Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 28(12): 2540-2552, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-32877695

RESUMEN

Enhancing the immunogenicity of tumor-associated antigens would represent a major advance for anti-tumor vaccination strategies. Here, we investigated structure-directed antigen destabilization as a strategy to improve the degradation, immunogenic epitope presentation, and T cell activation against a vesicular stomatitis virus (VSV)-encoded tumor antigen. We used the crystal structure of the model antigen ovalbumin to identify charge-disrupting amino acid mutations that were predicted to decrease the stability of the protein. One mutation, OVA-C12R, significantly reduced the half-life of the protein and was preferentially degraded in a 26-S proteasomal-dependent manner. The destabilized ovalbumin protein exhibited enhanced presentation of the major histocompatibility complex (MHC) class I immunogenic epitope, SIINFEKL, on the surface of B16F10 cells or murine bone marrow-derived dendritic cells (BMDCs) in vitro. Enhanced presentation correlated with better recognition by cognate CD8 OT-I T cells as measured by activation, proliferation, and effector cytokine production. Finally, VSV encoding the degradation-prone antigen was better able to prime an antigen ovalbumin-specific CD8 T cell response in vivo without altering the anti-viral CD8 T cell response. Our studies highlight that not only is the choice of antigen in cancer vaccines of importance, but that emphasis should be placed on modifying antigen quality to ensure optimal priming of anti-tumor responses.


Asunto(s)
Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos/inmunología , Inmunidad , Activación de Linfocitos , Ovalbúmina/genética , Vesiculovirus/genética , Animales , Presentación de Antígeno , Antígenos de Neoplasias/química , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Epítopos/inmunología , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Viroterapia Oncolítica/métodos , Ovalbúmina/química , Estabilidad Proteica
2.
Mol Ther ; 25(4): 962-975, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28237836

RESUMEN

Systemic viroimmunotherapy activates endogenous innate and adaptive immune responses against both viral and tumor antigens. We have shown that therapy with vesicular stomatitis virus (VSV) engineered to express a tumor-associated antigen activates antigen-specific adoptively transferred T cells (adoptive cell therapy, ACT) in vivo to generate effective therapy. The overall goal of this study was to phenotypically characterize the immune response to VSV+ACT therapy and use the information gained to rationally improve combination therapy. We observed rapid expansion of blood CD8+ effector cells acutely following VSV therapy with markedly high expression of the immune checkpoint molecules PD-1 and TIM-3. Using these data, we tested a treatment schedule incorporating mAb immune checkpoint inhibitors with VSV+ACT treatment. Unlike clinical scenarios, we delivered therapy at early time points following tumor establishment and treatment. Our goal was to potentiate the immune response generated by VSV therapy to achieve durable control of metastatic disease. Despite the high frequency of endogenous PD-1+ TIM-3+ CD8+ T cells following virus administration, antibody blockade did not improve survival. These findings provide highly significant information about response kinetics to viroimmunotherapy and juxtapose the clinical use of checkpoint inhibitors against chronically dysfunctional T cells and the acute T cell response to oncolytic viruses.


Asunto(s)
Traslado Adoptivo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vectores Genéticos/genética , Inmunoterapia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Virus de la Estomatitis Vesicular Indiana/genética , Animales , Modelos Animales de Enfermedad , Femenino , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Memoria Inmunológica , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Mortalidad , Metástasis de la Neoplasia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Resultado del Tratamiento
3.
Bioorg Med Chem ; 25(10): 2701-2712, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28372935

RESUMEN

As a continuation of our search for novel histamine H3 receptor ligands a series of twenty new tert-amyl phenoxyalkylamine derivatives (2-21) was synthesized. Compounds of four to eight carbon atoms spacer alkyl chain were evaluated on their binding properties at human histamine H3 receptor (hH3R). The highest affinities were observed for pentyl derivatives 6-8 (Ki=8.8-23.4nM range) and among them piperidine derivative 6 with Ki=8.8nM. Structures 6, 7 were also classified as antagonists in cAMP accumulation assay (with EC50=157 and 164nM, respectively). Moreover, new compounds were also evaluated for anticonvulsant activity in Antiepileptic Screening Program (ASP) at National Institute of Neurological Disorders and Stroke (USA). Seven compounds (2-4, 9, 11, 12 and 20) showed anticonvulsant activity at maximal electroshock (MES) test in the dose of 30mg/kg at 0.5h. In the subcutaneous pentetrazole (scMET) test compound 4 showed protection at 100 and 300mg/kg dose at mice, however compounds showed high neurotoxicity in rotarod test at used doses. Also, molecular modeling studies were undertaken, to explain affinity of compounds at hH3R (taking into the consideration X-ray analysis of compound 18). In order to estimate "drug-likeness" of selected compounds in silico and experimental evaluation of lipophilicity, metabolic stability and cytotoxicity was performed.


Asunto(s)
Anticonvulsivantes/síntesis química , Antagonistas de los Receptores Histamínicos H3/síntesis química , Piperidinas/química , Receptores Histamínicos H3/química , Animales , Anticonvulsivantes/metabolismo , Anticonvulsivantes/toxicidad , Sitios de Unión , Línea Celular , Proliferación Celular/efectos de los fármacos , Electrochoque , Células HEK293 , Antagonistas de los Receptores Histamínicos H3/metabolismo , Antagonistas de los Receptores Histamínicos H3/toxicidad , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Neuronas/efectos de los fármacos , Piperidinas/metabolismo , Piperidinas/toxicidad , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Receptores Histamínicos H3/metabolismo , Solubilidad
4.
Mol Ther ; 23(5): 845-856, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25544599

RESUMEN

We used a VSV-cDNA library to treat recurrent melanoma, identifying immunogenic antigens, allowing us to target recurrences with immunotherapy or chemotherapy. Primary B16 melanoma tumors were induced to regress by frontline therapy. Mice with recurrent tumors were treated with VSV-cDNA immunotherapy. A Th17 recall response was used to screen the VSV-cDNA library for individual viruses encoding rejection antigens, subsequently targeted using immunotherapy or chemotherapy. Recurrent tumors were effectively treated with a VSV-cDNA library using cDNA from recurrent B16 tumors. Recurrence-associated rejection antigens identified included Topoisomerase-IIα, YB-1, cdc7 kinase, and BRAF. Fourteen out of 16 recurrent tumors carried BRAF mutations (595-605 region) following frontline therapy, even though the parental B16 tumors were BRAF wild type. The emergence of mutated BRAF-containing recurrences served as an excellent target for BRAF-specific immune-(VSV-BRAF), or chemo-(PLX-4720) therapies. Successful PLX-4720 therapy of recurrent tumors was associated with the development of a broad spectrum of T-cell responses. VSV-cDNA technology can be used to identify recurrence specific antigens. Emergence of mutated BRAF may be a major effector of melanoma recurrence which could serve as a target for chemo or immune therapy. This study suggests a rationale for offering patients with initially wild-type BRAF melanomas an additional biopsy to screen for mutant BRAF upon recurrence.


Asunto(s)
Melanoma/genética , Melanoma/patología , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Antígenos de Neoplasias/inmunología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Secuencia de Bases , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Ganciclovir/farmacología , Expresión Génica , Humanos , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/farmacología , Masculino , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Melanoma Experimental , Ratones , Datos de Secuencia Molecular , Recurrencia Local de Neoplasia , Proteínas Proto-Oncogénicas B-raf/química , Timidina Quinasa/genética
5.
Sci Transl Med ; 14(640): eabn2231, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35417192

RESUMEN

Oncolytic viruses (OVs) encoding a variety of transgenes have been evaluated as therapeutic tools to increase the efficacy of chimeric antigen receptor (CAR)-modified T cells in the solid tumor microenvironment (TME). Here, using systemically delivered OVs and CAR T cells in immunocompetent mouse models, we have defined a mechanism by which OVs can potentiate CAR T cell efficacy against solid tumor models of melanoma and glioma. We show that stimulation of the native T cell receptor (TCR) with viral or virally encoded epitopes gives rise to enhanced proliferation, CAR-directed antitumor function, and distinct memory phenotypes. In vivo expansion of dual-specific (DS) CAR T cells was leveraged by in vitro preloading with oncolytic vesicular stomatitis virus (VSV) or reovirus, allowing for a further in vivo expansion and reactivation of T cells by homologous boosting. This treatment led to prolonged survival of mice with subcutaneous melanoma and intracranial glioma tumors. Human CD19 CAR T cells could also be expanded in vitro with TCR reactivity against viral or virally encoded antigens and was associated with greater CAR-directed cytokine production. Our data highlight the utility of combining OV and CAR T cell therapy and show that stimulation of the native TCR can be exploited to enhance CAR T cell activity and efficacy in mice.


Asunto(s)
Glioma , Melanoma , Viroterapia Oncolítica , Virus Oncolíticos , Receptores Quiméricos de Antígenos , Animales , Glioma/terapia , Inmunoterapia Adoptiva , Melanoma/terapia , Ratones , Virus Oncolíticos/fisiología , Receptores de Antígenos de Linfocitos T , Linfocitos T , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Bioorg Med Chem ; 19(9): 2850-8, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21498080

RESUMEN

Previous studies have shown that several imidazole derivatives possess affinity to histamine H(3) and H(4) receptors. Continuing our study on structural requirements responsible for affinity and selectivity for H(3)/H(4) receptor subtypes, two series of 3-(1H-imidazol-4-yl)propyl carbamates were prepared: a series of unsaturated alkyl derivatives (1-9) and a series possessing a cycloalkyl group different distances to the carbamate moiety (10-13). The compounds were tested for their affinities at the human histamine H(3) receptor, stably expressed in CHO-K1 cells. Compounds 1, 2, 5-7, 10-13 were investigated for their affinities at the human histamine H(4) receptor co-expressed with Gα(i2) and Gß(1)γ(2) subunits in Sf9 cells. To expand the pharmacological profile, compounds were further tested for their H(3) receptor antagonist activity on guinea pig ileum and in vivo after oral administration to mice. All tested compounds exhibited good affinity for the human histamine H(3) receptor with K(i) values in the range from 14 to 194nM. All compounds were active in vivo after peroral administration (p.o.) to Swiss mice, thus demonstrating their ability to cross the blood-brain barrier. The most potent H(3) receptor ligand of these series was compound 5, 3-(1H-imidazol-4-yl)propyl pent-4-enylcarbamate with the highest affinity (K(i)=14nM). Additionally, compound 3 showed remarkable central nervous system (CNS) H(3)R activity, increasing the N(τ)-methylhistamine levels in mice with an ED(50) value of 0.55mg/kg, p.o. evidencing therefore, a twofold increase of inverse agonist/antagonist potency compared to the reference inverse agonist/antagonist thioperamide. In this study, the imidazole propyloxy carbamate moiety was kept constant. The different lipophilic moieties connected to the carbamate functionality in the eastern part of the molecule had a range of influences on the human H(4) receptor affinity (154-1326nM).


Asunto(s)
Carbamatos/química , Antagonistas de los Receptores Histamínicos/química , Ftalimidas/química , Receptores Histamínicos H3/química , Carbamatos/síntesis química , Carbamatos/farmacología , Antagonistas de los Receptores Histamínicos/síntesis química , Antagonistas de los Receptores Histamínicos/farmacología , Humanos , Ligandos , Ftalimidas/síntesis química , Ftalimidas/farmacología , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Receptores Histamínicos H3/metabolismo
7.
Nat Med ; 9(9): 1215-9, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12925849

RESUMEN

Fusion of tumor cells with antigen-presenting cells (APCs) has been proposed for the preparation of cancer vaccines. However, generation of these hybrids, using physical or chemical methods such as electrofusion or polyethylene glycol (PEG), has been difficult to standardize. Characterization of cell fusion has also been problematic because of difficulties in differentiating fusion from cell aggregation, leakage of cellular dyes and dendritic-cell (DC) phagocytosis of tumor material. In this report, we describe a new method to generate hybrid cell vaccines, based on gene transfer of a viral fusogenic membrane glycoprotein (FMG) into tumor cells, and incorporate a genetic method by which true hybrid formation can be unambiguously detected. We describe a new class of tumor cell-DC hybrid that can be rapidly isolated after cell fusion. These hybrids are highly potent in in vitro antigen presentation assays, target lymph nodes in vivo and are powerful immunogens against established metastatic disease.


Asunto(s)
Vacunas contra el Cáncer/genética , Células Dendríticas/citología , Técnicas Genéticas , Animales , Antígenos de Neoplasias , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacología , Fusión Celular/métodos , Células Dendríticas/fisiología , Glicoproteínas/genética , Células Híbridas , Ganglios Linfáticos/patología , Melanoma/genética , Melanoma/patología , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Células Tumorales Cultivadas , Vacunación
8.
Chem Pharm Bull (Tokyo) ; 58(10): 1353-61, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20930404

RESUMEN

The histamine H(3) receptor (H(3)R) is a promising target in the development of new compounds for the treatment of mainly centrally occurring diseases. However, emerging novel therapeutic concepts have been introduced and some indications in the H(3)R field, e.g. migraine, pain or allergic rhinitis, might take advantage of peripherally acting ligands. In this work, kojic acid-derived structural elements were inserted into a well established H(3)R antagonist/inverse agonist scaffold to investigate the bioisosteric potential of γ-pyranones with respect to the different moieties of the H(3)R pharmacophore. The most affine compounds showed receptor binding in the low nanomolar concentration range. Evaluation and comparison of kojic acid-containing ligands and their corresponding phenyl analogues (3-7) revealed that the newly integrated scaffold greatly influences chemical properties (S Log P, topological polar surface area (tPSA)) and hence, potentially modifies the pharmacokinetic profile of the different derivatives. Benzyl-1-(4-(3-(piperidin-1-yl)propoxy)phenyl)methanamine ligands 3 and 4 belong to the centrally acting diamine-based class of H(3)R antagonist/inverse agonist, whereas kojic acid analogues 6 and 7 might act peripherally. The latter compounds state promising lead structures in the development of H(3)R ligands with a modified profile of action.


Asunto(s)
Antagonistas de los Receptores Histamínicos/química , Pironas/química , Receptores Histamínicos H3/química , Línea Celular , Agonismo Inverso de Drogas , Antagonistas de los Receptores Histamínicos/síntesis química , Humanos , Ligandos , Pironas/síntesis química , Receptores Histamínicos H3/metabolismo
9.
Nat Commun ; 11(1): 3187, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32581235

RESUMEN

The application of adoptive T cell therapies, including those using chimeric antigen receptor (CAR)-modified T cells, to solid tumors requires combinatorial strategies to overcome immune suppression associated with the tumor microenvironment. Here we test whether the inflammatory nature of oncolytic viruses and their ability to remodel the tumor microenvironment may help to recruit and potentiate the functionality of CAR T cells. Contrary to our hypothesis, VSVmIFNß infection is associated with attrition of murine EGFRvIII CAR T cells in a B16EGFRvIII model, despite inducing a robust proinflammatory shift in the chemokine profile. Mechanistically, type I interferon (IFN) expressed following infection promotes apoptosis, activation, and inhibitory receptor expression, and interferon-insensitive CAR T cells enable combinatorial therapy with VSVmIFNß. Our study uncovers an unexpected mechanism of therapeutic interference, and prompts further investigation into the interaction between CAR T cells and oncolytic viruses to optimize combination therapy.


Asunto(s)
Inmunoterapia Adoptiva , Interferón beta/metabolismo , Virus Oncolíticos/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Quimiocinas/metabolismo , Terapia Combinada , Femenino , Interferón beta/genética , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Viroterapia Oncolítica , Virus Oncolíticos/genética , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Bazo/inmunología
10.
Bioorg Med Chem Lett ; 19(23): 6682-5, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19846299

RESUMEN

A series of imidazole-containing (non-)chiral carbamates were tested at human histamine H(3) receptor (H(3)R). All compounds displayed K(i) values below 100 nM. A trend for a stereoselectivity at human H(3)R was observed for the chiral alpha-branched ligands. Selected compounds were also tested at human histamine H(4) receptor and showed moderate to weak affinities (118-1460 nM).


Asunto(s)
Carbamatos/farmacología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Histamínicos H3/efectos de los fármacos , Receptores Histamínicos/efectos de los fármacos , Carbamatos/síntesis química , Carbamatos/química , Diseño de Fármacos , Humanos , Imidazoles/química , Estructura Molecular , Receptores Histamínicos H4 , Estereoisomerismo , Relación Estructura-Actividad
11.
Bioorg Med Chem ; 17(20): 7186-96, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19773175

RESUMEN

The human histamine H(4) receptor (hH(4)R) is a promising new target in the therapy of inflammatory diseases and disorders of the immune system. For the development of new H(4)R antagonists a broad ligand-based virtual screening was performed resulting in two hits. The dissection of their common annelated aromatic core into its heteromonocyclic components showed that 2,4-diaminopyrimidine is a potent hH(4)R affinity scaffold, which was comprehensively investigated. Structure-activity relationship studies revealed that slight structural changes evoke extensive differences in functional activities and potencies: while o- and p-substituted benzyl amines mainly showed partial agonism, m-substituted and rigidified ones exhibited inverse agonist efficacy.


Asunto(s)
Pirimidinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Histamínicos/metabolismo , Ligandos , Espectroscopía de Resonancia Magnética , Receptores Histamínicos H4 , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad
12.
Cancer Res ; 67(1): 300-8, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17210711

RESUMEN

Adoptive T-cell transfer has achieved significant clinical success in advanced melanoma. However, therapeutic efficacy is limited by poor T-cell survival after adoptive transfer and by inefficient trafficking to tumor sites. Here, we report that intratumoral expression of the chemokine CCL21 enhances the efficacy of adoptive T-cell therapy in a mouse model of melanoma. Based on our novel observation that CCL21 is highly chemotactic for activated OT-1 T cells in vitro and down-regulates expression of CD62L, we hypothesized that tumor cell-mediated expression of this chemokine might recruit, and retain, adoptively transferred T cells to the sites of tumor growth. Mice bearing metastatic tumors stably transduced with CCL21 survived significantly longer following adoptive T-cell transfer than mice bearing non-CCL21-expressing tumors. However, although we could not detect increased trafficking of the adoptively transferred T cells to tumors, tumor-expressed CCL21 promoted the survival and cytotoxic activity of the adoptively transferred T cells and led to the priming of antitumor immunity following T-cell transfer. To translate these observations into a protocol of real clinical usefulness, we showed that adsorption of a retrovirus encoding CCL21 to OT-1 T cells before adoptive transfer increased the therapeutic efficacy of a subsequently administered dose of OT-1 T cells, resulting in cure of metastatic disease and the generation of immunologic memory in the majority of treated mice. These studies indicate a promising role for CCL21 in enhancing the therapeutic efficacy of adoptive T-cell therapy.


Asunto(s)
Quimiocinas CC/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Quimiocina CCL21 , Quimiocinas CC/biosíntesis , Quimiocinas CC/genética , Quimiotaxis de Leucocito/inmunología , Regulación hacia Abajo , Femenino , Selectina L/biosíntesis , Selectina L/metabolismo , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Transfección
13.
Clin Cancer Res ; 12(4): 1333-41, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16489091

RESUMEN

PURPOSE: Fusogenic membrane glycoproteins (FMG), such as the vesicular stomatitis virus G glycoprotein (VSV-G), represent a new class of gene therapy for cancer that cause cytotoxic fusion on expression in tumor cells. In addition, FMG-mediated tumor cell death stimulates antitumor immunity, suggesting potential applications for FMG-expressing cellular vaccines. This study addresses the promise of FMG-expressing allogeneic tumor cells, which are most practical for clinical use, as a novel platform for ex vivo and in situ vaccination. EXPERIMENTAL DESIGN: Murine B16 melanoma-derived cell lines expressing autologous or allogeneic MHC class I, expressing fusogenic or nonfusogenic VSV-G, were used to vaccinate mice in vivo against a live tumor challenge. Exosome-like vesicles released by fusing allogeneic cells (syncitiosomes) and intratumoral injection of fusing vaccines were also tested as novel therapeutic strategies for their antitumor effects. RESULTS: Expression of fusogenic VSV-G enhanced the immunogenicity of an allogeneic cellular vaccine, which was more effective than a fusing autologous vaccine. Allogeneic syncitiosomes were only as effective as cellular vaccines when administered with adjuvant, demonstrating that syncitiosomes cannot account entirely for the mechanism of immune priming. Intratumoral injection of FMG-expressing allogeneic cells led to significant tumor regression using both fusogenic or nonfusogenic VSV-G. However, specific priming against tumor-associated antigenic epitopes and protection against secondary rechallenge only occurred if the initial vaccine was competent for cell fusion. CONCLUSIONS: FMG-expressing allogeneic tumor cells are a potent source of antitumor vaccines. Syncitiosomes given with adjuvant and intratumoral injection of fusing cells represent novel strategies well-suited to clinical translation.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Glicoproteínas de Membrana/genética , Proteínas del Envoltorio Viral/genética , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Vesículas Citoplasmáticas/inmunología , Expresión Génica , Antígenos H-2/genética , Antígenos H-2/inmunología , Melanoma Experimental/genética , Melanoma Experimental/patología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Inducción de Remisión , Factores de Tiempo , Transfección , Trasplante Homólogo , Resultado del Tratamiento , Proteínas del Envoltorio Viral/inmunología
14.
Front Chem ; 5: 64, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28955709

RESUMEN

Neurleptic drugs, e.g., aripiprazole, targeting the dopamine D2S and D3 receptors (D2SR and D3R) in the central nervous system are widely used in the treatment of several psychotic and neurodegenerative diseases. Therefore, a new series of benzothiazole-based ligands (3-20) was synthesized by applying the bioisosteric approach derived from the selective D3Rs ligand BP-897 (1) and its structurally related benz[d]imidazole derivative (2). Herein, introduction of the benzothiazole moiety was well tolerated by D2SR and D3R binding sites leading to antagonist affinities in the low nanomolar concentration range at both receptor subtypes. However, all novel compounds showed lower antagonist affinity to D3R when compared to that of 1. Further exploration of different substitution patterns at the benzothiazole heterocycle and the basic 4-phenylpiperazine resulted in the discovery of high dually acting D2SR and D3R ligands. Moreover, the methoxy substitution at 2-position of 4-phenylpiperazine resulted in significantly (22-fold) increased D2SR binding affinity as compared to the parent ligand 1, and improved physicochemical and drug-likeness properties of ligands 3-11. However, the latter structural modifications failed to improve the drug-able properties in ligands having un-substituted 4-phenylpiperazine analogs (12-20). Accordingly, compound 9 showed in addition to high dual affinity at the D2SR and D3R [Ki (hD2SR) = 2.8 ± 0.8 nM; Ki (hD3R) = 3.0 ± 1.6 nM], promising clogS, clogP, LE (hD2SR, hD3R), LipE (hD2SR, hD3R), and drug-likeness score values of -4.7, 4.2, (0.4, 0.4), (4.4, 4.3), and 0.7, respectively. Also, the deaminated analog 10 [Ki (hD2SR) = 3.2 ± 0.4 nM; Ki (hD3R) = 8.5 ± 2.2 nM] revealed clogS, clogP, LE (hD2SR, hD3R), LipE (hD2SR, hD3R) and drug-likeness score values of -4.7, 4.2, (0.4, 0.4), (3.9, 3.5), and 0.4, respectively. The results observed for the newly developed benzothiazole-based ligands 3-20 provide clues for the diversity in structure activity relationships (SARs) at the D2SR and D3R subtypes.

15.
Cancer Immunol Res ; 5(11): 1029-1045, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29038298

RESUMEN

Understanding how incompletely cleared primary tumors transition from minimal residual disease (MRD) into treatment-resistant, immune-invisible recurrences has major clinical significance. We show here that this transition is mediated through the subversion of two key elements of innate immunosurveillance. In the first, the role of TNFα changes from an antitumor effector against primary tumors into a growth promoter for MRD. Second, whereas primary tumors induced a natural killer (NK)-mediated cytokine response characterized by low IL6 and elevated IFNγ, PD-L1hi MRD cells promoted the secretion of IL6 but minimal IFNγ, inhibiting both NK-cell and T-cell surveillance. Tumor recurrence was promoted by trauma- or infection-like stimuli inducing VEGF and TNFα, which stimulated the growth of MRD tumors. Finally, therapies that blocked PD-1, TNFα, or NK cells delayed or prevented recurrence. These data show how innate immunosurveillance mechanisms, which control infection and growth of primary tumors, are exploited by recurrent, competent tumors and identify therapeutic targets in patients with MRD known to be at high risk of relapse. Cancer Immunol Res; 5(11); 1029-45. ©2017 AACR.


Asunto(s)
Células Asesinas Naturales/inmunología , Melanoma Experimental/inmunología , Recurrencia Local de Neoplasia/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Anticuerpos/uso terapéutico , Línea Celular Tumoral , Citocinas/inmunología , Femenino , Ganciclovir/uso terapéutico , Vigilancia Inmunológica , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Ratones Transgénicos , Viroterapia Oncolítica , Paclitaxel/uso terapéutico , Reoviridae , Neoplasias Cutáneas/terapia , Linfocitos T/trasplante
16.
Cancer Res ; 63(12): 3173-80, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12810645

RESUMEN

We aimed to use cell-based carriers to direct vector production to target sites for systemic therapy. We used T cells engineered to express a chimeric T cell receptor that can specifically recognize target cells expressing the tumor-associated carcinoembryonic antigen (CEA). These T cells were modified to produce a retrovirus under tight pharmacological control using the rapamycin-inducible transcriptional regulatory system. The retroviral vectors produced were transcriptionally targeted to CEA-expressing target cells. We found that vector production and transgene expression from these T cells in vitro was dependent on pharmacological induction and expression of CEA in target cells, respectively. Mice bearing metastatic tumors that received cell carriers delivering the HSVtk gene demonstrated a significant increase in survival, but only in response to pharmacological induction of vector production. Interestingly, the therapeutic effect required the presence of the tumor-specific chimeric receptor on T cells. Further studies demonstrated that systemic delivery of tumor-specific T cells to mice bearing metastatic tumors caused recruitment of nonspecific T cells to the tumor site. We hypothesize that this enhanced targeting to tumor sites is responsible for the efficiency of T cell-mediated retroviral gene transfer and that this principle can be used to enhance systemic therapies using immune-cell carriers.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígeno Carcinoembrionario/inmunología , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Células Jurkat/virología , Virus de la Leucemia Murina de Moloney/genética , Proteínas Recombinantes de Fusión/inmunología , Sirolimus/farmacología , Transcripción Genética/efectos de los fármacos , Adenocarcinoma/patología , Adenocarcinoma/secundario , Animales , Neoplasias Colorrectales/patología , Sistemas de Liberación de Medicamentos , Colorantes Fluorescentes , Regulación Viral de la Expresión Génica , Genes Sintéticos , Vectores Genéticos/uso terapéutico , Humanos , Células Jurkat/trasplante , Neoplasias Hepáticas Experimentales/secundario , Neoplasias Hepáticas Experimentales/terapia , Melanoma/patología , Melanoma/secundario , Ratones , Virus de la Leucemia Murina de Moloney/fisiología , Especificidad de Órganos , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Simplexvirus/enzimología , Simplexvirus/genética , Secuencias Repetidas Terminales , Timidina Quinasa/genética , Transducción Genética , Transfección , Proteínas Virales/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 62(22): 6566-78, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12438252

RESUMEN

Expression of viral fusogenic membrane glycoproteins (FMGs) is a potent strategy for antitumor cytotoxic gene therapy in which tumor cells are fused into large multinucleated syncytia. To understand how local cell killing can potentiate activation of antitumor immune responses, we characterized the mechanism of FMG-mediated cell killing. Here, we show that syncytia are highly ordered structures over 24-48 h but then die through processes that, by multiple morphological and biochemical criteria, bear very little resemblance to classical apoptosis. Death of syncytia is associated with nuclear fusion and premature chromosome condensation as well as severe ATP depletion and autophagic degeneration, accompanied by release of vesicles reminiscent of exosomes (syncytiosomes). Dying syncytia produce significantly more syncytiosomes than normal cells or cells killed by irradiation, freeze thaw, or osmotic shock. These syncytiosomes also load dendritic cells (DCs) more effectively than exosomes from cells dying by other mechanisms. Finally, we demonstrate that syncytiosomes from either autologous or allogeneic fusing melanoma cells lead to cross-presentation of a defined tumor antigen, gp100, by DCs to a gp100-specific CTL clone. Cross-presentation was significantly more efficient than that with exosomes from normal, irradiated, or herpes simplex virus thymidine kinase/ganciclovir-killed tumor cells. Therefore, FMG-mediated cell killing combines very effective local tumor cell killing with the potential to be a highly immunogenic method of cytotoxic gene therapy. In addition, these data open the way for novel methods of loading DCs with relevant tumor-associated antigens for vaccine development.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Proteínas Virales de Fusión/inmunología , Apoptosis/fisiología , Caspasa 3 , Caspasas/metabolismo , Muerte Celular/inmunología , Grupo Citocromo c/metabolismo , Citoesqueleto/metabolismo , Citotoxicidad Inmunológica , Activación Enzimática , Precursores Enzimáticos/metabolismo , Fibrosarcoma/genética , Fibrosarcoma/inmunología , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Fase G2/fisiología , Terapia Genética/métodos , Células Gigantes/inmunología , Células Gigantes/patología , Humanos , Glicoproteínas de Membrana/metabolismo , Mitocondrias/metabolismo , Mitosis/fisiología , Transfección , Células Tumorales Cultivadas , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética
18.
Mol Ther Oncolytics ; 3: 16030, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27933315

RESUMEN

We showed previously that therapy with Vesicular Stomatitis Virus (VSV) expressing tumor-associated proteins eradicates established tumors. We show here that when cellular cDNA were cloned into VSV which retained their own poly-A signal, viral species emerged in culture which had deleted the cellular poly-A signal and also contained a truncated form of the protein coding sequence. Typically, the truncation occurred such that a Tyrosine-encoding codon was converted into a STOP codon. We believe that the truncation of tumor-associated proteins expressed from VSV in this way occurred to preserve the ability of the virus to replicate efficiently. Truncated cDNA expressed from VSV were significantly more effective than full length cDNA in treating established tumors. Moreover, tumor therapy with truncated cDNA was completely abolished by depletion of CD4+ T cells, whereas therapy with full length cDNA was CD8+ T cell dependent. These data show that the type/potency of antitumor immune responses against self-tumor-associated proteins can be manipulated in vivo through the nature of the self protein (full length or truncated). Therefore, in addition to generation of neoantigens through sequence mutation, immunological tolerance against self-tumor-associated proteins can be broken through manipulation of protein integrity, allowing for rational design of better self-immunogens for cancer immunotherapy.

20.
Eur J Med Chem ; 103: 238-51, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26360048

RESUMEN

Within the constantly growing number of histamine H4 (H4R) receptor ligands there is a large group of azine derivatives. A series of novel compounds in the group of 4-methylpiperazine-1,3,5-triazine-2-amines were designed and obtained. Considered structures were modified at the triazine 6-position by introduction of variously substituted arylethenyl moieties. Their affinities to histamine H4 receptors were evaluated in radioligand binding assays with use of Sf9 cells, transiently expressing human H4R. Pharmacological studies results allowed to identify 4-[(E)-2-(3-chlorophenyl)ethenyl]-6-(4-methylpiperazin-1-yl)-1,3,5-triazin-2-amine (Ki = 253 nM) as the most potent compound in the present series.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Receptores Histamínicos/metabolismo , Triazinas/metabolismo , Triazinas/farmacología , Animales , Antiinflamatorios/síntesis química , Antiinflamatorios/química , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Ratones , Estructura Molecular , Ensayo de Unión Radioligante , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Histamínicos H4 , Relación Estructura-Actividad , Especificidad por Sustrato , Triazinas/síntesis química , Triazinas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA