Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 32(2): 527-539, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38140726

RESUMO

Dexamethasone (dex) is a glucocorticoid that is a mainstay for the treatment of inflammatory pathologies, including immunotherapy-associated toxicities, yet the specific impact of dex on the activity of CAR T cells is not fully understood. We assessed whether dex treatment given ex vivo or as an adjuvant in vivo with CAR T cells impacted the phenotype or function of CAR T cells. We demonstrated that CAR T cell expansion and function were not inhibited by dex. We confirmed this observation using multiple CAR constructs and tumor models, suggesting that this is a general phenomenon. Moreover, we determined that dex upregulated interleukin-7 receptor α on CAR T cells and increased the expression of genes involved in activation, migration, and persistence when supplemented ex vivo. Direct delivery of dex and IL-7 into tumor-bearing mice resulted in increased persistence of adoptively transferred CAR T cells and complete tumor regression. Overall, our studies provide insight into the use of dex to enhance CAR T cell therapy and represent potential novel strategies for augmenting CAR T cell function during production as well as following infusion into patients.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Receptores de Interleucina-7 , Humanos , Animais , Camundongos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Imunoterapia Adotiva/métodos , Neoplasias/patologia , Linfócitos T , Dexametasona/farmacologia
2.
Gastroenterology ; 162(4): 1319-1333, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34999097

RESUMO

BACKGROUND & AIMS: Pancreatic cancer (PC) is the third leading cause of cancer-related death with a 5-year survival rate of approximately 10%. It typically presents as a late-stage incurable cancer and chemotherapy provides modest benefit. Here, we demonstrate the feasibility, safety, and potency of a novel human natural killer (NK) cell-based immunotherapy to treat PC. METHODS: The expression of prostate stem cell antigen (PSCA) was evaluated in primary PC at messenger RNA and protein levels. The processes of retroviral transduction, expansion, activation, and cryopreservation of primary human NK cells obtained from umbilical cord blood were optimized, allowing us to develop frozen, off-the-shelf, allogeneic PSCA chimeric antigen receptor (CAR) NK cells. The safety and efficacy of PSCA CAR NK cells also expressing soluble (s) interleukin 15 (PSCA CAR_s15 NK cells) were evaluated in vitro and in vivo. RESULTS: PSCA was elevated in primary human PC compared with the adjacent or other normal tissues. PSCA CAR_s15 NK cells displayed significant tumor-suppressive effects against PSCA(+) PC in vitro before and after 1 cycle of freeze-thaw. The viability of frozen PSCA CAR_s15 NK cells persisted more than 90 days in vivo after their last infusion and significantly prolonged the survival of mice engrafted with human PC. CONCLUSIONS: PSCA CAR_s15 NK cells showed therapeutic efficacy in human metastatic PC models without signs of systematic toxicity, providing a strong rationale to support clinical development.


Assuntos
Neoplasias Pancreáticas , Receptores de Antígenos Quiméricos , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Humanos , Imunoterapia Adotiva , Células Matadoras Naturais , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Neoplasias Pancreáticas/patologia , Próstata , Células-Tronco/metabolismo , Neoplasias Pancreáticas
3.
Mol Ther ; 29(7): 2335-2349, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-33647456

RESUMO

Chimeric antigen receptor (CAR) T cell therapy has led to impressive clinical responses in patients with hematological malignancies; however, its effectiveness in patients with solid tumors has been limited. While CAR T cells for the treatment of advanced prostate and pancreas cancer, including those targeting prostate stem cell antigen (PSCA), are being clinically evaluated and are anticipated to show bioactivity, their safety and the impact of the immunosuppressive tumor microenvironment (TME) have not been faithfully explored preclinically. Using a novel human PSCA knockin (hPSCA-KI) immunocompetent mouse model, we evaluated the safety and therapeutic efficacy of PSCA-CAR T cells. We demonstrated that cyclophosphamide (Cy) pre-conditioning significantly modified the immunosuppressive TME and was required to uncover the efficacy of PSCA-CAR T cells in metastatic prostate and pancreas cancer models, with no observed toxicities in normal tissues with endogenous expression of PSCA. This combination dampened the immunosuppressive TME, generated pro-inflammatory myeloid and T cell signatures in tumors, and enhanced the recruitment of antigen-presenting cells, as well as endogenous and adoptively transferred T cells, resulting in long-term anti-tumor immunity.


Assuntos
Ciclofosfamida/farmacologia , Imunoterapia Adotiva/métodos , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias Pancreáticas/terapia , Neoplasias da Próstata/terapia , Microambiente Tumoral , Animais , Antígenos de Neoplasias/genética , Apoptose , Proliferação de Células , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Agonistas Mieloablativos/farmacologia , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Treat Res ; 180: 297-326, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32215875

RESUMO

The past two decades have marked the beginning of an unprecedented success story for cancer therapy through redirecting antitumor immunity [1]. While the mechanisms that control the initial and ongoing immune responses against tumors remain a strong research focus, the clinical development of technologies that engage the immune system to target and kill cancer cells has become a translational research priority. Early attempts documented in the late 1800s aimed at sparking immunity with cancer vaccines were difficult to interpret but demonstrated an opportunity that more than 100 years later has blossomed into the current field of cancer immunotherapy. Perhaps the most recent and greatest illustration of this is the widespread appreciation that tumors actively shut down antitumor immunity, which has led to the emergence of checkpoint pathway inhibitors that re-invigorate the body's own immune system to target cancer [2, 3]. This class of drugs, with first FDA approvals in 2011, has demonstrated impressive durable clinical responses in several cancer types, including melanoma, lung cancer, Hodgkin's lymphoma, and renal cell carcinoma, with the ongoing investigation in others. The biology and ultimate therapeutic successes of these drugs led to the 2018 Nobel Prize in Physiology or Medicine, awarded to Dr. James Allison and Dr. Tasuku Honjo for their contributions to cancer therapy [4]. In parallel to the emerging science that aided in unleashing the body's own antitumor immunity with checkpoint pathway inhibitors, researchers were also identifying ways to re-engineer antitumor immunity through adoptive cellular immunotherapy approaches. Chimeric antigen receptor (CAR)-based T cell therapy has achieved an early head start in the field, with two recent FDA approvals in 2017 for the treatment of B-cell malignancies [5]. There is an explosion of preclinical and clinical efforts to expand the therapeutic indications for CAR T cell therapies, with a specific focus on improving their clinical utility, particularly for the treatment of solid tumors. In this chapter, we will highlight the recent progress, challenges, and future perspectives surrounding the development of CAR T cell therapies for solid tumors.


Assuntos
Vacinas Anticâncer , Imunoterapia Adotiva , Neoplasias/terapia , Humanos
5.
N Engl J Med ; 375(26): 2561-9, 2016 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-28029927

RESUMO

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 .).


Assuntos
Linfócitos T CD8-Positivos/imunologia , Glioblastoma/terapia , Imunoterapia Adotiva , Recidiva Local de Neoplasia/terapia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Engenharia Celular , Terapia Combinada , Humanos , Subunidade alfa2 de Receptor de Interleucina-13 , Masculino , Pessoa de Meia-Idade
6.
Cancer Treat Res ; 178: 189-205, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31209846

RESUMO

Repairing defects in anti-tumor immunity has been a longstanding challenge in cancer therapy, and in recent years, immunotherapy has emerged as a promising approach for treating advanced disease. While the interactions between the immune system and cancer have been studied for more than a century, only in recent years has the field realized the tremendous potential in stimulating the immune system to eradicate cancer. From early investigations by William Coley in using bacteria to treat cancer patients to more recent work in adoptively transferred engineered T cells to identify and kill cancer cells has opened up an entire field dedicated to re-educating the immune system in a cancer patient. A multitude of immunotherapy strategies have been proposed and tested in clinical trials, from recombinant proteins, agonistic antibodies, and checkpoint inhibitors designed to re-invigorate anti-tumor immunity, to vaccine approaches and adoptive T-cell strategies, we are now on the cusp of an exciting revolution that will ultimately become an arsenal of therapies to treat any cancer type, at any stage, with the hope of robust and durable responses in cancer patients. In this chapter, we will examine the various immunotherapy strategies under active clinical investigation, with a particular focus on the latest advances in cellular immunotherapies and the future of precision medicine-enabled immunotherapy.


Assuntos
Vacinas Anticâncer , Imunoterapia , Neoplasias , Medicina de Precisão , Humanos , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T
7.
Prostate ; 77(2): 123-144, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27679977

RESUMO

INTRODUCTION: The 2016 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, "Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer," was held from June 23 to June 26, 2016, in Coronado, California. METHODS: For the 4th year in a row, the Prostate Cancer Foundation (PCF) hosted the CHPCA Meeting, a think tank-structured scientific conference, which focuses on a specific topic of critical unmet need on the biology and treatment of advanced prostate cancer. The 2016 CHPCA Meeting was attended by 71 investigators from prostate cancer and other fields, who discussed the biology, study methodologies, treatment strategies, and critical unmet needs concerning metastatic prostate cancer, with the ultimate goal of advancing strategies to treat and eliminate this disease. RESULTS: The major topics of discussion included: the molecular landscape and molecular heterogeneity of metastatic prostate cancer, the role of the metastatic microenvironment, optimizing immunotherapy in metastatic prostate cancer, learning from exceptional responders and non-responders, targeting DNA repair deficiency in advanced prostate cancer, developing and applying novel biomarkers and imaging techniques, and potential roles for the microbiome in prostate cancer. DISCUSSION: This article reviews the topics presented and discussions held at the CHPCA Meeting, with a focus on the unknowns and next steps needed to advance our understanding of the biology and most effective treatment strategies for metastatic prostate cancer. Prostate 77:123-144, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Academias e Institutos/tendências , Antineoplásicos/administração & dosagem , Congressos como Assunto/tendências , Imunoterapia/tendências , Neoplasias da Próstata/terapia , Relatório de Pesquisa/tendências , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , California , Compreensão , Sistemas de Liberação de Medicamentos/métodos , Sistemas de Liberação de Medicamentos/tendências , Humanos , Imunoterapia/métodos , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/fisiologia
8.
Eur J Immunol ; 45(1): 71-81, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25310972

RESUMO

Increasing evidence suggests that premetastatic niches, consisting mainly of myeloid cells, provide microenvironment critical for cancer cell recruitment and survival to facilitate metastasis. While CD8(+) T cells exert immunosurveillance in primary human tumors, whether they can exert similar effects on myeloid cells in the premetastatic environment is unknown. Here, we show that CD8(+) T cells are capable of constraining premetastatic myeloid cell accumulation by inducing myeloid cell apoptosis in C57BL/6 mice. Ag-specific CD8(+) T-cell cytotoxicity against myeloid cells in premetastatic lymph nodes is compromised by Stat3. We demonstrate here that Stat3 ablation in myeloid cells leads to CD8(+) T-cell activation and increased levels of IFN-γ and granzyme B in the premetastatic environment. Furthermore, Stat3 negatively regulates soluble Ag cross-presentation by myeloid cells to CD8(+) T cells in the premetastatic niche. Importantly, in tumor-free lymph nodes of melanoma patients, infiltration of activated CD8(+) T cells inversely correlates with STAT3 activity, which is associated with a decrease in number of myeloid cells. Our study suggested a novel role for CD8(+) T cells in constraining myeloid cell activity through direct killing in the premetastatic environment, and the therapeutic potential by targeting Stat3 in myeloid cells to improve CD8(+) T-cell immunosurveillance against metastasis.


Assuntos
Regulação Neoplásica da Expressão Gênica/imunologia , Vigilância Imunológica , Macrófagos Peritoneais/imunologia , Melanoma Experimental/imunologia , Neoplasias Cutâneas/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Proliferação de Células , Granzimas/genética , Granzimas/imunologia , Humanos , Interferon gama/genética , Interferon gama/imunologia , Linfonodos/imunologia , Linfonodos/patologia , Metástase Linfática , Macrófagos Peritoneais/patologia , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias , Cultura Primária de Células , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Linfócitos T Citotóxicos/patologia
9.
Mol Ther ; 23(4): 757-68, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25366031

RESUMO

The success of adoptive therapy using chimeric antigen receptor (CAR)-expressing T cells partly depends on optimal CAR design. CARs frequently incorporate a spacer/linker region based on the constant region of either IgG1 or IgG4 to connect extracellular ligand-binding with intracellular signaling domains. Here, we evaluated the potential for the IgG4-Fc linker to result in off-target interactions with Fc gamma receptors (FcγRs). As proof-of-principle, we focused on a CD19-specific scFv-IgG4-CD28-zeta CAR and found that, in contrast to CAR-negative cells, CAR+ T cells bound soluble FcγRs in vitro and did not engraft in NSG mice. We hypothesized that mutations to avoid FcγR binding would improve CAR+ T cell engraftment and antitumor efficacy. Thus, we generated CD19-specific CARs with IgG4-Fc spacers that had either been mutated at two sites (L235E; N297Q) within the CH2 region (CD19R(EQ)) or incorporated a CH2 deletion (CD19Rch2Δ). These mutations reduced binding to soluble FcγRs without altering the ability of the CAR to mediate antigen-specific lysis. Importantly, CD19R(EQ) and CD19Rch2Δ T cells exhibited improved persistence and more potent CD19-specific antilymphoma efficacy in NSG mice. Together, these studies suggest that optimal CAR function may require the elimination of cellular FcγR interactions to improve T cell persistence and antitumor responses.


Assuntos
Imunoglobulina G/imunologia , Proteínas Mutantes Quiméricas/metabolismo , Mutação , Neoplasias Experimentais/terapia , Receptores de Antígenos/metabolismo , Receptores Fc/metabolismo , Linfócitos T/imunologia , Animais , Imunoterapia , Camundongos , Neoplasias Experimentais/imunologia , Ligação Proteica , Receptores de Antígenos/genética
10.
Proc Natl Acad Sci U S A ; 110(32): 13079-84, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23878227

RESUMO

Dysregulated inflammation in adipose tissue, marked by increased proinflammatory T-cell accumulation and reduced regulatory T cells (Tregs), contributes to obesity-associated insulin resistance. The molecular mechanisms underlying T-cell-mediated inflammation in adipose tissue remain largely unknown, however. Here we show a crucial role for signal transducer and activator of transcription 3 (Stat3) in T cells in skewing adaptive immunity in visceral adipose tissue (VAT), thereby contributing to diet-induced obesity (DIO) and insulin resistance. Stat3 activity is elevated in obese VAT and in VAT-resident T cells. Functional ablation of Stat3 in T cells reduces DIO, improves insulin sensitivity and glucose tolerance, and suppresses VAT inflammation. Importantly, Stat3 ablation reverses the high Th1/Treg ratio in VAT of DIO mice that is likely secondary to elevated IL-6 production, leading in turn to suppression of Tregs. In addition, Stat3 in T cells in DIO mice affects adipose tissue macrophage accumulation and M2 phenotype. Our study identifies Stat3 in VAT-resident T cells as an important mediator and direct target for regulating adipose tissue inflammation, DIO, and its associated metabolic dysfunctions.


Assuntos
Resistência à Insulina/imunologia , Gordura Intra-Abdominal/imunologia , Obesidade/imunologia , Fator de Transcrição STAT3/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Glicemia/metabolismo , Western Blotting , Dieta Hiperlipídica/efeitos adversos , Jejum/sangue , Feminino , Citometria de Fluxo , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Insulina/sangue , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/sangue , Obesidade/etiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo
11.
Curr Opin Oncol ; 27(6): 466-74, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26352543

RESUMO

PURPOSE OF REVIEW: Chimeric antigen receptors (CARs) are synthetic immunoreceptors, which can redirect T cells to selectively kill tumor cells, and as 'living drugs' have the potential to generate long-term antitumor immunity. Given their recent clinical successes for the treatment of refractory B-cell malignancies, there is a strong push toward advancing this immunotherapy to other hematological diseases and solid cancers. Here, we summarize the current state of the field, highlighting key variables for the optimal application of CAR T cells for cancer immunotherapy. RECENT FINDINGS: Advances in CAR T-cell therapy have highlighted intrinsic CAR design and T-cell manufacturing methods as critical components for maximal therapeutic success. Similarly, addressing the unique extrinsic challenges of each tumor type, including overcoming the immunosuppressive tumor microenvironment and tumor heterogeneity, and mitigating potential toxicity, will dominate the next wave of CAR T-cell development. SUMMARY: CAR T-cell therapeutic optimization, including intrinsic and extrinsic factors, is critical to developing effective CAR T-cell therapies for cancer. The excitement of CAR T-cell immunotherapy has just begun, and will continue with new insights revealed in laboratory research and in ongoing clinical investigations.


Assuntos
Imunoterapia/métodos , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Linfócitos T/imunologia , Diferenciação Celular , Humanos , Neoplasias/imunologia , Microambiente Tumoral/imunologia
12.
J Am Coll Surg ; 238(4): 436-447, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38214445

RESUMO

BACKGROUND: Chimeric antigen receptor (CAR) T cells targeting the B-cell antigen CD19 are standard therapy for relapsed or refractory B-cell lymphoma and leukemia. CAR T cell therapy in solid tumors is limited due to an immunosuppressive tumor microenvironment and a lack of tumor-restricted antigens. We recently engineered an oncolytic virus (CF33) with high solid tumor affinity and specificity to deliver a nonsignaling truncated CD19 antigen (CD19t), allowing targeting by CD19-CAR T cells. Here, we tested this combination against pancreatic cancer. STUDY DESIGN: We engineered CF33 to express a CD19t (CF33-CD19t) target. Flow cytometry and ELISA were performed to quantify CD19t expression, immune activation, and killing by virus and CD19-CAR T cells against various pancreatic tumor cells. Subcutaneous pancreatic human xenograft tumor models were treated with virus, CAR T cells, or virus+CAR T cells. RESULTS: In vitro, CF33-CD19t infection of tumor cells resulted in >90% CD19t cell-surface expression. Coculturing CD19-CAR T cells with infected cells resulted in interleukin-2 and interferon gamma secretion, upregulation of T-cell activation markers, and synergistic cell killing. Combination therapy of virus+CAR T cells caused significant tumor regression (day 13): control (n = 16, 485 ± 20 mm 3 ), virus alone (n = 20, 254 ± 23 mm 3 , p = 0.0001), CAR T cells alone (n = 18, 466 ± 25 mm 3 , p = NS), and virus+CAR T cells (n = 16, 128 ± 14 mm 3 , p < 0.0001 vs control; p = 0.0003 vs virus). CONCLUSIONS: Engineered CF33-CD19t effectively infects and expresses CD19t in pancreatic tumors, triggering cell killing and increased immunogenic response by CD19-CAR T cells. Notably, CF33-CD19t can turn cold immunologic tumors hot, enabling solid tumors to be targetable by agents designed against liquid tumor antigens.


Assuntos
Vírus Oncolíticos , Neoplasias Pancreáticas , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Linfócitos T/metabolismo , Linfócitos T/transplante , Antígenos CD19/metabolismo , Neoplasias Pancreáticas/terapia , Microambiente Tumoral
13.
Mol Ther Oncol ; 32(2): 200789, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38939825

RESUMO

Advancing chimeric antigen receptor (CAR)-engineered T cells for the treatment of solid tumors is a major focus in the field of cellular immunotherapy. Several hurdles have hindered similar CAR T cell clinical responses in solid tumors as seen in hematological malignancies. These challenges include on-target off-tumor toxicities, which have inspired efforts to optimize CARs for improved tumor antigen selectivity and overall safety. We recently developed a CAR T cell therapy targeting prostate stem cell antigen (PSCA) for prostate and pancreatic cancers, showing improved preclinical antitumor activity and T cell persistence by optimizing the intracellular co-stimulatory domain. Similar studies were undertaken to optimize HER2-directed CAR T cells with modifications to the intracellular co-stimulatory domain for selective targeting of breast cancer brain metastasis. In the present study, we evaluate various nonsignaling extracellular spacers in these CARs to further improve tumor antigen selectivity. Our findings suggest that length and structure of the extracellular spacer can dictate the ability of CARs to selectively target tumor cells with high antigen density, while sparing cells with low antigen density. This study contributes to CAR construct design considerations and expands our knowledge of tuning solid tumor CAR T cell therapies for improved safety and efficacy.

14.
Elife ; 132024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38251974

RESUMO

Chromatin-associated RNAs (caRNAs) form a relatively poorly recognized layer of the epigenome. The caRNAs reported to date are transcribed from the nuclear genome. Here, leveraging a recently developed assay for detection of caRNAs and their genomic association, we report that mitochondrial RNAs (mtRNAs) are attached to the nuclear genome and constitute a subset of caRNA, thus termed mt-caRNA. In four human cell types analyzed, mt-caRNAs preferentially attach to promoter regions. In human endothelial cells (ECs), the level of mt-caRNA-promoter attachment changes in response to environmental stress that mimics diabetes. Suppression of a non-coding mt-caRNA in ECs attenuates stress-induced nascent RNA transcription from the nuclear genome, including that of critical genes regulating cell adhesion, and abolishes stress-induced monocyte adhesion, a hallmark of dysfunctional ECs. Finally, we report increased nuclear localization of multiple mtRNAs in the ECs of human diabetic donors, suggesting many mtRNA translocate to the nucleus in a cell stress and disease-dependent manner. These data nominate mt-caRNAs as messenger molecules responsible for mitochondrial-nuclear communication and connect the immediate product of mitochondrial transcription with the transcriptional regulation of the nuclear genome.


Assuntos
Células Endoteliais , RNA , Humanos , RNA Mitocondrial/genética , Cromatina , Bioensaio
15.
Nat Med ; 30(6): 1636-1644, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38867077

RESUMO

Despite recent therapeutic advances, metastatic castration-resistant prostate cancer (mCRPC) remains lethal. Chimeric antigen receptor (CAR) T cell therapies have demonstrated durable remissions in hematological malignancies. We report results from a phase 1, first-in-human study of prostate stem cell antigen (PSCA)-directed CAR T cells in men with mCRPC. The starting dose level (DL) was 100 million (M) CAR T cells without lymphodepletion (LD), followed by incorporation of LD. The primary end points were safety and dose-limiting toxicities (DLTs). No DLTs were observed at DL1, with a DLT of grade 3 cystitis encountered at DL2, resulting in addition of a new cohort using a reduced LD regimen + 100 M CAR T cells (DL3). No DLTs were observed in DL3. Cytokine release syndrome of grade 1 or 2 occurred in 5 of 14 treated patients. Prostate-specific antigen declines (>30%) occurred in 4 of 14 patients, as well as radiographic improvements. Dynamic changes indicating activation of peripheral blood endogenous and CAR T cell subsets, TCR repertoire diversity and changes in the tumor immune microenvironment were observed in a subset of patients. Limited persistence of CAR T cells was observed beyond 28 days post-infusion. These results support future clinical studies to optimize dosing and combination strategies to improve durable therapeutic outcomes. ClinicalTrials.gov identifier NCT03873805 .


Assuntos
Antígenos de Neoplasias , Proteínas Ligadas por GPI , Imunoterapia Adotiva , Proteínas de Neoplasias , Neoplasias de Próstata Resistentes à Castração , Humanos , Masculino , Neoplasias de Próstata Resistentes à Castração/terapia , Neoplasias de Próstata Resistentes à Castração/imunologia , Neoplasias de Próstata Resistentes à Castração/patologia , Idoso , Pessoa de Meia-Idade , Antígenos de Neoplasias/imunologia , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Proteínas Ligadas por GPI/imunologia , Proteínas de Neoplasias/imunologia , Receptores de Antígenos Quiméricos/imunologia , Metástase Neoplásica , Linfócitos T/imunologia , Linfócitos T/transplante , Antígeno Prostático Específico/sangue
16.
Cancers (Basel) ; 15(23)2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-38067366

RESUMO

Precision immune oncology capitalizes on identifying and targeting tumor-specific antigens to enhance anti-tumor immunity and improve the treatment outcomes of solid tumors. Gastric cancer (GC) is a molecularly heterogeneous disease where monoclonal antibodies against human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor (VEGF), and programmed cell death 1 (PD-1) combined with systemic chemotherapy have improved survival in patients with unresectable or metastatic GC. However, intratumoral molecular heterogeneity, variable molecular target expression, and loss of target expression have limited antibody use and the durability of response. Often immunogenically "cold" and diffusely spread throughout the peritoneum, GC peritoneal carcinomatosis (PC) is a particularly challenging, treatment-refractory entity for current systemic strategies. More adaptable immunotherapeutic approaches, such as oncolytic viruses (OVs) and chimeric antigen receptor (CAR) T cells, have emerged as promising GC and GCPC treatments that circumvent these challenges. In this study, we provide an up-to-date review of the pre-clinical and clinical efficacy of CAR T cell therapy for key primary antigen targets and provide a translational overview of the types, modifications, and mechanisms for OVs used against GC and GCPC. Finally, we present a novel, summary-based discussion on the potential synergistic interplay between OVs and CAR T cells to treat GCPC.

17.
J Immunother Cancer ; 11(4)2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37019471

RESUMO

BACKGROUND: Gastric cancer (GC) that metastasizes to the peritoneum is fatal. CF33 and its genetically modified derivatives show cancer selectivity and oncolytic potency against various solid tumors. CF33-hNIS and CF33-hNIS-antiPDL1 have entered phase I trials for intratumoral and intravenous treatments of unresectable solid tumors (NCT05346484) and triple-negative breast cancer (NCT05081492). Here, we investigated the antitumor activity of CF33-oncolytic viruses (OVs) against GC and CF33-hNIS-antiPDL1 in the intraperitoneal (IP) treatment of GC peritoneal metastases (GCPM). METHODS: We infected six human GC cell lines AGS, MKN-45, MKN-74, KATO III, SNU-1, and SNU-16 with CF33, CF33-GFP, or CF33-hNIS-antiPDL1 at various multiplicities of infection (0.01, 0.1, 1.0, and 10.0), and performed viral proliferation and cytotoxicity assays. We used immunofluorescence imaging and flow cytometric analysis to verify virus-encoded gene expression. We evaluated the antitumor activity of CF33-hNIS-antiPDL1 following IP treatment (3×105 pfu × 3 doses) in an SNU-16 human tumor xenograft model using non-invasive bioluminescence imaging. RESULTS: CF33-OVs showed dose-dependent infection, replication, and killing of both diffuse and intestinal subtypes of human GC cell lines. Immunofluorescence imaging showed virus-encoded GFP, hNIS, and anti-PD-L1 antibody scFv expression in CF33-OV-infected GC cells. We confirmed GC cell surface PD-L1 blockade by virus-encoded anti-PD-L1 scFv using flow cytometry. In the xenograft model, CF33-hNIS-antiPDL1 (IP; 3×105 pfu × 3 doses) treatment significantly reduced peritoneal tumors (p<0.0001), decreased amount of ascites (62.5% PBS vs 25% CF33-hNIS-antiPDL1) and prolonged animal survival. At day 91, seven out of eight mice were alive in the virus-treated group versus one out of eight in the control group (p<0.01). CONCLUSIONS: Our results show that CF33-OVs can deliver functional proteins and demonstrate effective antitumor activity in GCPM models when delivered intraperitoneally. These preclinical results will inform the design of future peritoneal-directed therapy in GCPM patients.


Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos , Neoplasias Peritoneais , Neoplasias Gástricas , Humanos , Camundongos , Animais , Vírus Oncolíticos/genética , Neoplasias Peritoneais/terapia , Terapia Viral Oncolítica/métodos , Peritônio/patologia , Neoplasias Gástricas/patologia
18.
bioRxiv ; 2023 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-36711615

RESUMO

Chimeric antigen receptor (CAR) T cell therapeutic responses are hampered by limited T cell trafficking, persistence, and durable anti-tumor activity in solid tumor microenvironments. However, these challenges can be largely overcome by relatively unconstrained synthetic engineering strategies, which are being harnessed to improve solid tumor CAR T cell therapies. Here, we describe fully optimized CAR T cells targeting tumor-associated glycoprotein-72 (TAG72) for the treatment of solid tumors, identifying the CD28 transmembrane domain upstream of the 4-1BB co-stimulatory domain as a driver of potent anti-tumor activity and IFNγ secretion. These findings have culminated into a phase 1 trial evaluating safety, feasibility, and bioactivity of TAG72-CAR T cells for the treatment of patients with advanced ovarian cancer ( NCT05225363 ). Preclinically, we found that CAR T cell-mediated IFNγ production facilitated by IL-12 signaling was required for tumor cell killing, which was recapitulated by expressing an optimized membrane-bound IL-12 (mbIL12) molecule on CAR T cells. Critically, mbIL12 cell surface expression and downstream signaling was induced and sustained only following CAR T cell activation. CAR T cells with mbIL12 demonstrated improved antigen-dependent T cell proliferation and potent cytotoxicity in recursive tumor cell killing assays in vitro and showed robust in vivo anti-tumor efficacy in human xenograft models of ovarian cancer peritoneal metastasis. Further, locoregional administration of TAG72-CAR T cells with antigen-dependent IL-12 signaling promoted durable anti-tumor responses against both regional and systemic disease in mice and was associated with improved systemic T cell persistence. Our study features a clinically-applicable strategy to improve the overall efficacy of locoregionally-delivered CAR T cells engineered with antigen-dependent immune-modulating cytokines in targeting both regional and systemic disease.

19.
Nat Commun ; 14(1): 4737, 2023 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-37550294

RESUMO

Chimeric antigen receptor (CAR) T cell therapeutic responses are hampered by limited T cell trafficking, persistence, and durable anti-tumor activity in solid tumors. However, these challenges can be largely overcome by relatively unconstrained synthetic engineering strategies. Here, we describe CAR T cells targeting tumor-associated glycoprotein-72 (TAG72), utilizing the CD28 transmembrane domain upstream of the 4-1BB co-stimulatory domain as a driver of potent anti-tumor activity and IFNγ secretion. CAR T cell-mediated IFNγ production facilitated by IL-12 signaling is required for tumor cell killing, which is recapitulated by engineering an optimized membrane-bound IL-12 (mbIL12) molecule in CAR T cells. These T cells show improved antigen-dependent T cell proliferation and recursive tumor cell killing in vitro, with robust in vivo efficacy in human ovarian cancer xenograft models. Locoregional administration of mbIL12-engineered CAR T cells promotes durable anti-tumor responses against both regional and systemic disease in mice. Safety and efficacy of mbIL12-engineered CAR T cells is demonstrated using an immunocompetent mouse model, with beneficial effects on the immunosuppressive tumor microenvironment. Collectively, our study features a clinically-applicable strategy to improve the efficacy of locoregionally-delivered CAR T cells engineered with antigen-dependent immune-modulating cytokines in targeting regional and systemic disease.


Assuntos
Neoplasias Ovarianas , Receptores de Antígenos Quiméricos , Feminino , Humanos , Camundongos , Animais , Imunoterapia Adotiva , Interleucina-12 , Receptores de Antígenos Quiméricos/genética , Linfócitos T , Neoplasias Ovarianas/terapia , Ensaios Antitumorais Modelo de Xenoenxerto , Linhagem Celular Tumoral , Microambiente Tumoral
20.
Nat Commun ; 14(1): 2041, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-37041154

RESUMO

Six transmembrane epithelial antigen of the prostate 1 (STEAP1) is a cell surface antigen for therapeutic targeting in prostate cancer. Here, we report broad expression of STEAP1 relative to prostate-specific membrane antigen (PSMA) in lethal metastatic prostate cancers and the development of a STEAP1-directed chimeric antigen receptor (CAR) T cell therapy. STEAP1 CAR T cells demonstrate reactivity in low antigen density, antitumor activity across metastatic prostate cancer models, and safety in a human STEAP1 knock-in mouse model. STEAP1 antigen escape is a recurrent mechanism of treatment resistance and is associated with diminished tumor antigen processing and presentation. The application of tumor-localized interleukin-12 (IL-12) therapy in the form of a collagen binding domain (CBD)-IL-12 fusion protein combined with STEAP1 CAR T cell therapy enhances antitumor efficacy by remodeling the immunologically cold tumor microenvironment of prostate cancer and combating STEAP1 antigen escape through the engagement of host immunity and epitope spreading.


Assuntos
Neoplasias da Próstata , Receptores de Antígenos Quiméricos , Masculino , Camundongos , Animais , Humanos , Linfócitos T , Interleucina-12 , Linhagem Celular Tumoral , Neoplasias da Próstata/patologia , Imunoterapia , Microambiente Tumoral , Antígenos de Neoplasias , Oxirredutases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA