Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Mol Ther ; 28(2): 548-560, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31870622

RESUMEN

The prognosis of patients diagnosed with advanced ovarian or endometrial cancer remains poor, and effective therapeutic strategies are limited. The Müllerian inhibiting substance type 2 receptor (MISIIR) is a transforming growth factor ß (TGF-ß) receptor family member, overexpressed by most ovarian and endometrial cancers while absent in most normal tissues. Restricted tissue expression, coupled with an understanding that MISIIR ligation transmits apoptotic signals to cancer cells, makes MISIIR an attractive target for tumor-directed therapeutics. However, the development of clinical MISIIR-targeted agents has been challenging. Prompted by the responses achieved in patients with blood malignancies using chimeric antigen receptor (CAR) T cell therapy, we hypothesized that MISIIR targeting may be achieved using a CAR T cell approach. Herein, we describe the development and evaluation of a CAR that targets MISIIR. T cells expressing the MISIIR-specific CAR demonstrated antigen-specific reactivity in vitro and eliminated MISIIR-overexpressing tumors in vivo. MISIIR CAR T cells also recognized a panel of human ovarian and endometrial cancer cell lines, and they lysed a battery of patient-derived tumor specimens in vitro, without mediating cytotoxicity of a panel of normal primary human cells. In conclusion, these results indicate that MISIIR targeting for the treatment of ovarian cancer and other gynecologic malignancies is achievable using CAR technology.


Asunto(s)
Neoplasias de los Genitales Femeninos/inmunología , Inmunoterapia Adoptiva , Neoplasias Ováricas/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores de Péptidos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epítopos/genética , Epítopos/inmunología , Femenino , Neoplasias de los Genitales Femeninos/terapia , Humanos , Ratones , Neoplasias Ováricas/terapia , Receptores Quiméricos de Antígenos/metabolismo , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Int J Mol Sci ; 22(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070369

RESUMEN

Folate receptor beta (FRß) is a folate binding receptor expressed on myeloid lineage hematopoietic cells. FRß is commonly expressed at high levels on malignant blasts in patients with acute myeloid leukemia (AML), as well as on M2 polarized tumor-associated macrophages (TAMs) in the tumor microenvironment of many solid tumors. Therefore, FRß is a potential target for both direct and indirect cancer therapy. We demonstrate that FRß is expressed in both AML cell lines and patient-derived AML samples and that a high-affinity monoclonal antibody against FRß (m909) has the ability to cause dose- and expression-dependent ADCC against these cells in vitro. Importantly, we find that administration of m909 has a significant impact on tumor growth in a humanized mouse model of AML. Surprisingly, m909 functions in vivo with and without the infusion of human NK cells as mediators of ADCC, suggesting potential involvement of mouse macrophages as effector cells. We also found that TAMs from primary ovarian ascites samples expressed appreciable levels of FRß and that m909 has the ability to cause ADCC in these samples. These results indicate that the targeting of FRß using m909 has the potential to limit the outgrowth of AML in vitro and in vivo. Additionally, m909 causes cytotoxicity to TAMs in the tumor microenvironment of ovarian cancer warranting further investigation of m909 and its derivatives as therapeutic agents in patients with FRß-expressing cancers.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Receptor 2 de Folato , Inmunoterapia , Leucemia Mieloide Aguda , Proteínas de Neoplasias , Neoplasias Ováricas , Animales , Células CHO , Cricetulus , Femenino , Receptor 2 de Folato/antagonistas & inhibidores , Receptor 2 de Folato/inmunología , Células HL-60 , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Células THP-1 , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Am Chem Soc ; 142(14): 6554-6568, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32191035

RESUMEN

Universal immune receptors represent a rapidly emerging form of adoptive T-cell therapy with the potential to overcome safety and antigen escape challenges faced by conventional chimeric antigen receptor (CAR) T-cell therapy. By decoupling antigen recognition and T-cell signaling domains via bifunctional antigen-specific targeting ligands, universal immune receptors can regulate T-cell effector function and target multiple antigens with a single receptor. Here, we describe the development of the SpyCatcher immune receptor, the first universal immune receptor that allows for the post-translational covalent attachment of targeting ligands at the T-cell surface through the application of SpyCatcher-SpyTag chemistry. The SpyCatcher immune receptor redirected primary human T cells against a variety of tumor antigens via the addition of SpyTag-labeled targeting ligands, both in vitro and in vivo. SpyCatcher T-cell activity relied upon the presence of both target antigen and SpyTag-labeled targeting ligand, allowing for dose-dependent control of function. The mutational disruption of covalent bond formation between the receptor and the targeting ligand still permitted redirected T-cell function but significantly compromised antitumor function. Thus, the SpyCatcher immune receptor allows for rapid antigen-specific receptor assembly, multiantigen targeting, and controllable T-cell activity.


Asunto(s)
Ingeniería Genética/métodos , Linfocitos T/inmunología , Humanos , Ligandos
4.
Blood ; 125(22): 3466-76, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25887778

RESUMEN

T cells expressing a chimeric antigen receptor (CAR) can produce dramatic results in lymphocytic leukemia patients; however, therapeutic strategies for myeloid leukemia remain limited. Folate receptor ß (FRß) is a myeloid-lineage antigen expressed on 70% of acute myeloid leukemia (AML) patient samples. Here, we describe the development and evaluation of the first CARs specific for human FRß (m909) in vitro and in vivo. m909 CAR T cells exhibited selective activation and lytic function against engineered C30-FRß as well as endogenous FRß(+) AML cell lines in vitro. In mouse models of human AML, m909 CAR T cells mediated the regression of engrafted FRß(+) THP1 AML in vivo. In addition, we demonstrated that treatment of AML with all-trans retinoic acid (ATRA) enhanced FRß expression, resulting in improved immune recognition by m909 CAR T cells. Because many cell surface markers are shared between AML blasts and healthy hematopoietic stem and progenitor cells (HSCs), we evaluated FRß expression and recognition of HSCs by CAR T cells. m909 CAR T cells were not toxic against healthy human CD34(+) HSCs in vitro. Our results indicate that FRß is a promising target for CAR T-cell therapy of AML, which may be augmented by combination with ATRA.


Asunto(s)
Receptor 2 de Folato/antagonistas & inhibidores , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Animales , Células Cultivadas , Femenino , Receptor 2 de Folato/genética , Terapia Genética/métodos , Células HEK293 , Humanos , Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Terapia Molecular Dirigida , Proteínas Mutantes Quiméricas/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
5.
Mol Ther ; 24(11): 1987-1999, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27439899

RESUMEN

B7-H4 protein is frequently overexpressed in ovarian cancer. Here, we engineered T cells with novel B7-H4-specific chimeric antigen receptors (CARs) that recognized both human and murine B7-H4 to test the hypothesis that B7-H4 CAR T cell therapy can be applied safely in preclinical models. B7-H4 CAR T cells specifically secreted IFN-γ and lysed B7-H4(+) targets. In vivo, B7-H4 CAR T cells displayed antitumor reactivity against B7-H4(+) human ovarian tumor xenografts. Unexpectedly, B7-H4 CAR T cell treatment reproducibly showed delayed, lethal toxicity 6-8 weeks after therapy. Comprehensive assessment of murine B7-H4 protein distribution uncovered expression in ductal and mucosal epithelial cells in normal tissues. Postmortem analysis revealed the presence of widespread histologic lesions that correlated with B7-H4(+) expression, and were inconsistent with graft versus host disease. Lastly, expression patterns of B7-H4 protein in normal human tissue were comparable to distribution in mice, advancing our understanding of B7-H4. We conclude that B7-H4 CAR therapy mediates control of cancer outgrowth. However, long-term engraftment of B7-H4 CAR T cells mediates lethal, off-tumor toxicity that is likely due to wide expression of B7-H4 in healthy mouse organs. This model system provides a unique opportunity for preclinical evaluation of safety approaches that limit CAR-mediated toxicity after tumor destruction in vivo.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Neoplasias Ováricas/terapia , Receptores de Antígenos/metabolismo , Linfocitos T/inmunología , Inhibidor 1 de la Activación de Células T con Dominio V-Set/inmunología , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias Ováricas/inmunología , Linfocitos T/trasplante , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Blood ; 119(3): 696-706, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22117050

RESUMEN

The costimulatory effects of CD27 on T lymphocyte effector function and memory formation has been confined to evaluations in mouse models, in vitro human cell culture systems, and clinical observations. Here, we tested whether CD27 costimulation actively enhances human T-cell function, expansion, and survival in vitro and in vivo. Human T cells transduced to express an antigen-specific chimeric antigen receptor (CAR-T) containing an intracellular CD3 zeta (CD3ζ) chain signaling module with the CD27 costimulatory motif in tandem exerted increased antigen-stimulated effector functions in vitro, including cytokine secretion and cytotoxicity, compared with CAR-T with CD3ζ alone. After antigen stimulation in vitro, CD27-bearing CAR-T cells also proliferated, up-regulated Bcl-X(L) protein expression, resisted apoptosis, and underwent increased numerical expansion. The greatest impact of CD27 was noted in vivo, where transferred CAR-T cells with CD27 demonstrated heightened persistence after infusion, facilitating improved regression of human cancer in a xenogeneic allograft model. This tumor regression was similar to that achieved with CD28- or 4-1BB-costimulated CARs, and heightened persistence was similar to 4-1BB but greater than CD28. Thus, CD27 costimulation enhances expansion, effector function, and survival of human CAR-T cells in vitro and augments human T-cell persistence and antitumor activity in vivo.


Asunto(s)
Neoplasias de la Mama/terapia , Antígenos CD28/inmunología , Inmunoterapia Adoptiva , Mesotelioma/terapia , Neoplasias Ováricas/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Antígenos CD28/genética , Proliferación Celular , Citocinas/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Activación de Linfocitos , Mesotelioma/genética , Mesotelioma/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Receptores de Antígenos de Linfocitos T/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Ther ; 20(3): 633-43, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22127019

RESUMEN

Cancer regression by gene-modified T cells bearing a chimeric antigen receptor (CAR) exodomain of mouse origin can be limited by the induction of transgene immunogenicity resulting in poor persistence and function in vivo. The development of functionally-active CAR of human origin can address this issue. Here, we constructed and evaluated fully human anti-mesothelin CARs comprised of a human mesothelin-specific single-chain antibody variable fragment (P4 scFv) coupled to T cell signaling domains. Primary human T cells expressing P4 CAR specifically produced proinflammatory cytokines, degranulated and exerted potent cytolytic functions when cultured with mesothelin-expressing tumors in vitro. P4 CAR T cells also mediated bystander killing of mesothelin-negative cancer cells during coculture. CAR reactivity was not abrogated by soluble tumor-secreted or recombinant mesothelin protein even at supraphysiological levels. Importantly, adoptive transfer of P4 CAR-expressing T cells mediated the regression of large, established tumor in the presence of soluble mesothelin in a xenogenic model of human ovarian cancer. Thus, primary human T cells expressing fully human anti-mesothelin CAR efficiently kill mesothelin-expressing tumors in vitro and in vivo and have the potential to overcome the issue of transgene immunogenicity that may limit CAR T cell trials that utilize scFvs of mouse origin.


Asunto(s)
Proteínas Ligadas a GPI/inmunología , Neoplasias Ováricas/inmunología , Receptores de Antígenos de Linfocitos T/genética , Anticuerpos de Cadena Única/genética , Linfocitos T/inmunología , Animales , Efecto Espectador/inmunología , Línea Celular , Citotoxicidad Inmunológica , Epítopos/inmunología , Femenino , Proteínas Ligadas a GPI/metabolismo , Orden Génico , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Mesotelina , Ratones , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Anticuerpos de Cadena Única/inmunología , Linfocitos T/metabolismo , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Blood Adv ; 7(9): 1885-1898, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-36053778

RESUMEN

Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of lymphoid malignancies associated with poor prognosis due to ineffective treatment options and high rates of relapse. The success of chimeric antigen receptor T-cell (CART) therapy for certain hematologic malignancies makes it an attractive treatment option for PTCLs. However, shared expression of potential target antigens by both malignant and healthy T cells poses a challenge. Current prospective CART approaches cause a high degree of on-target, off-tumor activity, resulting in fratricide during CART expansion, depletion of healthy T cells in vivo, and immune compromise in the patient. To limit off-tumor targeting, we sought to develop a CART platform specific for a given T-cell receptor vß (TCRvß) family that would endow CAR-modified T cells with the ability to mediate lysis of the clonal malignant population while preserving the majority of healthy T cells. Here, CAR constructs specific for multiple TCRvß family members were designed and validated. Our results demonstrate that TCRvß-family-specific CARTs (TCRvß-CARTs) recognize and kill TCRvß-expressing target cells. This includes specific self-depletion of the targeted cell subpopulation in the CART product and lysis of cell lines engineered to express a target TCRvß family. Furthermore, TCRvß-CARTs eliminated the dominant malignant TCRvß clone in 2 patient samples. Finally, in immunodeficient mice, TCRvß-CARTs eradicated malignant cells in a TCRvß-dependent manner. Importantly, the nontargeted TCRvß families were spared in all cases. Thus, TCRvß-CART therapy provides a potential option for high-precision treatment of PTCL with limited healthy T-cell depletion.


Asunto(s)
Linfoma de Células T Periférico , Receptores Quiméricos de Antígenos , Ratones , Animales , Linfocitos T , Recurrencia Local de Neoplasia , Receptores de Antígenos de Linfocitos T/genética , Linfoma de Células T Periférico/terapia , Células Clonales
9.
Cancer Cell ; 40(12): 1470-1487.e7, 2022 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-36513049

RESUMEN

Despite the success of CAR-T cell cancer immunotherapy, challenges in efficacy and safety remain. Investigators have begun to enhance CAR-T cells with the expression of accessory molecules to address these challenges. Current systems rely on constitutive transgene expression or multiple viral vectors, resulting in unregulated response and product heterogeneity. Here, we develop a genetic platform that combines autonomous antigen-induced production of an accessory molecule with constitutive CAR expression in a single lentiviral vector called Uni-Vect. The broad therapeutic application of Uni-Vect is demonstrated in vivo by activation-dependent expression of (1) an immunostimulatory cytokine that improves efficacy, (2) an antibody that ameliorates cytokine-release syndrome, and (3) transcription factors that modulate T cell biology. Uni-Vect is also implemented as a platform to characterize immune receptors. Overall, we demonstrate that Uni-Vect provides a foundation for a more clinically actionable next-generation cellular immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T , Humanos , Inmunoterapia Adoptiva/métodos , Linfocitos T , Vectores Genéticos/genética , Citocinas/metabolismo
10.
J Immunother Cancer ; 9(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34253637

RESUMEN

BACKGROUND: Bispecific T cell engagers represent the majority of bispecific antibodies (BsAbs) entering the clinic to treat metastatic cancer. The ability to apply these agents safely and efficaciously in the clinic, particularly for solid tumors, has been challenging. Many preclinical studies have evaluated parameters related to the activity of T cell engaging BsAbs, but many questions remain. MAIN BODY: This study investigates the impact of affinity of T cell engaging BsAbs with regards to potency, efficacy, and induction of immunomodulatory receptors/ligands using HER-2/CD3 BsAbs as a model system. We show that an IgG BsAb can be as efficacious as a smaller BsAb format both in vitro and in vivo. We uncover a dichotomous relationship between tumor-associated antigen (TAA) affinity and CD3 affinity requirements for cells that express high versus low levels of TAA. HER-2 affinity directly correlated with the CD3 engager lysis potency of HER-2/CD3 BsAbs when HER-2 receptor numbers are high (~200 K/cell), while the CD3 affinity did not impact potency until its binding affinity was extremely low (<600 nM). When HER-2 receptor numbers were lower (~20 K/cell), both HER-2 and CD3 affinity impacted potency. The high affinity anti-HER-2/low CD3 affinity BsAb also demonstrated lower cytokine induction levels in vivo and a dosing paradigm atypical of extremely high potency T cell engaging BsAbs reaching peak efficacy at doses >3 mg/kg. This data confirms that low CD3 affinity provides an opportunity for improved safety and dosing for T cell engaging BsAbs. T cell redirection also led to upregulation of Programmed cell death 1 (PD-1) and 4-1BB, but not CTLA-4 on T cells, and to Programmed death-ligand 1 (PD-L1) upregulation on HER-2HI SKOV3 tumor cells, but not on HER-2LO OVCAR3 tumor cells. Using this information, we combined anti-PD-1 or anti-4-1BB monoclonal antibodies with the HER-2/CD3 BsAb in vivo and demonstrated significantly increased efficacy against HER-2HI SKOV3 tumors via both combinations. CONCLUSIONS: Overall, these studies provide an informational dive into the optimization process of CD3 engaging BsAbs for solid tumors indicating that a reduced affinity for CD3 may enable a better therapeutic index with a greater selectivity for the target tumor and a reduced cytokine release syndrome. These studies also provide an additional argument for combining T cell checkpoint inhibition and co-stimulation to achieve optimal efficacy.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Linfocitos T/inmunología , Animales , Anticuerpos Biespecíficos/farmacología , Humanos , Ratones
11.
Nat Commun ; 12(1): 877, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33563975

RESUMEN

The immunosuppressive tumor microenvironment (TME) represents a major barrier for effective immunotherapy. Tumor-associated macrophages (TAMs) are highly heterogeneous and plastic cell components of the TME which can either promote tumor progression (M2-like) or boost antitumor immunity (M1-like). Here, we demonstrate that a subset of TAMs that express folate receptor ß (FRß) possess an immunosuppressive M2-like profile. In syngeneic tumor mouse models, chimeric antigen receptor (CAR)-T cell-mediated selective elimination of FRß+ TAMs in the TME results in an enrichment of pro-inflammatory monocytes, an influx of endogenous tumor-specific CD8+ T cells, delayed tumor progression, and prolonged survival. Preconditioning of the TME with FRß-specific CAR-T cells also improves the effectiveness of tumor-directed anti-mesothelin CAR-T cells, while simultaneous co-administration of both CAR products does not. These results highlight the pro-tumor role of FRß+ TAMs in the TME and the therapeutic implications of TAM-depleting agents as preparative adjuncts to conventional immunotherapies that directly target tumor antigens.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Receptor 2 de Folato/inmunología , Receptor 2 de Folato/metabolismo , Humanos , Terapia de Inmunosupresión , Mesotelina , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Neoplasias/inmunología , Células Tumorales Cultivadas , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/metabolismo
13.
J Clin Invest ; 109(8): 1111-20, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11956249

RESUMEN

Susceptibility to myasthenia gravis (MG) is positively linked to expression of HLA-DQ8 and DR3 molecules and negatively linked to expression of the DQ6 molecule. To elucidate the molecular basis of this association, we have induced experimental autoimmune MG (EAMG) in mice transgenic for HLA-DQ8, DQ6, and DR3, and in DQ8xDQ6 and DQ8xDR3 F(1) transgenic mice, by immunization with human acetylcholine receptor (H-AChR) in CFA. Mice expressing transgenes for one or both of the HLA class II molecules positively associated with MG (DQ8 and DR3) developed EAMG. T cells from DQ8 transgenic mice responded well to three cytoplasmic peptide sequences of H-AChR (alpha320-337, alpha304-322, and alpha419-437), of which the response to alpha320-337 was the most intense. DR3 transgenic mice also responded to this sequence very strongly. H-AChR- and alpha320-337 peptide-specific lymphocyte responses were restricted by HLA class II molecules. Disease resistance in DQ6 transgenic mice was associated with reduced synthesis of anti-AChR IgG, IgG(2b), and IgG(2c) Ab's and reduced IL-2 and IFN-gamma secretion by H-AChR- and peptide alpha320-337-specific lymphocytes. Finally, we show that DQ8 imparts susceptibility to EAMG and responsiveness to an epitope within the sequence alpha320-337 as a dominant trait.


Asunto(s)
Antígenos HLA/genética , Miastenia Gravis/genética , Miastenia Gravis/inmunología , Receptores Colinérgicos/genética , Receptores Colinérgicos/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Mapeo Epitopo , Epítopos/genética , Antígenos HLA-DQ/genética , Antígeno HLA-DR3/genética , Humanos , Inmunización , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular
14.
J Hematol Oncol ; 9(1): 56, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27439908

RESUMEN

BACKGROUND: The poor prognosis and the limited efficacy of targeted therapy in patients with triple-negative breast cancer (TNBC) have raised the need for alternative therapies. Recent studies have demonstrated that folate receptor-alpha (FRα) may represent an ideal tumor-associated marker for immunotherapy for TNBC. METHODS: The aim of the present study was to apply a chimeric antigen receptor (CAR) approach for the targeting of FRα expressed on TNBC cells and evaluate the antitumor activity of CAR-engineered T cells in vitro and in vivo. RESULTS: We found that human T cells expressing a FRα-specific CAR were potent and specific killers of TNBC cells that express moderate levels of FRα in vitro and significantly inhibited tumor outgrowth following infusion into immunodeficient mice bearing an MDA-MB-231 tumor xenograft. However, the antitumor activity of the FRα CAR T cells was modest when compared to the same CAR T cells applied in an ovarian tumor xenograft model where FRα expression is more abundant. Notably, FRα CAR T cells induced superior tumor regression in vivo against MDA-MB-231 that was engineered for overexpression of FRα. CONCLUSIONS: Taken together, our results show that FRα CAR T cells can mediate antitumor activity against established TNBC tumor, particularly when FRα is expressed at higher levels. These results have significant implications for the pre-selection of patients with high antigen expression levels when utilizing CAR-based adoptive T cell therapies of cancer in future clinical trials.


Asunto(s)
Receptor 1 de Folato/análisis , Inmunoterapia Adoptiva/métodos , Neoplasias Ováricas/terapia , Neoplasias de la Mama Triple Negativas/terapia , Animales , Células Cultivadas , Femenino , Receptor 1 de Folato/inmunología , Xenoinjertos , Humanos , Inmunoterapia Adoptiva/normas , Ratones , Neoplasias Ováricas/química , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Linfocitos T/trasplante , Neoplasias de la Mama Triple Negativas/química
15.
Oncotarget ; 7(50): 82354-82368, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27409425

RESUMEN

Exogenous cytokines are widely applied to enhance the anti-tumor ability of immune cells. However, systematic comparative studies of their effects on chimeric antigen receptor (CAR)-engineered T (CART) cells are lacking. In this study, CART cells targeting folate receptor-alpha were generated and expanded ex vivo in the presence of different cytokines (IL-2, IL-7, IL-15, IL-18, and IL-21), and their expansion, phenotype and cytotoxic capacity were evaluated, in vitro and in vivo. Moreover, the effect of the administration of these cytokines along with CART cells in vivo was also studied. IL-2, IL-7, and IL-15 favored the ex vivo expansion of CART cells compared to other cytokines or no cytokine treatment. IL-7 induced the highest proportion of memory stem cell-like CART cells in the final product, and IL-21 supported the expansion of CART cells with a younger phenotype, while IL-2 induced more differentiated CART cells. IL-2 and IL-15-exposed CART cells secreted more proinflammatory cytokines and presented stronger tumor-lysis ability in vitro. However, when tested in vivo, CART cells exposed to IL-2 ex vivo showed the least anti-tumor effect. In contrast, the administration of IL-15 and IL-21 in combination with CART cells in vivo increased their tumor killing capacity. According to our results, IL-7 and IL-15 show promise to promote ex vivo expansion of CART cells, while IL-15 and IL-21 seem better suited for in vivo administration after CART cell infusion. Collectively, these results may have a profound impact on the efficacy of CART cells in both hematologic and solid cancers.


Asunto(s)
Citocinas/farmacología , Citotoxicidad Inmunológica/efectos de los fármacos , Inmunoterapia Adoptiva , Activación de Linfocitos/efectos de los fármacos , Neoplasias Ováricas/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/trasplante , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Femenino , Humanos , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Immunol Res ; 3(10): 1130-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26112923

RESUMEN

Adoptive transfer of T cells engineered to express chimeric immunoreceptors is an effective strategy to treat hematologic cancers; however, the use of this type of therapy for solid cancers, such as ovarian cancer, remains challenging because a safe and effective immunotherapeutic target has not yet been identified. Here, we constructed and evaluated a novel redirected T-cell-based immunotherapy targeting human follicle-stimulating hormone receptor (FSHR), a highly conserved molecule in vertebrate animals with expression limited to gonadal tissues, ovarian cancer, and cancer-associated vasculature. Receptor ligand-based anti-FSHR immunoreceptors were constructed that contained small binding fragments from the ligand for FSHR, FSH, fused to T-cell transmembrane and T-cell signaling domains. Human T cells transduced to express anti-FSHR immunoreceptors were specifically immunoreactive against FSHR-expressing human and mouse ovarian cancer cell lines in an MHC-nonrestricted manner and mediated effective lysis of FHSR-expressing tumor cells, but not FSHR-deficient targets, in vitro. Similarly, the outgrowth of human ovarian cancer xenografts in immunodeficient mice was significantly inhibited by the adoptive transfer of FSHR-redirected T cells. Our experimental observations show that FSHR is a promising immunotherapeutic target for ovarian cancer and support further exploration of FSHR-targeted immune therapy approaches for patients with cancer.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias/genética , Neoplasias/inmunología , Receptores de HFE/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Línea Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Orden Génico , Vectores Genéticos/genética , Humanos , Inmunoterapia Adoptiva/métodos , Ratones , Neoplasias/terapia , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Receptores de HFE/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Oncotarget ; 6(25): 21533-46, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26101914

RESUMEN

Chimeric antigen receptors (CARs) can redirect T cells against antigen-expressing tumors in an HLA-independent manner. To date, various CARs have been constructed using mouse single chain antibody variable fragments (scFvs) of high affinity that are immunogenic in humans and have the potential to mediate "on-target" toxicity. Here, we developed and evaluated a fully human CAR comprised of the human C4 folate receptor-alpha (αFR)-specific scFv coupled to intracellular T cell signaling domains. Human T cells transduced to express the C4 CAR specifically secreted proinflammatory cytokine and exerted cytolytic functions when cultured with αFR-expressing tumors in vitro. Adoptive transfer of C4 CAR T cells mediated the regression of large, established human ovarian cancer in a xenogeneic mouse model. Relative to a murine MOv19 scFv-based αFR CAR, C4 CAR T cells mediated comparable cytotoxic tumor activity in vitro and in vivo but had lower affinity for αFR protein and exhibited reduced recognition of normal cells expressing low levels of αFR. Thus, T cells expressing a fully human CAR of intermediate affinity can efficiently kill antigen-expressing tumors in vitro and in vivo and may overcome issues of transgene immunogenicity and "on-target off-tumor" toxicity that plague trials utilizing CARs containing mouse-derived, high affinity scFvs.


Asunto(s)
Receptor 1 de Folato/química , Antígenos HLA/química , Trasplante de Neoplasias , Neoplasias Ováricas/terapia , Receptores de Antígenos/química , Secuencias de Aminoácidos , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Separación Celular , Citocinas/metabolismo , Femenino , Citometría de Flujo , Células HEK293 , Humanos , Inmunoterapia Adoptiva , Inflamación , Lentivirus/metabolismo , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias Ováricas/metabolismo , Estructura Terciaria de Proteína , Transducción de Señal , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/química , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Oncotarget ; 6(30): 28911-28, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26359629

RESUMEN

Using lentiviral technology, we recently demonstrated that incorporation of CD27 costimulation into CARs greatly improves antitumor activity and T cell persistence. Still, virus-mediated gene transfer is expensive, laborious and enables long-term persistence, creating therapies which cannot be easily discontinued if toxic. To address these concerns, we utilized a non-integrating RNA platform to engineer human T cells to express FRα-specific, CD27 CARs and tested their capacity to eliminate human FRα(+) cancer. Novel CARs comprised of human components were constructed, C4-27z and C4opt-27z, a codon-optimized variant created for efficient expression. Following RNA electroporation, C4-27z and C4opt-27z CAR expression is initially ubiquitous but progressively declines across T cell populations. In addition, C4-27z and C4opt-27z RNA CAR T cells secrete high levels of Th-1 cytokines and display strong cytolytic function against human FRα(+) cancers in a time- and antigen-dependent manner. Further, C4-27z and C4opt-27z CAR T cells exhibit significant proliferation in vivo, facilitate the complete regression of fully disseminated human ovarian cancer xenografts in mice and reduce the progression of solid ovarian cancer. These results advocate for rapid progression of C4opt-27z RNA CAR to the clinic and establish a new paradigm for preclinical optimization and validation of RNA CAR candidates destined for clinical translation.


Asunto(s)
Receptor 1 de Folato/metabolismo , Terapia Genética/métodos , Inmunoterapia Adoptiva/métodos , Linfocitos Infiltrantes de Tumor/trasplante , Neoplasias Glandulares y Epiteliales/terapia , Neoplasias Ováricas/terapia , ARN/genética , Linfocitos T/trasplante , Animales , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular , Terapia Combinada , Citocinas/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica , Electroporación , Femenino , Receptor 1 de Folato/inmunología , Regulación de la Expresión Génica , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fenotipo , ARN/metabolismo , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
FEBS Lett ; 537(1-3): 111-6, 2003 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-12606041

RESUMEN

Eosinophil-derived neurotoxin (EDN) is one of the four basic proteins stored in specific eosinophil granules. Here we demonstrate that EDN can also be detected at the surface of granulocytes. Reduction of EDN membrane expression after phosphatidylinositol-specific phospholipase C treatment suggests that a glycosylphosphatidylinositol (GPI) anchor is involved in the membrane association of EDN. The presence of a GPI anchor was confirmed by a lower expression of membrane EDN on granulocytes from patients with paroxysmal nocturnal hemoglobinuria which present cells lacking GPI anchor proteins. Furthermore, metabolic labeling with GPI anchor components supports biochemical evidence of GPI anchoring of EDN.


Asunto(s)
Glicosilfosfatidilinositoles/fisiología , Granulocitos/efectos de los fármacos , Ribonucleasas/toxicidad , Donantes de Sangre , Gránulos Citoplasmáticos/química , Neurotoxina Derivada del Eosinófilo , Eosinofilia/sangre , Eosinófilos/química , Citometría de Flujo , Granulocitos/citología , Granulocitos/patología , Hemoglobinuria Paroxística/sangre , Humanos , Recuento de Leucocitos , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Valores de Referencia , Ribonucleasas/sangre
20.
J Neuroimmunol ; 125(1-2): 51-8, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11960640

RESUMEN

To analyze the role of the Th2 cytokine interleukin-5 (IL-5) in experimental autoimmune myasthenia gravis (EAMG) pathogenesis, we induced clinical EAMG in C57BL/6 and IL-5 gene-knockout (KO) mice in the C57BL/6 background. IL-5 KO mice had a significantly reduced incidence and severity of EAMG. Despite their increased resistance to EAMG, IL-5 KO mice displayed intact secondary antibody and lymphoproliferative responses to acetylcholine receptor (AChR) after immunization with this molecule. However, the relative resistance of IL-5 KO mice was associated with a reduced primary lymphocyte response to AChR, and reduced C3 levels in muscle extracts compared to those in C57BL/6 mice.


Asunto(s)
Interleucina-5/genética , Interleucina-5/inmunología , Miastenia Gravis/genética , Miastenia Gravis/inmunología , Receptores Colinérgicos/inmunología , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Animales , Autoanticuerpos/sangre , Complemento C3/metabolismo , Modelos Animales de Enfermedad , Epítopos/inmunología , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-5/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Pronóstico , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA