Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Trends Immunol ; 44(10): 748-750, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37652814

RESUMEN

Broadening immune responses through antigen spreading remains the 'Holy Grail' of cancer immunotherapy. A study by Ma and colleagues reveals that vaccine boosting of chimeric antigen receptor (CAR)-T cells in mice promotes endogenous immunity and elicits antigen spread to eliminate antigenically heterogenous solid tumors through a mechanism crucially dependent on interferon (IFN)γ.


Asunto(s)
Neoplasias , Receptores de Antígenos de Linfocitos T , Ratones , Animales , Inmunoterapia Adoptiva , Neoplasias/terapia , Linfocitos T
2.
Immunol Rev ; 290(1): 60-84, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31355493

RESUMEN

Malignant brain tumors, including glioblastoma, represent some of the most difficult to treat of solid tumors. Nevertheless, recent progress in immunotherapy, across a broad range of tumor types, provides hope that immunological approaches will have the potential to improve outcomes for patients with brain tumors. Chimeric antigen receptors (CAR) T cells, a promising immunotherapeutic modality, utilizes the tumor targeting specificity of any antibody or receptor ligand to redirect the cytolytic potency of T cells. The remarkable clinical response rates of CD19-targeted CAR T cells and early clinical experiences in glioblastoma demonstrating safety and evidence for disease modifying activity support the potential of further advancements ultimately providing clinical benefit for patients. The brain, however, is an immune specialized organ presenting unique and specific challenges to immune-based therapies. Remaining barriers to be overcome for achieving effective CAR T cell therapy in the central nervous system (CNS) include tumor antigenic heterogeneity, an immune-suppressive microenvironment, unique properties of the CNS that limit T cell entry, and risks of immune-based toxicities in this highly sensitive organ. This review will summarize preclinical and clinical data for CAR T cell immunotherapy in glioblastoma and other malignant brain tumors, including present obstacles to advancement.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Neoplasias Encefálicas/patología , Ingeniería Genética , Humanos , Inmunidad , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Resultado del Tratamiento
3.
N Engl J Med ; 375(26): 2561-9, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-28029927

RESUMEN

A patient with recurrent multifocal glioblastoma received chimeric antigen receptor (CAR)-engineered T cells targeting the tumor-associated antigen interleukin-13 receptor alpha 2 (IL13Rα2). Multiple infusions of CAR T cells were administered over 220 days through two intracranial delivery routes - infusions into the resected tumor cavity followed by infusions into the ventricular system. Intracranial infusions of IL13Rα2-targeted CAR T cells were not associated with any toxic effects of grade 3 or higher. After CAR T-cell treatment, regression of all intracranial and spinal tumors was observed, along with corresponding increases in levels of cytokines and immune cells in the cerebrospinal fluid. This clinical response continued for 7.5 months after the initiation of CAR T-cell therapy. (Funded by Gateway for Cancer Research and others; ClinicalTrials.gov number, NCT02208362 .).


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Glioblastoma/terapia , Inmunoterapia Adoptiva , Recurrencia Local de Neoplasia/terapia , Receptores de Antígenos de Linfocitos T/uso terapéutico , Ingeniería Celular , Terapia Combinada , Humanos , Subunidad alfa2 del Receptor de Interleucina-13 , Masculino , Persona de Mediana Edad
4.
Bioconjug Chem ; 29(5): 1659-1668, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29526082

RESUMEN

Even when treated with aggressive current therapies, patients with glioblastoma usually survive less than two years and exhibit a high rate of recurrence. CpG is an oligonucleotide that activates the innate immune system via Toll-like receptor 9 (TLR9) activation. Injection of CpG into glioblastoma tumors showed promise as an immunotherapy in mouse models but proved disappointing in human trials. One aspect of glioma that is not addressed by CpG therapy alone is the highly invasive nature of glioma cells, which is associated with resistance to radiation and chemotherapy. Here, we demonstrate that single-walled carbon nanotubes noncovalently functionalized with CpG (SWNT/CpG), which retain the immunostimulatory property of the CpG, selectively inhibit the migration of glioma cells and not macrophages without affecting cell viability or proliferation. SWNT/CpG also selectively decreased NF-κB activation in glioma cells, while activating macrophages by induction of the TLR9/NF-κB pathway, as we have previously reported. The migration inhibition of glioma cells was correlated with selective reduction of intracellular levels of reactive oxygen species (ROS), suggesting that an antioxidant-based mechanism mediates the observed effects. To the best of our knowledge, SWNT/CpG is the first nanomaterial that inhibits the migration of cancer cells while stimulating the immune system.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Glioma/tratamiento farmacológico , Nanotubos de Carbono/química , Oligodesoxirribonucleótidos/farmacología , Adyuvantes Inmunológicos/química , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioma/patología , Humanos , Ratones , Oligodesoxirribonucleótidos/química
5.
Eur J Immunol ; 44(8): 2489-2499, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24777831

RESUMEN

Primarily defined by their antigen-presenting property, dendritic cells (DCs) are being implemented as cancer vaccines in immunotherapeutic interventions. DCs can also function as direct tumor cell killers. How DC cytotoxic activity can be efficiently harnessed and the mechanisms controlling this nonconventional property are not fully understood. We report here that the tumoricidal potential of mouse DCs generated from myeloid precursors with GM-CSF and IL-15 (IL-15 DCs) can be triggered with the Toll-like receptor (TLR) 4 ligand lipopolysaccharide to a similar extent compared with that of their counterparts, conventionally generated with IL-4 (IL-4 DCs). The mechanism of tumor cell killing depends on the induction of iNOS expression by DCs. In contrast, interferon (IFN)-γ induces the cytotoxic activity of IL-4 but not IL-15 DCs. Although the IFN-γ-STAT-1 signaling pathway is overall functional in IL-15 DCs, IFN-γ fails to induce iNOS expression in these cells. iNOS expression is negatively controlled in IFN-γ-stimulated IL-15 DCs by the cooperation between the E3 SUMO ligase PIAS1 and STAT-3, and can be partially restored with PIAS1 siRNA and STAT-3 inhibitors.


Asunto(s)
Células Dendríticas/metabolismo , Interferón gamma/metabolismo , Interleucina-15/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-4/metabolismo , Ligandos , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo
6.
Crit Rev Immunol ; 33(1): 1-21, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23510023

RESUMEN

Universally viewed as the sentinels and messengers of the immune system and traditionally referred to as professional antigen-presenting cells, dendritic cells (DCs) play a fundamental role in antitumor immunity. DCs are uniquely equipped with the ability to acquire, process, and present to T lymphocytes tumor-derived antigens. They can drive the differentiation of naive T cells into activated tumor-specific effector lymphocytes. DCs also dictate the type and regulate the strength and duration of T-cell responses. In addition, they contribute to natural killer and natural killer T-cell antitumoral function and to B-cell-mediated immunity. Besides this cardinal role as orchestrators of innate and adaptive immune responses, many studies have provided evidence that DCs can also function as direct cytotoxic effectors against tumors. This less conventional aspect of DC function has, however, raised controversy as it relates to the origin of these cells and the induction, regulation, and mechanisms underlying their tumoricidal activity. The possible impact of the cytotoxic function of DCs on their capability to present antigens also has been the focus of intensive research. This review examines these questions and discusses the biological significance of this nontraditional property and possible strategies to exploit the killing potential of DCs in cancer immunotherapy.


Asunto(s)
Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Inmunoterapia , Neoplasias/terapia , Animales , Humanos , Neoplasias/inmunología
7.
Cells ; 13(13)2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38994929

RESUMEN

Standard-of-care treatment for Glioblastoma Multiforme (GBM) is comprised of surgery and adjuvant chemoradiation. Chimeric Antigen Receptor (CAR) T cell therapy has demonstrated disease-modifying activity in GBM and holds great promise. Radiation, a standard-of-care treatment for GBM, has well-known immunomodulatory properties and may overcome the immunosuppressive tumor microenvironment (TME); however, radiation dose optimization and integration with CAR T cell therapy is not well defined. Murine immunocompetent models of GBM were treated with titrated doses of stereotactic radiosurgery (SRS) of 5, 10, and 20 Gray (Gy), and the TME was analyzed using Nanostring. A conditioning dose of 10 Gy was determined based on tumor growth kinetics and gene expression changes in the TME. We demonstrate that a conditioning dose of 10 Gy activates innate and adaptive immune cells in the TME. Mice treated with 10 Gy in combination with mCAR T cells demonstrated enhanced antitumor activity and superior memory responses to rechallenge with IL13Rα2-positive tumors. Furthermore, 10 Gy plus mCAR T cells also protected against IL13Rα2-negative tumors through a mechanism that was, in part, c-GAS-STING pathway-dependent. Together, these findings support combination conditioning with low-dose 10 Gy radiation in combination with mCAR T cells as a therapeutic strategy for GBM.


Asunto(s)
Glioblastoma , Receptores Quiméricos de Antígenos , Microambiente Tumoral , Glioblastoma/terapia , Glioblastoma/inmunología , Glioblastoma/radioterapia , Glioblastoma/patología , Animales , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Ratones , Microambiente Tumoral/inmunología , Humanos , Línea Celular Tumoral , Inmunoterapia Adoptiva/métodos , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Linfocitos T/inmunología , Ratones Endogámicos C57BL , Inmunomodulación , Femenino
8.
Nat Med ; 30(4): 1001-1012, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38454126

RESUMEN

Chimeric antigen receptor T cell (CAR-T) therapy is an emerging strategy to improve treatment outcomes for recurrent high-grade glioma, a cancer that responds poorly to current therapies. Here we report a completed phase I trial evaluating IL-13Rα2-targeted CAR-T cells in 65 patients with recurrent high-grade glioma, the majority being recurrent glioblastoma (rGBM). Primary objectives were safety and feasibility, maximum tolerated dose/maximum feasible dose and a recommended phase 2 dose plan. Secondary objectives included overall survival, disease response, cytokine dynamics and tumor immune contexture biomarkers. This trial evolved to evaluate three routes of locoregional T cell administration (intratumoral (ICT), intraventricular (ICV) and dual ICT/ICV) and two manufacturing platforms, culminating in arm 5, which utilized dual ICT/ICV delivery and an optimized manufacturing process. Locoregional CAR-T cell administration was feasible and well tolerated, and as there were no dose-limiting toxicities across all arms, a maximum tolerated dose was not determined. Probable treatment-related grade 3+ toxicities were one grade 3 encephalopathy and one grade 3 ataxia. A clinical maximum feasible dose of 200 × 106 CAR-T cells per infusion cycle was achieved for arm 5; however, other arms either did not test or achieve this dose due to manufacturing feasibility. A recommended phase 2 dose will be refined in future studies based on data from this trial. Stable disease or better was achieved in 50% (29/58) of patients, with two partial responses, one complete response and a second complete response after additional CAR-T cycles off protocol. For rGBM, median overall survival for all patients was 7.7 months and for arm 5 was 10.2 months. Central nervous system increases in inflammatory cytokines, including IFNγ, CXCL9 and CXCL10, were associated with CAR-T cell administration and bioactivity. Pretreatment intratumoral CD3 T cell levels were positively associated with survival. These findings demonstrate that locoregional IL-13Rα2-targeted CAR-T therapy is safe with promising clinical activity in a subset of patients. ClinicalTrials.gov Identifier: NCT02208362 .


Asunto(s)
Glioblastoma , Glioma , Receptores Quiméricos de Antígenos , Humanos , Recurrencia Local de Neoplasia , Glioma/terapia , Linfocitos T , Glioblastoma/terapia , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos
9.
J Immunol ; 187(12): 6310-7, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22075702

RESUMEN

Dendritic cells (DCs) encompass a heterogeneous population of cells capable of orchestrating innate and adaptive immune responses. The ability of DCs to act as professional APCs has been the foundation for the development and use of these cells as vaccines in cancer immunotherapy. DCs are also endowed with the nonconventional property of directly killing tumor cells. The current study investigates the regulation of murine DC cytotoxic function by T lymphocytes. We provide evidence that CD4(+) Th-1, but not Th-2, Th-17 cells, or regulatory T cells, are capable of inducing DC cytotoxic function. IFN-γ was identified as the major factor responsible for Th-1-induced DC tumoricidal activity. Tumor cell killing mediated by Th-1-activated killer DCs was dependent on inducible NO synthase expression and NO production. Importantly, Th-1-activated killer DCs were capable of presenting the acquired Ags from the killed tumor cells to T lymphocytes in vitro or in vivo. These observations offer new possibilities for the application of killer DCs in cancer immunotherapy.


Asunto(s)
Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Interferón gamma/fisiología , Neoplasias Mamarias Experimentales/inmunología , Melanoma Experimental/inmunología , Receptores de Interferón/fisiología , Células TH1/inmunología , Células TH1/metabolismo , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Citotoxicidad Inmunológica/genética , Células Dendríticas/metabolismo , Femenino , Interferón gamma/metabolismo , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptor de Interferón gamma
10.
Clin Dev Immunol ; 2013: 957878, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24454480

RESUMEN

While the role of T helper 17 lymphocytes (Th17) in the pathogenesis of autoimmune diseases and in infectious immunity has been relatively well defined, the impact of these cells and their associated cytokines on cancer development is still under debate. Although multiple reports have indicated that Th17 can promote anticancer immunity, others have argued that these cells may exhibit tumor-promoting properties. This dichotomy in the function of Th17 lymphocytes in cancer may be related to the versatile nature of these cells, being capable of differentiating into either proinflammatory Th1 or suppressive FoxP3-expressing Treg cells or hybrid T cell subsets depending on the underlying environmental conditions. In the current review, we examine the role of Th17 lymphocytes and Th17-associated cytokines in cancer and discuss how factors that control their final lineage commitment decision may influence the balance between their tumor-promoting versus tumor-suppressing properties.


Asunto(s)
Citocinas/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Animales , Diferenciación Celular/inmunología , Humanos , Inmunoterapia , Interleucina-17/biosíntesis , Neoplasias/terapia , Células Th17/citología
11.
Cancer Res ; 83(17): 2813-2815, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37404063

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy has transformed clinical care against blood malignancies and is seeing encouraging progress against solid tumors. While scientific advancement has been rapid, our mechanistic understanding of intrinsic features of CAR-engineered T cells is still evolving. CAR products typically consist of CD4+ and CD8+ T-cell subsets at variable ratios, yet a clear understanding of how each subset contributes together and independently to therapeutic response is lacking. CD8+ CAR T cells are well characterized for their perforin-dependent killing effects; however, the role of CD4+ CAR T cells as "helpers" versus "killers" has been variable across models and warrants more in-depth investigation. A recent study by Boulch and colleagues published in Nature Cancer demonstrates that CD4+ CAR T cells, alone, can exert potent antitumor activity through a mechanism involving IFNγ. CD4+ CAR T-cell production of IFNγ creates a cytokine field that can act at a distance to kill both antigen-positive and -negative tumor cells that are sensitive to the proapoptotic effects of IFNγ. These new findings reveal important insights for the antitumor effects mediated by CD4+ CAR T cells, which could have significant clinical implications.

12.
Cancer Res Commun ; 3(1): 66-79, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36968221

RESUMEN

Chimeric antigen receptor (CAR) T cell immunotherapy is emerging as a powerful strategy for cancer therapy; however, an important safety consideration is the potential for off-tumor recognition of normal tissue. This is particularly important as ligand-based CARs are optimized for clinical translation. Our group has developed and clinically translated an IL13(E12Y) ligand-based CAR targeting the cancer antigen IL13Rα2 for treatment of glioblastoma (GBM). There remains limited understanding of how IL13-ligand CAR design impacts the activity and selectivity for the intended tumor-associated target IL13Rα2 versus the more ubiquitous unintended target IL13Rα1. In this study, we functionally compared IL13(E12Y)-CARs incorporating different intracellular signaling domains, including first-generation CD3ζ-containing CARs (IL13ζ), second-generation 4-1BB (CD137)-containing or CD28-containing CARs (IL13-BBζ or IL13-28ζ), and third-generation CARs containing both 4-1BB and CD28 (IL13-28BBζ). In vitro coculture assays at high tumor burden establish that second-generation IL13-BBζ or IL13-28ζ outperform first-generation IL13ζ and third-generation IL13-28BBζ CAR designs, with IL13-BBζ providing superior CAR proliferation and in vivo antitumor potency in human xenograft mouse models. IL13-28ζ displayed a lower threshold for antigen recognition, resulting in higher off-target IL13Rα1 reactivity both in vitro and in vivo. Syngeneic mouse models of GBM also demonstrate safety and antitumor potency of murine IL13-BBζ CAR T cells delivered systemically after lymphodepletion. These findings support the use of IL13-BBζ CARs for greater selective recognition of IL13Rα2 over IL13Rα1, higher proliferative potential, and superior antitumor responsiveness. This study exemplifies the potential of modulating factors outside the antigen targeting domain of a CAR to improve selective tumor recognition. Significance: This study reveals how modulating CAR design outside the antigen targeting domain improves selective tumor recognition. Specifically, this work shows improved specificity, persistence, and efficacy of 4-1BB-based IL13-ligand CARs. Human clinical trials evaluating IL13-41BB-CAR T cells are ongoing, supporting the clinical significance of these findings.


Asunto(s)
Glioblastoma , Subunidad alfa2 del Receptor de Interleucina-13 , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Receptores de Antígenos de Linfocitos T , Receptores Quiméricos de Antígenos/genética , Linfocitos T , Inmunoterapia Adoptiva/métodos , Subunidad alfa2 del Receptor de Interleucina-13/genética , Interleucina-13/genética , Antígenos CD28/genética , Ligandos , Glioblastoma/terapia , Modelos Animales de Enfermedad
13.
Glia ; 59(3): 486-98, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21264954

RESUMEN

Despite significant infiltration into tumors, the effector function of macrophages (MPs) and microglia (MG) appears to be suppressed in gliomas. Although STAT3 pathway is thought to play a role in this process, the exact mechanism by which gliomas induce STAT3 activation in MPs and MG is not known. Because activation of receptor for advanced glycation end products (RAGE) can induce STAT3, and because gliomas express high levels of S100B, a RAGE ligand, we hypothesized that MP/MG STAT3 activity may be modulated through S100B-RAGE interaction. Exposure of N9 MG and bone marrow-derived monocytes (BMM) to GL261 glioma condition medium (GCM) and low (nM) levels of S100B increased RAGE expression, induced STAT3 and suppressed MG function in vitro. Furthermore, neutralization of S100B in GCM, partially reversed IL-1ß suppression in BMM, suggesting that the inhibitory effect of GCM to be in part due to S100B. Finally, blockage of S100B-RAGE interaction inhibited STAT3 activation in N9 MG and in glioma MG/MP in vivo. These findings suggest that the RAGE pathway may play an important role in STAT3 induction in glioma-associated MG/MPs, and that this process may be mediated through S100B.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Microglía/metabolismo , Factores de Crecimiento Nervioso/fisiología , Proteínas S100/fisiología , Factor de Transcripción STAT3/fisiología , Transducción de Señal/fisiología , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/fisiología , Subunidad beta de la Proteína de Unión al Calcio S100 , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética
14.
Cancer Discov ; 11(9): 2248-2265, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33837065

RESUMEN

Chimeric antigen receptor (CAR) T cells mediate potent antigen-specific antitumor activity; however, their indirect effects on the endogenous immune system are not well characterized. Remarkably, we demonstrate that CAR T-cell treatment of mouse syngeneic glioblastoma (GBM) activates intratumoral myeloid cells and induces endogenous T-cell memory responses coupled with feed-forward propagation of CAR T-cell responses. IFNγ production by CAR T cells and IFNγ responsiveness of host immune cells are critical for tumor immune landscape remodeling to promote a more activated and less suppressive tumor microenvironment. The clinical relevance of these observations is supported by studies showing that human IL13Rα2-CAR T cells activate patient-derived endogenous T cells and monocytes/macrophages through IFNγ signaling and induce the generation of tumor-specific T-cell responses in a responding patient with GBM. These studies establish that CAR T-cell therapy has the potential to shape the tumor microenvironment, creating a context permissible for eliciting endogenous antitumor immunity. SIGNIFICANCE: Our findings highlight the critical role of IFNγ signaling for a productive CAR T-cell therapy in GBM. We establish that CAR T cells can activate resident myeloid populations and promote endogenous T-cell immunity, emphasizing the importance of host innate and adaptive immunity for CAR T-cell therapy of solid tumors.This article is highlighted in the In This Issue feature, p. 2113.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Inmunoterapia Adoptiva , Interferón gamma/metabolismo , Células Mieloides/inmunología , Receptores Quiméricos de Antígenos/inmunología , Animales , Glioblastoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Discov ; 11(5): 1192-1211, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33328215

RESUMEN

Glioblastoma (GBM) contains self-renewing GBM stem cells (GSC) potentially amenable to immunologic targeting, but chimeric antigen receptor (CAR) T-cell therapy has demonstrated limited clinical responses in GBM. Here, we interrogated molecular determinants of CAR-mediated GBM killing through whole-genome CRISPR screens in both CAR T cells and patient-derived GSCs. Screening of CAR T cells identified dependencies for effector functions, including TLE4 and IKZF2. Targeted knockout of these genes enhanced CAR antitumor efficacy. Bulk and single-cell RNA sequencing of edited CAR T cells revealed transcriptional profiles of superior effector function and inhibited exhaustion responses. Reciprocal screening of GSCs identified genes essential for susceptibility to CAR-mediated killing, including RELA and NPLOC4, the knockout of which altered tumor-immune signaling and increased responsiveness of CAR therapy. Overall, CRISPR screening of CAR T cells and GSCs discovered avenues for enhancing CAR therapeutic efficacy against GBM, with the potential to be extended to other solid tumors. SIGNIFICANCE: Reciprocal CRISPR screening identified genes in both CAR T cells and tumor cells regulating the potency of CAR T-cell cytotoxicity, informing molecular targeting strategies to potentiate CAR T-cell antitumor efficacy and elucidate genetic modifications of tumor cells in combination with CAR T cells to advance immuno-oncotherapy.This article is highlighted in the In This Issue feature, p. 995.


Asunto(s)
Neoplasias Encefálicas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Glioblastoma/genética , Células Madre Neoplásicas/metabolismo , Receptores Quiméricos de Antígenos/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Glioblastoma/patología , Humanos
16.
Plant Cell Physiol ; 51(2): 312-22, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20061301

RESUMEN

In the unicellular green alga Chlamydomonas reinhardtii, translation of the chloroplast-encoded psbA mRNA is regulated by the light-dependent binding of a nuclear-encoded protein complex (RB38, RB47, RB55 and RB60) to the 5'-untranslated region of the RNA. Despite the absence of any report identifying a red light photoreceptor within this alga, we show that the expression of the rb38, rb47 and rb60 genes, as well as the nuclear-encoded psbO gene that directs the synthesis of OEE1 (oxygen evolving enhancer 1), is differentially regulated by red light. Further elucidation of the signal transduction pathway shows that calmodulin is an important messenger in the signaling cascade that leads to the expression of rb38, rb60 and psbO, and that a chloroplast signal affects rb47 at the translational level. While there may be several factors involved in the cascade of events from the perception of red light to the expression of the rb and psbO genes, our data suggest the involvement of a red light photoreceptor. Future studies will elucidate this receptor and the additional components of this red light signaling expression pathway in C. reinhardtii.


Asunto(s)
Proteínas Algáceas/metabolismo , Calmodulina/metabolismo , Chlamydomonas reinhardtii/genética , Luz , Complejo de Proteína del Fotosistema II/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Algáceas/genética , Chlamydomonas reinhardtii/metabolismo , Chlamydomonas reinhardtii/efectos de la radiación , Cloroplastos/metabolismo , Regulación de la Expresión Génica , Fotosíntesis , Complejo de Proteína del Fotosistema II/genética , ARN de Algas/genética , Proteínas de Unión al ARN/genética , Transducción de Señal
17.
Nanomedicine ; 6(2): 382-90, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19836468

RESUMEN

The goal of this study was to evaluate the mechanism of cyclodextrin-based nanoparticle (CDP-NP) uptake into a murine glioma model. Using mixed in vitro culture systems, we demonstrated that CDP-NPs were preferentially taken up by BV2 and N9 microglia (MG) cells compared with GL261 glioma cells. Fluorescent microscopy and flow cytometry analysis of intracranial GL261 gliomas confirmed these findings and demonstrated a predominant CDP-NP uptake by macrophages (MPs) and MG within and around the tumor site. Notably, in mice bearing bilateral intracranial tumor, MG and MPs carrying CDP-NPs were able to migrate to the contralateral tumors. In conclusion, these studies better characterize the cellular distribution of CDP-NPs in intracranial tumors and demonstrate that MPs and MG could potentially be used as nanoparticle drug carriers into malignant brain tumors. FROM THE CLINICAL EDITOR: The goal of this study was to evaluate the mechanism of cyclodextrin-based nanoparticle (CDP-NP) uptake into a murine glioma model. CDP-NP was preferentially taken up microglia (MG) cells as compared to glioma cells. A predominant CDP-NP uptake by macrophages and MG was also shown in and around the tumor site. Macrophages and MG could potentially be used as nanoparticle drug carriers into malignant brain tumors.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Ciclodextrinas/química , Ciclodextrinas/farmacocinética , Portadores de Fármacos/química , Glioma/metabolismo , Macrófagos/metabolismo , Nanopartículas/química , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioma/patología , Ratones , Nanopartículas/uso terapéutico
18.
Clin Cancer Res ; 26(11): 2740-2753, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32019861

RESUMEN

PURPOSE: Unlike most cancers, no clear epidemiological correlation between diabetes (Db) and malignant glioma progression exists. Because hyperglycemia activates proinflammatory pathways through the receptor for advanced glycation endproducts (RAGE), we hypothesized that Db can also promote malignant glioma progression. EXPERIMENTAL DESIGN: We compared the growth of two phenotypically diverse syngeneic glioma models in control and diabetic mice. Tumor growth and antitumor immune responses were evaluated in orthotopic and heterotopic models and correlated to RAGE and RAGE ligand expression. RESULTS: Irrespective of tumor implantation site, growth of a "classical" glioma model, GL261, increased in hyperglycemic mice and was mediated by upregulation of RAGE and its ligand, HMGB1. However, growth of a "mesenchymal" glioma subtype, K-Luc, depended on tumor implantation site. Whereas heterotopic K-Luc tumors progressed rapidly in Db mice, intracranial K-Luc tumors grew slower. We further showed that hyperglycemia inhibited the innate antitumor inflammatory responses in both models. Although this contributed to the accelerated growth of heterotopic tumors, suppression of tumor inflammatory responses dampened the growth of orthotopic K-Luc gliomas. CONCLUSIONS: Hyperglycemia may enhance glioma growth through promotion of RAGE expression and suppression of antitumor immune responses. However, abrogation of the proinflammatory milieu in tumors may also dampen the growth of inflammatory glioma subtypes in the brains of diabetic mice. This dichotomy in glioma growth response to hyperglycemia may partly explain why conflicting epidemiological studies show both an increased risk and a protective effect of Db in patients with malignant gliomas.


Asunto(s)
Neoplasias Encefálicas/patología , Diabetes Mellitus Experimental/fisiopatología , Glioma/patología , Hiperglucemia/complicaciones , Inmunidad Innata/inmunología , Animales , Apoptosis , Neoplasias Encefálicas/etiología , Movimiento Celular , Proliferación Celular , Glioma/etiología , Humanos , Hiperglucemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Invasividad Neoplásica , Pronóstico , Células Tumorales Cultivadas
19.
Sci Transl Med ; 12(533)2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32132216

RESUMEN

Although chimeric antigen receptor (CAR) T cells have demonstrated signs of antitumor activity against glioblastoma (GBM), tumor heterogeneity remains a critical challenge. To achieve broader and more effective GBM targeting, we developed a peptide-bearing CAR exploiting the GBM-binding potential of chlorotoxin (CLTX). We find that CLTX peptide binds a great proportion of tumors and constituent tumor cells. CAR T cells using CLTX as the targeting domain (CLTX-CAR T cells) mediate potent anti-GBM activity and efficiently target tumors lacking expression of other GBM-associated antigens. Treatment with CLTX-CAR T cells resulted in tumor regression in orthotopic xenograft GBM tumor models. CLTX-CAR T cells do not exhibit observable off-target effector activity against normal cells or after adoptive transfer into mice. Effective targeting by CLTX-CAR T cells requires cell surface expression of matrix metalloproteinase-2. Our results pioneer a peptide toxin in CAR design, expanding the repertoire of tumor-selective CAR T cells with the potential to reduce antigen escape.


Asunto(s)
Glioblastoma , Venenos de Escorpión , Animales , Línea Celular Tumoral , Glioblastoma/terapia , Inmunoterapia Adoptiva , Metaloproteinasa 2 de la Matriz , Ratones , Receptores de Antígenos de Linfocitos T , Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto
20.
JAMA Oncol ; 6(12): 1947-1951, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33030521

RESUMEN

IMPORTANCE: Little is known about the penetration and bioactivity of systemically administered programmed cell death 1 (PD-1) antibodies in the central nervous system. Such information is critical for advancing checkpoint antibody therapies for treatment of brain tumors. OBJECTIVE: To evaluate pembrolizumab concentrations and PD-1 blockade on T cells in the cerebrospinal fluid (CSF) after intravenous administration. DESIGN, SETTING, AND PARTICIPANTS: Cerebrospinal fluid and blood samples were collected from 10 adult patients with high-grade gliomas who were participating in clinical trials of intracranially administered chimeric antigen receptor (CAR) T cells and intravenous pembrolizumab at City of Hope in Duarte, California, from 2017 through 2019. Neuropharmacokinetic and immunologic correlative studies were performed on CSF and serum samples. INTERVENTIONS OR EXPOSURES: Pembrolizumab, 200 mg, was given intravenously every 3 weeks with a median of 2 cycles (range, 1-8). CAR T cells were administered intracranially every 1 to 4 weeks. Cerebrospinal fluid and blood samples were collected on the day of CAR T-cell administration and then 24 hours later for a total of 100 paired samples. MAIN OUTCOMES AND MEASURES: Pembrolizumab concentrations were measured by enzyme-linked immunosorbent assay, PD-1 blocking on T cells by flow cytometry, and results of PD-1 blockade on CAR T-cell function by in vitro tumor rechallenge assays. RESULTS: Of the 10 patients included in this study, the mean (SD) age was 45.7 (11.0) years, and 6 (60%) were women. Steady-state pembrolizumab concentrations in the CSF were achieved by 24 hours after initial intravenous administration, with a mean CSF:serum ratio of 0.009 (95% CI, 0.004-0.014). The CSF concentrations of pembrolizumab effectively blocked PD-1 on both endogenous T cells and intracranially administered CAR T cells in the CSF, with flow cytometric detection of surface PD-1 on the T cells decreasing from a mean (SD) of 39.3% (20.2%) before pembrolizumab to a mean (SD) of 3.8% (5.8%) 24 hours after pembrolizumab infusion. Steady-state concentrations in the CSF were maintained throughout the 21-day cycle of pembrolizumab, as was the PD-1 blocking effect, evidenced by no increase in detectable surface PD-1 on T cells in the CSF during that time period. Incubation of PD-1-expressing T cells with CSF samples from patients treated with pembrolizumab also resulted in PD-1 blockade. CONCLUSIONS AND RELEVANCE: Results of this study demonstrate steady-state concentrations of pembrolizumab in CSF after intravenous administration as well as CSF concentrations that are sufficient for blocking PD-1 on endogenous and adoptively transferred T cells. This provides mechanistic insight regarding the ability of systemically administered PD-1 blocking antibodies to modulate T-cell activity in the brain.


Asunto(s)
Neoplasias , Receptor de Muerte Celular Programada 1 , Femenino , Humanos , Inmunoterapia/métodos , Recuento de Linfocitos , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA