Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
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
3.
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
4.
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.

6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Cancer Immunol Res ; 7(5): 759-772, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30890531

RESUMEN

Improvements in the quality and fitness of chimeric antigen receptor (CAR)-engineered T cells, through CAR design or manufacturing optimizations, could enhance the therapeutic potential of CAR-T cells. One parameter influencing the effectiveness of CAR-T cell therapy is the differentiation status of the final product: CAR-T cells that are less-differentiated and less exhausted are more therapeutically effective. In the current study, we demonstrate that CAR-T cells expanded in IL15 (CAR-T/IL15) preserve a less-differentiated stem cell memory (Tscm) phenotype, defined by expression of CD62L+CD45RA+ CCR7+, as compared with cells cultured in IL2 (CAR-T/IL2). CAR-T/IL15 cells exhibited reduced expression of exhaustion markers, higher antiapoptotic properties, and increased proliferative capacity upon antigen challenge. Furthermore, CAR-T/IL15 cells exhibited decreased mTORC1 activity, reduced expression of glycolytic enzymes and improved mitochondrial fitness. CAR-T/IL2 cells cultured in rapamycin (mTORC1 inhibitor) shared phenotypic features with CAR-T/IL15 cells, suggesting that IL15-mediated reduction of mTORC1 activity is responsible for preserving the Tscm phenotype. CAR-T/IL15 cells promoted superior antitumor responses in vivo in comparison with CAR-T/IL2 cells. Inclusion of cytokines IL7 and/or IL21 in addition to IL15 reduced the beneficial effects of IL15 on CAR-T phenotype and antitumor potency. Our findings show that IL15 preserves the CAR-T cell Tscm phenotype and improves their metabolic fitness, which results in superior in vivo antitumor activity, thus opening an avenue that may improve future adoptive T-cell therapies.


Asunto(s)
Inmunoterapia Adoptiva , Interleucina-15/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Células Madre/inmunología , Linfocitos T/trasplante , Animales , Línea Celular Tumoral , Humanos , Memoria Inmunológica , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Fenotipo , Linfocitos T/inmunología
14.
J Vis Exp ; (144)2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30882787

RESUMEN

The field of chimeric antigen receptor (CAR) T cell therapy is rapidly advancing with improvements in CAR design, gene-engineering approaches and manufacturing optimizations. One challenge for these development efforts, however, has been the establishment of in vitro assays that can robustly inform selection of the optimal CAR T cell products for in vivo therapeutic success. Standard in vitro tumor-lysis assays often fail to reflect the true antitumor potential of the CAR T cells due to the relatively short co-culture time and high T cell to tumor ratio. Here, we describe an in vitro co-culture method to evaluate CAR T cell recursive killing potential at high tumor cell loads. In this assay, long-term cytotoxic function and proliferative capacity of CAR T cells is examined in vitro over 7 days with additional tumor targets administered to the co-culture every other day. This assay can be coupled with profiling T cell activation, exhaustion and memory phenotypes. Using this assay, we have successfully distinguished the functional and phenotypic differences between CD4+ and CD8+ CAR T cells against glioblastoma (GBM) cells, reflecting their differential in vivo antitumor activity in orthotopic xenograft models. This method provides a facile approach to assess CAR T cell potency and to elucidate the functional variations across different CAR T cell products.


Asunto(s)
Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Glioblastoma/terapia , Humanos , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos , Linfocitos T/inmunología
15.
JCI Insight ; 3(10)2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29769444

RESUMEN

Chimeric antigen receptor-modified (CAR-modified) T cells have shown promising therapeutic effects for hematological malignancies, yet limited and inconsistent efficacy against solid tumors. The refinement of CAR therapy requires an understanding of the optimal characteristics of the cellular products, including the appropriate composition of CD4+ and CD8+ subsets. Here, we investigated the differential antitumor effect of CD4+ and CD8+ CAR T cells targeting glioblastoma-associated (GBM-associated) antigen IL-13 receptor α2 (IL13Rα2). Upon stimulation with IL13Rα2+ GBM cells, the CD8+ CAR T cells exhibited robust short-term effector function but became rapidly exhausted. By comparison, the CD4+ CAR T cells persisted after tumor challenge and sustained their effector potency. Mixing with CD4+ CAR T cells failed to ameliorate the effector dysfunction of CD8+ CAR T cells, while surprisingly, CD4+ CAR T cell effector potency was impaired when coapplied with CD8+ T cells. In orthotopic GBM models, CD4+ outperformed CD8+ CAR T cells, especially for long-term antitumor response. Further, maintenance of the CD4+ subset was positively correlated with the recursive killing ability of CAR T cell products derived from GBM patients. These findings identify CD4+ CAR T cells as a highly potent and clinically important T cell subset for effective CAR therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Glioblastoma/patología , Humanos
16.
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
17.
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
18.
PLoS One ; 11(12): e0165118, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27936099

RESUMEN

Microglia (MG) and macrophages (MPs) represent a significant component of the inflammatory response to gliomas. When activated, MG/MP release a variety of pro-inflammatory cytokines, however, they also secrete anti-inflammatory factors that limit their cytotoxic function. The balance between pro and anti-inflammatory functions dictates their antitumor activity. To evaluate potential variations in MG and MP function in gliomas, we isolated these cells (and other Gr1+ cells) from intracranial GL261 murine gliomas by FACS and evaluated their gene expression profiles by microarray analysis. As expected, arginase 1 (Arg1, M2 marker) was highly expressed by tumor-associated Gr1+, MG and MP. However, in contrast to MP and Gr1+ cells that expressed Arg1 shortly after tumor trafficking, Arg1 expression in MG was delayed and occurred in larger tumors. Interestingly, depletion of MPs in tumors did not prevent MG polarization, suggesting direct influence of tumor-specific factors on MG Arg1 upregulation. Finally, Arg1 expression was confirmed in human GBM samples, but most Arg1+ cells were neutrophils and not MPs. These findings confirm variations in tumor MG and MP polarization states and its dependency on tumor microenvironmental factors.


Asunto(s)
Arginasa/genética , Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Macrófagos/metabolismo , Microglía/metabolismo , Proteínas de Neoplasias/genética , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Arginasa/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Humanos , Activación de Macrófagos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Especificidad de Órganos , Cultivo Primario de Células , Transducción de Señal , Factores de Tiempo , Microambiente Tumoral/genética
19.
PLoS One ; 11(2): e0148139, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26829221

RESUMEN

Even when treated with aggressive current therapies, most patients with glioblastoma survive less than two years. Rapid tumor growth, an invasive nature, and the blood-brain barrier, which limits the penetration of large molecules into the brain, all contribute to the poor tumor response associated with conventional therapies. Immunotherapy has emerged as a therapeutic approach that may overcome these challenges. We recently reported that single-walled carbon nanotubes (SWCNTs) can be used to dramatically increase the immunotherapeutic efficacy of CpG oligonucleotides in a mouse model of glioma. Following implantation in the mouse brain, the tumor cell line used in these previous studies (GL261) tends to form a spherical tumor with limited invasion into healthy brain. In order to evaluate SWCNT/CpG therapy under more clinically-relevant conditions, here we report the treatment of a more invasive mouse glioma model (K-Luc) that better recapitulates human disease. In addition, a CpG sequence previously tested in humans was used to formulate the SWCNT/CpG which was combined with temozolomide, the standard of care chemotherapy for glioblastoma patients. We found that, following two intracranial administrations, SWCNT/CpG is well-tolerated and improves the survival of mice bearing invasive gliomas. Interestingly, the efficacy of SWCNT/CpG was enhanced when combined with temozolomide. This enhanced anti-tumor efficacy was correlated to an increase of tumor-specific cytotoxic activity in splenocytes. These results reinforce the emerging understanding that immunotherapy can be enhanced by combining it with chemotherapy and support the continued development of SWCNT/CpG.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Inmunoterapia , Nanotubos de Carbono/química , Oligodesoxirribonucleótidos/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Glioma/patología , Inflamación/patología , Lípidos/química , Ratones Endogámicos C57BL , Invasividad Neoplásica , Polietilenglicoles/química , Bazo/patología , Temozolomida , Resultado del Tratamiento
20.
Methods Mol Biol ; 1371: 101-13, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26530797

RESUMEN

CD4(+) T helper (Th) lymphocytes are essential elements of the complex cellular networks regulating the initiation, development, and termination of adaptive immune responses. Different independent and specialized subsets of Th cells can be distinguished based on their dedicated transcription factor and cytokine expression profiles. Th17 lymphocytes have been described about a decade ago as CD4(+) Th cells producing high quantity of IL-17A as a signature cytokine. Since their initial discovery, Th17 have drawn intense scrutiny for their dominant role in the pathogenesis of multiple autoimmune, infectious diseases and allergy. The influence of Th17 lymphocytes in cancer remains however ambiguous. The plethoric functions of Th17 may rely on the remarkable plasticity of these cells, endowed with the ability to trans-differentiate into other Th subpopulations depending on the environmental cytokine context. The possibility to generate Th17 ex vivo has facilitated the elucidation of the signals and transcription factors required for their differentiation and functions and has allowed for the evaluation of their functions following adoptive transfer in vivo. Several protocols have been developed to produce Th17 in vitro. The intent of this chapter is to provide examples of procedures for generating and expanding Th17 ex vivo.


Asunto(s)
Técnicas de Cultivo de Célula , Subgrupos de Linfocitos T/citología , Células Th17/citología , Animales , Separación Celular/métodos , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo/métodos , Humanos , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Subgrupos de Linfocitos T/metabolismo , Células Th17/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA