédica , Neoplasias , Humanos , Estados Unidos , New York , Qualidade de Vida , Neoplasias/terapia , Polônia
7.
Mol Ther Oncolytics ; 27: 224-238, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36420307

RESUMO

Treatment of many cancers, particularly those that remain difficult to treat or are refractive after standard-of-care therapies, has been challenging with cell-based therapies. Although relatively safe as allogeneic therapies and innately effective against cancers without the need for antigen sensitization, natural killer (NK) cells have necessitated use of genetic manipulation approaches to enhance their specificity, persistence, and homing. Chimeric antigen receptor (CAR) and gene-edited NK cell therapies have emerged as a potent treatment modality, addressing many of the issues that have plagued such gene-based therapies with other cell types. Early examples of engineered NK cell therapies have largely leveraged their activity against hematological malignancies in combination with conventional construct architectures or by editing putative genetic targets of immunosuppression. As the motivation to tackle more complex solid tumors grows, so has the sophistication and emergence of NK-specific constructs and engineering approaches. Multi-CARs, combinations with diverse genome editing technologies, as well as responsive and sensing CARs have appeared in the context of NK cell therapy. Here we discuss engineering approaches for NK cell therapy, the latest developments in the field, and what stands in the way of those promises en route to clinical translation.

8.
Elife ; 112022 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-35815945

RESUMO

Immunometabolic reprogramming due to adenosine produced by CD73 (encoded by the 5'-ectonucleotidase gene NT5E) is a recognized immunosuppressive mechanism contributing to immune evasion in solid tumors. Adenosine is not only known to contribute to tumor progression, but it has specific roles in driving dysfunction of immune cells, including natural killer (NK) cells. Here, we engineered human NK cells to directly target the CD73-adenosine axis by blocking the enzymatic activity of CD73. In doing so, the engineered NK cells not only impaired adenosinergic metabolism driven by the hypoxic uptake of ATP by cancer cells in a model of non-small-cell lung cancer, but also mediated killing of tumor cells due to the specific recognition of overexpressed CD73. This resulted in a 'single agent' immunotherapy that combines antibody specificity, blockade of purinergic signaling, and killing of targets mediated by NK cells. We also showed that CD73-targeted NK cells are potent in vivo and result in tumor arrest, while promoting NK cell infiltration into CD73+ tumors and enhancing intratumoral activation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Adenosina/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Humanos , Imunoterapia/métodos , Células Matadoras Naturais , Neoplasias Pulmonares/metabolismo
9.
Cancer Immunol Immunother ; 71(12): 3043-3056, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35622118

RESUMO

The production of adenosine by CD73 on cancer cells in the tumor microenvironment is a recognized immunosuppressive mechanism contributing to immune evasion in many solid tumors. While NK cells have been purported to overexpress CD73 under certain conditions, this phenomenon has remained elusive and unclear. We have found that while NK cells are able to upregulate expression of CD73 on their surface when exposed to CD73+ cancer cells, this upregulation is not universal, nor is it often substantial. Rather, our data point to the extent of CD73 expression on NK cells to be both cancer-specific and environmentally-driven, and largely limited in intensity. We found that NK cell overexpression of CD73 responds to the level of CD73 on cancer cells and is enhanced in hypoxia. Interestingly, human CD73+ NK cells appear hyperfunctional in vitro compared to CD73- NK cells, suggesting that CD73 expression could be a bystander of NK cell activation. In addition, glioblastoma patient data show that tumor-infiltrating NK cells express CD73 variably, depending on donor, and present lower expression of CD16, alongside patient-specific changes in CEACAM1, CXCR3 and TIM-3, suggesting some functional changes in NK cell responses associated with expression of CD73 on NK cells in vivo. Taken together, our study is the first to show that while NK cells are largely resistant to the upregulation of CD73, CD73 expression is inducible on NK cells in response to CD73 on cancer cells, and these cells are associated with distinct functional signatures.


Assuntos
Glioblastoma , Células Matadoras Naturais , Humanos , Adenosina/metabolismo , Glioblastoma/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Células Matadoras Naturais/metabolismo , Microambiente Tumoral
10.
Front Immunol ; 13: 756034, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35309302

RESUMO

Neutrophil migration and activation are essential for defense against pathogens. However, this process may also lead to collateral tissue injury. We used microRNA overexpression as a platform and discovered protein-coding genes that regulate neutrophil migration. Here we show that miR-99 decreased the chemotaxis of zebrafish neutrophils and human neutrophil-like cells. In zebrafish neutrophils, miR-99 directly targets the transcriptional factor RAR-related orphan receptor alpha (roraa). Inhibiting RORα, but not the closely related RORγ, reduced chemotaxis of zebrafish and primary human neutrophils without causing cell death, and increased susceptibility of zebrafish to bacterial infection. Expressing a dominant-negative form of Rorα or disrupting the roraa locus specifically in zebrafish neutrophils reduced cell migration. At the transcriptional level, RORα regulates transmembrane signaling receptor activity and protein phosphorylation pathways. Our results, therefore, reveal previously unknown functions of miR-99 and RORα in regulating neutrophil migration and anti-microbial defense.


Assuntos
MicroRNAs , Peixe-Zebra , Animais , Movimento Celular/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Neutrófilos/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
11.
Front Immunol ; 13: 823618, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35222404

RESUMO

Over the past decade, lung cancer treatment has undergone a major paradigm shift. A greater understanding of lung cancer biology has led to the development of many effective targeted therapies as well as of immunotherapy. Immune checkpoint inhibitors (ICIs) have shown tremendous benefit in the treatment of non-small cell lung cancer (NSCLC) and are now being used as first-line therapies in metastatic disease, consolidation therapy following chemoradiation in unresectable locally advanced disease, and adjuvant therapy following surgical resection and chemotherapy in resectable disease. Despite these benefits, predicting who will respond to ICIs has proven to be difficult and there remains a need to discover new predictive immunotherapy biomarkers. Furthermore, resistance to ICIs in lung cancer is frequent either because of a lack of response or disease progression after an initial response. The utility of ICIs in the treatment of small cell lung cancer (SCLC) remains limited to first-line treatment of extensive stage disease in combination with chemotherapy with modest impact on overall survival. It is thus important to explore and exploit additional targets to reap the full benefits of immunotherapy in the treatment of lung cancer. Here, we will summarize the current state of immunotherapy in lung cancer, discuss novel targets, and explore the intersection between DNA repair defects and immunotherapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Humanos , Fatores Imunológicos/uso terapêutico , Imunoterapia , Neoplasias Pulmonares/patologia , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico
12.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34740973

RESUMO

Tumor antigen heterogeneity, a severely immunosuppressive tumor microenvironment (TME) and lymphopenia resulting in inadequate immune intratumoral trafficking, have rendered glioblastoma (GBM) highly resistant to therapy. To address these obstacles, here we describe a unique, sophisticated combinatorial platform for GBM: a cooperative multifunctional immunotherapy based on genetically engineered human natural killer (NK) cells bearing multiple antitumor functions including local tumor responsiveness that addresses key drivers of GBM resistance to therapy: antigen escape, immunometabolic reprogramming of immune responses, and poor immune cell homing. We engineered dual-specific chimeric antigen receptor (CAR) NK cells to bear a third functional moiety that is activated in the GBM TME and addresses immunometabolic suppression of NK cell function: a tumor-specific, locally released antibody fragment which can inhibit the activity of CD73 independently of CAR signaling and decrease the local concentration of adenosine. The multifunctional human NK cells targeted patient-derived GBM xenografts, demonstrated local tumor site-specific activity in the tissue, and potently suppressed adenosine production. We also unveil a complex reorganization of the immunological profile of GBM induced by inhibiting autophagy. Pharmacologic impairment of the autophagic process not only sensitized GBM to antigenic targeting by NK cells but promoted a chemotactic profile favorable to NK infiltration. Taken together, our study demonstrates a promising NK cell-based combinatorial strategy that can target multiple clinically recognized mechanisms of GBM progression simultaneously.


Assuntos
Engenharia Genética , Glioblastoma/terapia , Imunoterapia Adotiva , Células Matadoras Naturais , Microambiente Tumoral/imunologia , Animais , Autofagia , Glioblastoma/imunologia , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
BioDrugs ; 35(5): 529-545, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34427899

RESUMO

Dimethylsufoxide (DMSO) being universally used as a cryoprotectant in clinical adoptive cell-therapy settings to treat hematological malignancies and solid tumors is a growing concern, largely due to its broad toxicities. Its use has been associated with significant clinical side effects-cardiovascular, neurological, gastrointestinal, and allergic-in patients receiving infusions of cell-therapy products. DMSO has also been associated with altered expression of natural killer (NK) and T-cell markers and their in vivo function, not to mention difficulties in scaling up DMSO-based cryoprotectants, which introduce manufacturing challenges for autologous and allogeneic cellular therapies, including chimeric antigen receptor (CAR)-T and CAR-NK cell therapies. Interest in developing alternatives to DMSO has resulted in the evaluation of a variety of sugars, proteins, polymers, amino acids, and other small molecules and osmolytes as well as modalities to efficiently enable cellular uptake of these cryoprotectants. However, the DMSO-free cryopreservation of NK and T cells remains difficult. They represent heterogeneous cell populations that are sensitive to freezing and thawing. As a result, clinical use of cryopreserved cell-therapy products has not moved past the use of DMSO. Here, we present the state of the art in the development and use of cryopreservation options that do not contain DMSO toward clinical solutions to enable the global deployment of safer adoptively transferred cell-based therapies.


Assuntos
Criopreservação , Imunoterapia Adotiva , Células Matadoras Naturais/citologia , Linfócitos T/citologia , Dimetil Sulfóxido/farmacologia , Humanos , Receptores de Antígenos Quiméricos
14.
Cytotherapy ; 23(10): 939-952, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34272175

RESUMO

BACKGROUND AIMS: Traditionally, natural killer (NK) cells are sourced from the peripheral blood of donors-a laborious and highly donor-specific process. Processes for generating NK cells from induced pluripotent stem cells (iPSCs) have demonstrated that it is possible to successfully generate renewable alloreactive NK cells that are not only functional in vivo but can also be genetically engineered for enhanced function. However, poor standardization and cumbersome differentiation procedures suggest that further improvements in the control of the differentiation process are necessary. METHODS: Here the authors evaluated the potential of differentiating NK cells from centrally authenticated iPSCs under entirely chemically defined and serum-free conditions as well as their immunotherapeutic potential, after expansion in feeder-free media, against solid tumors targets. To address limitations of current differentiation approaches, the authors did not utilize feeder or stromal cell layers, TrypLE adaptation or peripheral blood during the differentiation process. The authors also evaluated the feasibility of utilizing centrally authenticated iPSC lines, thus circumventing protocol- and donor-induced variability associated with reprogramming approaches, and characterized these iPSC-NK cells in terms of cytotoxicity, cytokine production and degranulation potential against solid tumor cell lines and patient-derived targets. RESULTS: Differentiation of iPSCs generated NK cells that were predominantly CD56+/CD16+/CD3- and expressed NK activation markers NKG2D, NKp30, NKp44, NKp46 and DNAM-1. These iPSC-NK cells mediated effector functions, including cytotoxicity, degranulation and IFN-γ production, in response to solid tumor targets, including patient-derived cancer cells, and could be cryopreserved and expanded in culture. CONCLUSIONS: The ability to produce NK cells under defined conditions and the functional responses elicited by these iPSC-NK cells suggest that they could represent promising effectors in clinical adoptive transfer settings as a renewable source of donor-independent NK cells for immunotherapy of solid tumors.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Linhagem Celular Tumoral , Humanos , Imunoterapia , Células Matadoras Naturais
15.
Cytokine Growth Factor Rev ; 59: 36-45, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33495094

RESUMO

Natural killer (NK) cell-based cell therapy has been emerging as a powerful weapon in the treatment of multiple malignancies. However, the inadequate infiltration of the therapeutic NK cells into solid tumors remains a big challenge to their clinical utility. Chemokine networks, which play essential roles in the migration of lymphocytes, have been recognized as critical in driving the intratumoral infiltration of NK cells via interactions between soluble chemokines and their receptors. Often, such interactions are complex and disease-specific. In the context of NK cells, chemokine receptors of note have included CCR2, CCR5, CCR7, CXCR3, and CX3CR1. The immunobiology of chemokine-receptor interactions has fueled the development of approaches that hope to improve the infiltration of NK cells into the microenvironment of solid tumors. Stimulation of NK cells ex vivo in the presence of various cytokines (such as IL-2, IL-15, and IL-21) and genetic engineering of NK cells have been utilized to alter the chemokine receptor profile and generate NK cells with higher infiltrating capacity. Additionally, the immune-suppressive tumor microenvironment has also been targeted, by introducing, either directly or indirectly, chemokine ligands which NK cells are able to respond to, ultimately creating a more hospitable niche for NK cell trafficking. Such strategies have promoted the infiltration and activity of infused NK cells into multiple solid tumors. In this review, we discuss how chemokine receptors and their ligands coordinate and how they can be manipulated to regulate the trafficking, distribution, and residence of NK cells in solid tumors.


Assuntos
Células Matadoras Naturais , Neoplasias , Quimiocinas , Humanos , Imunoterapia Adotiva , Neoplasias/terapia , Microambiente Tumoral
16.
Cancers (Basel) ; 12(9)2020 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-32858904

RESUMO

Among natural killer (NK) cell receptors, the T-cell immunoglobulin and mucin-containing domain (TIM-3) has been associated with both inhibitory and activating functions, depending on context and activation pathway. Ex vivo and in vitro, expression of TIM-3 is inducible and depends on activation stimulus. Here, we report that TIM-3 expression can be downregulated on NK cells under specific conditions. When NK cells are exposed to cancer targets, they synergize with stimulation conditions to induce a substantial decrease in TIM-3 expression on their surface. We found that such downregulation occurs following prior NK activation. Downregulated TIM-3 expression correlated to lower cytotoxicity and lower interferon gamma (IFN-γ) expression, fueling the notion that TIM-3 might function as a benchmark for human NK cell dysfunction.

17.
Cell Oncol (Dordr) ; 43(4): 577-600, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32488848

RESUMO

BACKGROUND: The unique ability of NK cells to target cancer cells without antigen specificity makes them an attractive prospect for immunotherapy of solid tumors. However, the complexity of the tumor microenvironment (TME), particularly its heterogeneity and associated immunosuppressive properties, enables solid tumor cells to escape NK cell immune-surveillance by impairing their infiltration and cytotoxic functions. As a result, NK cells that have been able to infiltrate solid tumors are dysfunctional, exhausted and metabolically and functionally impaired. Understanding the status of NK cells in solid tumors and the interplay between the tumor-promoting functions of the TME and the immunometabolic reprogramming events that NK cells endure as a result is essential to developing approaches to improve the clinical outcome of NK cell-based immunotherapies against solid tumors. CONCLUSIONS: In this review, we address the current knowledge on the presence and immunometabolic roles of NK cells in solid tumors as well as the strategies developed to restore NK cell activities in these conditions, with the ultimate goal of enhancing persistence, trafficking, cytotoxicity and metabolic functions.


Assuntos
Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Neoplasias/imunologia , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia , Animais , Reprogramação Celular/imunologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo
18.
J Hematol Oncol ; 13(1): 76, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32532329

RESUMO

Natural killer (NK) cells are powerful immune effectors, modulating their anti-tumor function through a balance activating and inhibitor ligands on their cell surface. Though still emerging, cancer immunotherapies utilizing NK cells are proving promising as a modality for the treatment of a number of solid tumors, including glioblastoma (GBM) and other gliomas, but are often limited due to complex immunosuppression associated with the GBM tumor microenvironment which includes overexpression of inhibitory receptors on GBM cells. CD155, or poliovirus receptor (PVR), has recently emerged as a pro-tumorigenic antigen, overexpressed on GBM and contributing to increased GBM migration and aggressiveness. CD155 has also been established as an immunomodulatory receptor, able to both activate NK cells through interactions with CD226 (DNAM-1) and CD96 and inhibit them through interaction with TIGIT. However, NK cell TIGIT expression has been shown to be upregulated in cancer, establishing CD155 as a predominantly inhibitory receptor within the context of GBM and other solid tumors, and rendering it of interest as a potential target for antigen-specific NK cell-based immunotherapy. This review will explore the function of CD155 within GBM as it relates to tumor migration and NK cell immunoregulation, as well as pre-clinical and clinical targeting of CD155/TIGIT and the potential that this pathway holds for the development of emerging NK cell-based immunotherapies.


Assuntos
Glioblastoma/terapia , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Invasividade Neoplásica/imunologia , Receptores Virais/antagonistas & inibidores , Animais , Antígenos CD/imunologia , Antígenos CD/fisiologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos de Diferenciação de Linfócitos T/fisiologia , Antineoplásicos Imunológicos/uso terapêutico , Adesão Celular , Movimento Celular , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Células Matadoras Naturais/transplante , Camundongos , Invasividade Neoplásica/prevenção & controle , Metástase Neoplásica , Terapia Viral Oncolítica , Poliovirus/fisiologia , Vírus Reordenados/fisiologia , Receptores Imunológicos/imunologia , Receptores Imunológicos/fisiologia , Receptores Virais/imunologia , Rhinovirus/fisiologia , Microambiente Tumoral/imunologia
19.
Adv Sci (Weinh) ; 7(9): 1902938, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32382476

RESUMO

The ability of natural killer (NK) cells to mediate potent antitumor immunity in clinical adoptive transfer settings relies, in large part, on their ability to retain cytotoxic function following cryopreservation. To avoid potential systemic toxicities associated with infusions of NK cells into patients in the presence of dimethylsulfoxide (DMSO), interest in alternative cryoprotective agents (CPAs) with improved safety profiles has grown. Despite the development of various sugars, amino acids, polyols, and polyampholytes as cryoprotectants, their ability to promote protection from intracellular cryodamage is limited because they mostly act outside of the cell. Though ways to shuttle cryoprotectants intracellularly exist, NK cells' high aversity to manipulation and freezing has meant they are highly understudied as targets for the development of new cryopreservation approaches. Here, the first example of a safe and efficient platform for the intracellular delivery of non-DMSO CPAs to NK cells is presented. Biocompatible chitosan-based nanoparticles are engineered to mediate the efficient DMSO-free cryopreservation of NK cells. NK cells cryopreserved in this way retain potent cytotoxic, degranulation, and cytokine production functions against tumor targets. This not only represents the first example of delivering nanoparticles to NK cells, but illustrates the clinical potential in manufacturing safer allogeneic adoptive immunotherapies "off the shelf."

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA