Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Am Coll Surg ; 238(4): 436-447, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38214445

RESUMEN

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.


Asunto(s)
Virus Oncolíticos , Neoplasias Pancreáticas , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/genética , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Linfocitos T/metabolismo , Linfocitos T/trasplante , Antígenos CD19/metabolismo , Neoplasias Pancreáticas/terapia , Microambiente Tumoral
2.
Elife ; 132024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38251974

RESUMEN

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.


Asunto(s)
Células Endoteliales , ARN , Humanos , ARN Mitocondrial/genética , Cromatina , Bioensayo
3.
Mol Ther ; 32(2): 527-539, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38140726

RESUMEN

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.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Receptores de Interleucina-7 , Humanos , Animales , Ratones , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Inmunoterapia Adoptiva/métodos , Neoplasias/patología , Linfocitos T , Dexametasona/farmacología
4.
Nat Commun ; 14(1): 4737, 2023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37550294

RESUMEN

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.


Asunto(s)
Neoplasias Ováricas , Receptores Quiméricos de Antígenos , Femenino , Humanos , Ratones , Animales , Inmunoterapia Adoptiva , Interleucina-12 , Receptores Quiméricos de Antígenos/genética , Linfocitos T , Neoplasias Ováricas/terapia , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Microambiente Tumoral
5.
Nat Commun ; 14(1): 2041, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37041154

RESUMEN

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.


Asunto(s)
Neoplasias de la Próstata , Receptores Quiméricos de Antígenos , Masculino , Ratones , Animales , Humanos , Linfocitos T , Interleucina-12 , Línea Celular Tumoral , Neoplasias de la Próstata/patología , Inmunoterapia , Microambiente Tumoral , Antígenos de Neoplasias , Oxidorreductasas
6.
bioRxiv ; 2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36711615

RESUMEN

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.

8.
J Immunother Cancer ; 10(6)2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35738799

RESUMEN

BACKGROUND: The immune suppressive tumor microenvironment (TME) that inhibits T cell infiltration, survival, and antitumor activity has posed a major challenge for developing effective immunotherapies for solid tumors. Chimeric antigen receptor (CAR)-engineered T cell therapy has shown unprecedented clinical response in treating patients with hematological malignancies, and intense investigation is underway to achieve similar responses with solid tumors. Immunologically cold tumors, including prostate cancers, are often infiltrated with abundant tumor-associated macrophages (TAMs), and infiltration of CD163+ M2 macrophages correlates with tumor progression and poor responses to immunotherapy. However, the impact of TAMs on CAR T cell activity alone and in combination with TME immunomodulators is unclear. METHODS: To model this in vitro, we utilized a novel co-culture system with tumor cells, CAR T cells, and polarized M1 or M2 macrophages from CD14+ peripheral blood mononuclear cells collected from healthy human donors. Tumor cell killing, T cell activation and proliferation, and macrophage phenotypes were evaluated by flow cytometry, cytokine production, RNA sequencing, and functional blockade of signaling pathways using antibodies and small molecule inhibitors. We also evaluated the TME in humanized mice following CAR T cell therapy for validation of our in vitro findings. RESULTS: We observed inhibition of CAR T cell activity with the presence of M2 macrophages, but not M1 macrophages, coinciding with a robust induction of programmed death ligand-1 (PD-L1) in M2 macrophages. We observed similar PD-L1 expression in TAMs following CAR T cell therapy in the TME of humanized mice. PD-L1, but not programmed cell death protein-1, blockade in combination with CAR T cell therapy altered phenotypes to more M1-like subsets and led to loss of CD163+ M2 macrophages via interferon-γ signaling, resulting in improved antitumor activity of CAR T cells. CONCLUSION: This study reveals an alternative mechanism by which the combination of CAR T cells and immune checkpoint blockade modulates the immune landscape of solid tumors to enhance therapeutic efficacy of CAR T cells.


Asunto(s)
Antígeno B7-H1 , Inmunoterapia , Macrófagos , Neoplasias , Linfocitos T , Animales , Antígenos CD , Antígenos de Diferenciación Mielomonocítica , Humanos , Interferón gamma/metabolismo , Leucocitos Mononucleares , Macrófagos/inmunología , Ratones , Neoplasias/terapia , Receptores de Superficie Celular , Linfocitos T/inmunología , Microambiente Tumoral
9.
JCO Precis Oncol ; 6: e2100239, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35357903

RESUMEN

PURPOSE: Hyperthermic intraperitoneal chemotherapy (HIPEC) confers a survival benefit in epithelial ovarian cancer (EOC) and in preclinical models. However, the molecular changes induced by HIPEC have not been corroborated in humans. PATIENTS AND METHODS: A feasibility trial evaluated clinical and safety outcomes of HIPEC with cisplatin during optimal cytoreductive surgery (CRS) in patients with EOC diagnosed with stage III, IV, or recurrent EOC. Pre- and post-HIPEC biopsies were comprehensively profiled with genomic and transcriptomic sequencing to identify mutational and RNAseq signatures correlating with response; the tumor microenvironment was profiled to identify potential immune biomarkers; and transcriptional signatures of tumors and normal samples before and after HIPEC were compared to investigate HIPEC-induced acute transcriptional changes. RESULTS: Thirty-five patients had HIPEC at the time of optimal CRS; all patients had optimal CRS. The median progression-free survival (PFS) was 24.7 months for primary patients and 22.4 for recurrent patients. There were no grade 4 or 5 adverse events. Anemia was the most common grade 3 adverse event (43%). Hierarchical cluster analyses identified distinct transcriptomic signatures of good versus poor responders to HIPEC correlating with a PFS of 29.9 versus 7.3 months, respectively. Among good responders, significant HIPEC-induced molecular changes included immune pathway upregulation and DNA repair pathway downregulation. Within cancer islands, % programmed cell death protein 1 expression in CD8+ T cells significantly increased after HIPEC. An exceptional responder (PFS 58 months) demonstrated the highest programmed cell death protein 1 increase. Heat shock proteins comprised the top differentially upregulated genes in HIPEC-treated tumors. CONCLUSION: Distinct transcriptomic signatures identify responders to HIPEC, and preclinical model findings are confirmed for the first time in a human cohort.


Asunto(s)
Carcinoma Epitelial de Ovario , Quimioterapia Intraperitoneal Hipertérmica , Neoplasias Ováricas , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Estudios de Factibilidad , Femenino , Humanos , Quimioterapia Intraperitoneal Hipertérmica/efectos adversos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Microambiente Tumoral
10.
Gastroenterology ; 162(4): 1319-1333, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34999097

RESUMEN

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.


Asunto(s)
Neoplasias Pancreáticas , Receptores Quiméricos de Antígenos , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Inmunoterapia Adoptiva , Células Asesinas Naturales , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Neoplasias Pancreáticas/patología , Próstata , Células Madre/metabolismo , Neoplasias Pancreáticas
11.
Clin Cancer Res ; 28(4): 576-584, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34675084

RESUMEN

Immunotherapy has failed to achieve durable remissions in advanced prostate cancer patients. More potent T-cell-redirecting strategies may be needed to overcome the immunologically exclusive and suppressive tumor microenvironment. Clinical trials are underway, seeking to define the optimal target for T-cell redirection, such as PSMA, PSCA, or STEAP-1, as well as the optimal strategy, with CAR or bispecific antibodies. As results continue to emerge from these trials, understanding differential toxicity and efficacy of these therapies based on their targets and functional modifications will be key to advancing these promising therapies toward clinical practice. This review provides a unique depth and breadth of perspective regarding the diverse immunotherapy strategies currently under clinical investigation for men with advanced prostate cancer.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias de la Próstata , Anticuerpos Biespecíficos/uso terapéutico , Humanos , Inmunoterapia/métodos , Masculino , Neoplasias de la Próstata/patología , Linfocitos T , Microambiente Tumoral
12.
Nat Biomed Eng ; 5(11): 1348-1359, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34385695

RESUMEN

Treating solid malignancies with chimeric antigen receptor (CAR) T cells typically results in poor responses. Immunomodulatory biologics delivered systemically can augment the cells' activity, but off-target toxicity narrows the therapeutic window. Here we show that the activity of intratumoural CAR T cells can be controlled photothermally via synthetic gene switches that trigger the expression of transgenes in response to mild temperature elevations (to 40-42 °C). In vitro, heating engineered primary human T cells for 15-30 min led to over 60-fold-higher expression of a reporter transgene without affecting the cells' proliferation, migration and cytotoxicity. In mice, CAR T cells photothermally heated via gold nanorods produced a transgene only within the tumours. In mouse models of adoptive transfer, the systemic delivery of CAR T cells followed by intratumoural production, under photothermal control, of an interleukin-15 superagonist or a bispecific T cell engager bearing an NKG2D receptor redirecting T cells against NKG2D ligands enhanced antitumour activity and mitigated antigen escape. Localized photothermal control of the activity of engineered T cells may enhance their safety and efficacy.


Asunto(s)
Receptores Quiméricos de Antígenos , Animales , Deriva y Cambio Antigénico , Línea Celular Tumoral , Factores Inmunológicos , Inmunoterapia Adoptiva , Ratones , Receptores Quiméricos de Antígenos/genética , Linfocitos T
13.
Mol Ther ; 29(7): 2335-2349, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-33647456

RESUMEN

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.


Asunto(s)
Ciclofosfamida/farmacología , Inmunoterapia Adoptiva/métodos , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Pancreáticas/terapia , Neoplasias de la Próstata/terapia , Microambiente Tumoral , Animales , Antígenos de Neoplasias/genética , Apoptosis , Proliferación Celular , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Agonistas Mieloablativos/farmacología , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancer Immunol Res ; 9(1): 75-88, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33093217

RESUMEN

Lymphomas with central nervous system (CNS) involvement confer a worse prognosis than those without CNS involvement, and patients currently have limited treatment options. T cells genetically engineered with CD19-targeted chimeric antigen receptors (CAR) are effective against B-cell malignancies and show tremendous potential in the treatment of systemic lymphoma. We aimed to leverage this strategy toward a more effective therapy for patients with lymphoma with CNS disease. NOD-scid IL2Rgammanull (NSG) mice with CNS and/or systemic lymphoma were treated with CD19-CAR T cells via intracerebroventricular (ICV) or intravenous (IV) injection. CAR T cells isolated after treatment were rigorously examined for phenotype, gene expression, and function. We observed that CAR T cells infused ICV, but not IV, completely and durably eradicated both CNS and systemic lymphoma. CAR T cells delivered ICV migrated efficiently to the periphery, homed to systemic tumors, and expanded in vivo, leading to complete elimination of disease and resistance to tumor rechallenge. Mechanistic studies indicated that ICV-delivered CAR T cells are conditioned by exposure to cerebrospinal fluid in the ICV environment for superior antilymphoma activity and memory function compared with IV-delivered CAR T cells. Further analysis suggested that manipulating cellular metabolism or preactivating therapeutic CAR T cells with antigen ex vivo may improve the efficacy of CAR T cells in vivo Our demonstration that ICV-delivered CD19-CAR T cells had activity against CNS and systemic lymphoma could offer a valuable new strategy for treatment of B-cell malignancies with CNS involvement.


Asunto(s)
Neoplasias del Sistema Nervioso Central/terapia , Inmunoterapia Adoptiva/métodos , Linfoma/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/metabolismo , Animales , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Humanos , Inyecciones Intravenosas , Inyecciones Intraventriculares , Linfoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores Quiméricos de Antígenos/genética , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Cancer Ther ; 20(1): 173-182, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33262221

RESUMEN

Although it is known that oncolytic viruses can inflame and recruit immune cells to otherwise immunosuppressed tumor microenvironments, the influence of the antiviral immune response on antitumor immunity is less clear across viral platforms and tumor types. CF33 is a recombinant orthopoxvirus backbone effective against colon cancer. We tested derivatives of CF33 with and without immune-checkpoint inhibition (anti-PD-L1) in mouse models of colon cancer. Results showed that the efficacy of CF33 backbone with J2R deletion (single-deleted) against colon cancer is not altered by additional deletion of F14.5L in vitro or in vivo CF33 infection upregulated PD-L1 expression on tumor cells and led to an increased influx of lymphocytes and macrophages in tumors. Also, the levels of active CD8+ (IFNγ+) T cells in the virus-treated tumors were higher than those in control-treated tumors. Furthermore, a combination of CF33 derivatives with anti-PD-L1 resulted in durable tumor regression and long-term survival, resistant to tumor rechallenge. Analysis of immune cells from the treated mice showed that tumor-specific T cell activation occurred more robustly in tumors treated with the virus and that T cells were more strongly activated against the virus than against tumor, in an MHC-I-dependent manner. Our findings warrant further studies on the role of cross-priming of T cells against viral and tumor antigens, in the overall success of viroimmunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/inmunología , Neoplasias del Colon/virología , Reactividad Cruzada/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunidad , Orthopoxvirus/inmunología , Linfocitos T/inmunología , Animales , Línea Celular , Neoplasias del Colon/tratamiento farmacológico , Reactividad Cruzada/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Memoria Inmunológica/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Recombinación Genética/genética , Linfocitos T/efectos de los fármacos , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
16.
Sci Transl Med ; 12(559)2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32878978

RESUMEN

Chimeric antigen receptor (CAR)-engineered T cell therapy for solid tumors is limited by the lack of both tumor-restricted and homogeneously expressed tumor antigens. Therefore, we engineered an oncolytic virus to express a nonsignaling, truncated CD19 (CD19t) protein for tumor-selective delivery, enabling targeting by CD19-CAR T cells. Infecting tumor cells with an oncolytic vaccinia virus coding for CD19t (OV19t) produced de novo CD19 at the cell surface before virus-mediated tumor lysis. Cocultured CD19-CAR T cells secreted cytokines and exhibited potent cytolytic activity against infected tumors. Using several mouse tumor models, delivery of OV19t promoted tumor control after CD19-CAR T cell administration. OV19t induced local immunity characterized by tumor infiltration of endogenous and adoptively transferred T cells. CAR T cell-mediated tumor killing also induced release of virus from dying tumor cells, which propagated tumor expression of CD19t. Our study features a combination immunotherapy approach using oncolytic viruses to promote de novo CAR T cell targeting of solid tumors.


Asunto(s)
Neoplasias , Virus Oncolíticos , Animales , Antígenos CD19 , Inmunoterapia , Inmunoterapia Adoptiva , Ratones , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T
17.
Cancer Treat Res ; 180: 297-326, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32215875

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Inmunoterapia Adoptiva , Neoplasias/terapia , Humanos
18.
Cancer Treat Res ; 178: 189-205, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31209846

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Inmunoterapia , Neoplasias , Medicina de Precisión , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T
19.
Front Immunol ; 9: 2268, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30510550

RESUMEN

Impressive clinical efficacy of chimeric antigen receptor (CAR)-engineered T cell therapy for hematological malignancies have prompted significant efforts in achieving similar responses in solid tumors. The lack of truly restricted and uniform expression of tumor-associated antigens, as well as limited T cell persistence and/or tumor trafficking pose major challenges for successful translation of CAR T cell therapy in solid tumors. Recent studies have demonstrated that aberrantly glycosylated cell surface proteins on tumor cells are amenable CAR targets. Tumor-associated glycoprotein 72 (TAG72) antigen is the sialyl-Tn found on multiple O-glycoproteins expressed at high levels on the surface of several cancer types, including ovarian cancer. Here, we developed a humanized TAG72-specific CAR containing a 4-1BB intracellular co-stimulatory signaling domain (TAG72-BBζ). TAG72-BBζ CAR T cells showed potent antigen-dependent cytotoxicity and cytokine production against multiple TAG72+ ovarian cancer cell lines and patient-derived ovarian cancer ascites. Using in vivo xenograft models of peritoneal ovarian tumors, regional intraperitoneal delivery of TAG72-BBζ CAR T cells significantly reduced tumor growth, extended overall survival of mice, and was further improved with repeat infusions of CAR T cells. However, reduced TAG72 expression was observed in early recurring tumors, which coincided with a lack of T cell persistence. Taken together, we demonstrate efficacy with TAG72-CAR T cells in ovarian cancer, warranting further investigations as a CAR T cell therapeutic strategy for this disease.


Asunto(s)
Glicoproteínas/antagonistas & inhibidores , Inmunoterapia Adoptiva/métodos , Neoplasias Ováricas/terapia , Neoplasias Peritoneales/terapia , Receptores Quiméricos de Antígenos/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Femenino , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Humanos , Estimación de Kaplan-Meier , Ratones , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Peritoneales/inmunología , Neoplasias Peritoneales/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Carga Tumoral/inmunología
20.
J Nucl Med ; 59(10): 1531-1537, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29728514

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy is a promising clinical approach for reducing tumor progression and prolonging patient survival. However, improvements in both the safety and the potency of CAR T cell therapy demand quantitative imaging techniques to determine the distribution of cells after adoptive transfer. The purpose of this study was to optimize 89Zr-oxine labeling of CAR T cells and evaluate PET as a platform for imaging adoptively transferred CAR T cells. Methods: CAR T cells were labeled with 0-1.4 MBq of 89Zr-oxine per 106 cells and assessed for radioactivity retention, viability, and functionality. In vivo trafficking of 89Zr-oxine-labeled CAR T cells was evaluated in 2 murine xenograft tumor models: glioblastoma brain tumors with intracranially delivered IL13Rα2-targeted CAR T cells, and subcutaneous prostate tumors with intravenously delivered prostate stem cell antigen (PSCA)-targeted CAR T cells. Results: CAR T cells were efficiently labeled (75%) and retained more than 60% of the 89Zr over 6 d. In vitro cytokine production, migration, and tumor cytotoxicity, as well as in vivo antitumor activity, were not significantly reduced when labeled with 70 kBq/106 cells. IL13Rα2-CAR T cells delivered intraventricularly were detectable by PET for at least 6 d throughout the central nervous system and within intracranial tumors. When intravenously administered, PSCA-CAR T cells also showed tumor tropism, with a 9-fold greater tumor-to-muscle ratio than for CAR-negative T cells. Conclusion:89Zr-oxine can be used for labeling and imaging CAR T cells while maintaining cell viability and function. On the basis of these studies, we conclude that 89Zr-oxine is a clinically translatable platform for real-time assessment of cell therapies.


Asunto(s)
Inmunoterapia Adoptiva , Oxiquinolina/metabolismo , Radioisótopos , Linfocitos T/inmunología , Circonio , Animales , Línea Celular Tumoral , Humanos , Marcaje Isotópico , Masculino , Ratones , Oxiquinolina/farmacocinética , Linfocitos T/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...