Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Immunity ; 53(3): 658-671.e6, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32937153

RESUMEN

Identifying signals in the tumor microenvironment (TME) that shape CD8+ T cell phenotype can inform novel therapeutic approaches for cancer. Here, we identified a gradient of increasing glucocorticoid receptor (GR) expression and signaling from naïve to dysfunctional CD8+ tumor-infiltrating lymphocytes (TILs). Conditional deletion of the GR in CD8+ TILs improved effector differentiation, reduced expression of the transcription factor TCF-1, and inhibited the dysfunctional phenotype, culminating in tumor growth inhibition. GR signaling transactivated the expression of multiple checkpoint receptors and promoted the induction of dysfunction-associated genes upon T cell activation. In the TME, monocyte-macrophage lineage cells produced glucocorticoids and genetic ablation of steroidogenesis in these cells as well as localized pharmacologic inhibition of glucocorticoid biosynthesis improved tumor growth control. Active glucocorticoid signaling associated with failure to respond to checkpoint blockade in both preclinical models and melanoma patients. Thus, endogenous steroid hormone signaling in CD8+ TILs promotes dysfunction, with important implications for cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Glucocorticoides/metabolismo , Macrófagos/metabolismo , Melanoma Experimental/patología , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Hematopoyesis/inmunología , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Inhibidores de Puntos de Control Inmunológico , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/inmunología
2.
Immunity ; 50(1): 181-194.e6, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30635236

RESUMEN

An improved understanding of the anti-tumor CD8+ T cell response after checkpoint blockade would enable more informed and effective therapeutic strategies. Here we examined the dynamics of the effector response of CD8+ tumor-infiltrating lymphocytes (TILs) after checkpoint blockade therapy. Bulk and single-cell RNA profiles of CD8+ TILs after combined Tim-3+PD-1 blockade in preclinical models revealed significant changes in the transcriptional profile of PD-1- TILs. These cells could be divided into subsets bearing characterstics of naive-, effector-, and memory-precursor-like cells. Effector- and memory-precursor-like TILs contained tumor-antigen-specific cells, exhibited proliferative and effector capacity, and expanded in response to different checkpoint blockade therapies across different tumor models. The memory-precursor-like subset shared features with CD8+ T cells associated with response to checkpoint blockade in patients and was compromised in the absence of Tcf7. Expression of Tcf7/Tcf1 was requisite for the efficacy of diverse immunotherapies, highlighting the importance of this transcriptional regulator in the development of effective CD8+ T cell responses upon immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Experimentales/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Subgrupos de Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Memoria Inmunológica/genética , Inmunoterapia , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Transcriptoma
3.
Nature ; 510(7504): 235-240, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24870228

RESUMEN

Targeted genome editing by artificial nucleases has brought the goal of site-specific transgene integration and gene correction within the reach of gene therapy. However, its application to long-term repopulating haematopoietic stem cells (HSCs) has remained elusive. Here we show that poor permissiveness to gene transfer and limited proficiency of the homology-directed DNA repair pathway constrain gene targeting in human HSCs. By tailoring delivery platforms and culture conditions we overcame these barriers and provide stringent evidence of targeted integration in human HSCs by long-term multilineage repopulation of transplanted mice. We demonstrate the therapeutic potential of our strategy by targeting a corrective complementary DNA into the IL2RG gene of HSCs from healthy donors and a subject with X-linked severe combined immunodeficiency (SCID-X1). Gene-edited HSCs sustained normal haematopoiesis and gave rise to functional lymphoid cells that possess a selective growth advantage over those carrying disruptive IL2RG mutations. These results open up new avenues for treating SCID-X1 and other diseases.


Asunto(s)
Marcación de Gen/métodos , Genoma Humano/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Reparación del Gen Blanco/métodos , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Animales , Antígenos CD34/metabolismo , ADN Complementario/genética , Endonucleasas/metabolismo , Sangre Fetal/citología , Sangre Fetal/metabolismo , Sangre Fetal/trasplante , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Masculino , Ratones , Mutación/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
4.
Blood ; 130(5): 606-618, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28637663

RESUMEN

Transfer of T-cell receptors (TCRs) specific for tumor-associated antigens is a promising approach for cancer immunotherapy. We developed the TCR gene editing technology that is based on the knockout of the endogenous TCR α and ß genes, followed by the introduction of tumor-specific TCR genes, and that proved safer and more effective than conventional TCR gene transfer. Although successful, complete editing requires extensive cell manipulation and 4 transduction procedures. Here we propose a novel and clinically feasible TCR "single editing" (SE) approach, based on the disruption of the endogenous TCR α chain only, followed by the transfer of genes encoding for a tumor-specific TCR. We validated SE with the clinical grade HLA-A2 restricted NY-ESO-1157-165-specific TCR. SE allowed the rapid production of high numbers of tumor-specific T cells, with optimal TCR expression and preferential stem memory and central memory phenotype. Similarly to unedited T cells redirected by TCR gene transfer (TCR transferred [TR]), SE T cells efficiently killed NY-ESO-1pos targets; however, although TR cells proved highly alloreactive, SE cells showed a favorable safety profile. Accordingly, when infused in NSG mice previously engrafted with myeloma, SE cells mediated tumor rejection without inducing xenogeneic graft-versus-host disease, thus resulting in significantly higher survival than that observed in mice treated with TR cells. Overall, single TCR gene editing represents a clinically feasible approach that is able to increase the safety and efficacy of cancer adoptive immunotherapy.


Asunto(s)
Traslado Adoptivo , Edición Génica/métodos , Memoria Inmunológica , Mieloma Múltiple , Proteínas de Neoplasias , Fragmentos de Péptidos , Receptores de Antígenos de Linfocitos T , Linfocitos T , Animales , Línea Celular Tumoral , Femenino , Técnicas de Transferencia de Gen , Enfermedad Injerto contra Huésped , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Mieloma Múltiple/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/trasplante , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cell Rep Med ; 5(7): 101640, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38959885

RESUMEN

CD8+ T cells must persist and function in diverse tumor microenvironments to exert their effects. Thus, understanding common underlying expression programs could better inform the next generation of immunotherapies. We apply a generalizable matrix factorization algorithm that recovers both shared and context-specific expression programs from diverse datasets to a single-cell RNA sequencing (scRNA-seq) compendium of 33,161 CD8+ T cells from 132 patients with seven human cancers. Our meta-single-cell analyses uncover a pan-cancer T cell dysfunction program that predicts clinical non-response to checkpoint blockade in melanoma and highlights CXCR6 as a pan-cancer marker of chronically activated T cells. Cxcr6 is trans-activated by AP-1 and repressed by TCF1. Using mouse models, we show that Cxcr6 deletion in CD8+ T cells increases apoptosis of PD1+TIM3+ cells, dampens CD28 signaling, and compromises tumor growth control. Our study uncovers a TCF1:CXCR6 axis that counterbalances PD1-mediated suppression of CD8+ cell responses and is essential for effective anti-tumor immunity.


Asunto(s)
Antígenos CD28 , Linfocitos T CD8-positivos , Factor Nuclear 1-alfa del Hepatocito , Receptores CXCR6 , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Animales , Humanos , Antígenos CD28/metabolismo , Antígenos CD28/genética , Antígenos CD28/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , Ratones , Receptores CXCR6/metabolismo , Receptores CXCR6/genética , Neoplasias/inmunología , Neoplasias/genética , Neoplasias/patología , Análisis de la Célula Individual/métodos , Transducción de Señal , Microambiente Tumoral/inmunología , Ratones Endogámicos C57BL
6.
Mucosal Immunol ; 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38925529

RESUMEN

Dietary proteins are taken up by intestinal dendritic cells (DCs), cleaved into peptides, loaded to major histocompatibility complexes, and presented to T cells to generate an immune response. Amino acid (AA)-diets do not have the same effects because AAs cannot bind to major histocompatibility complex to activate T cells. Here, we show that impairment in regulatory T cell generation and loss of tolerance in mice fed a diet lacking whole protein is associated with major transcriptional changes in intestinal DCs including downregulation of genes related to DC maturation, activation and decreased gene expression of immune checkpoint molecules. Moreover, the AA-diet had a profound effect on microbiome composition, including an increase in Akkermansia muciniphilia and Oscillibacter and a decrease in Lactococcus lactis and Bifidobacterium. Although microbiome transfer experiments showed that AA-driven microbiome modulates intestinal DC gene expression, most of the unique transcriptional change in DC was linked to the absence of whole protein in the diet. Our findings highlight the importance of dietary proteins for intestinal DC function and mucosal tolerance.

7.
Cancer Cell ; 41(9): 1662-1679.e7, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37625402

RESUMEN

Stem-like CD8+ T cells are regulated by T cell factor 1 (TCF1) and are considered requisite for immune checkpoint blockade (ICB) response. However, recent findings indicate that reliance on TCF1+CD8+ T cells for ICB efficacy may differ across tumor contexts. We find that TCF1 is essential for optimal priming of tumor antigen-specific CD8+ T cells and ICB response in poorly immunogenic tumors that accumulate TOX+ dysfunctional T cells, but is dispensable for T cell priming and therapy response in highly immunogenic tumors that efficiently expand transitory effectors. Importantly, improving T cell priming by vaccination or by enhancing antigen presentation on tumors rescues the defective responses of TCF1-deficient CD8+ T cells upon ICB in poorly immunogenic tumors. Our study highlights TCF1's role during the early stages of anti-tumor CD8+ T cell responses with important implications for guiding optimal therapeutic interventions in cancers with low TCF1+CD8+ T cells and low-neo-antigen expression.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Factor 1 de Transcripción de Linfocitos T , Humanos , Anticuerpos , Antígenos de Neoplasias , Inmunoterapia , Factor 1 de Transcripción de Linfocitos T/genética , Neoplasias/inmunología , Neoplasias/terapia
8.
Trends Cancer ; 8(8): 642-654, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35527216

RESUMEN

Uncovering the mechanisms that control CD8+ T cell function is a major focus of cancer research. Advances in flow cytometry and single-cell transcriptomics have provided unprecedented in-depth resolution of CD8+ T cell states in cancer. However, these technologies fail to capture important spatial information, including cell-cell interactions and tissue localization. The discovery that intra-tumoral immune niches, tertiary lymphoid structures, and the tumor-draining lymph node are key sites of inter-cellular communication has evoked interest in understanding the spatial determinants that regulate CD8+ T cell functions at these sites. We focus on the cellular, as well as the soluble and surface-bound signals that regulate CD8+ T cell phenotypes and functions in a topologically-regulated manner, highlighting where new spatial transcriptomics and imaging technologies can uncover mechanistic insights.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Comunicación Celular , Diferenciación Celular/genética , Humanos , Activación de Linfocitos , Neoplasias/genética , Neoplasias/patología
9.
EMBO Mol Med ; 13(10): e13598, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34459560

RESUMEN

The immunosuppressive microenvironment surrounding tumor cells represents a key cause of treatment failure. Therefore, immunotherapies aimed at reprogramming the immune system have largely spread in the past years. We employed gene transfer into hematopoietic stem and progenitor cells to selectively express anti-tumoral cytokines in tumor-infiltrating monocytes/macrophages. We show that interferon-γ (IFN-γ) reduced tumor progression in mouse models of B-cell acute lymphoblastic leukemia (B-ALL) and colorectal carcinoma (MC38). Its activity depended on the immune system's capacity to respond to IFN-γ and drove the counter-selection of leukemia cells expressing surrogate antigens. Gene-based IFN-γ delivery induced antigen presentation in the myeloid compartment and on leukemia cells, leading to a wave of T cell recruitment and activation, with enhanced clonal expansion of cytotoxic CD8+ T lymphocytes. The activity of IFN-γ was further enhanced by either co-delivery of tumor necrosis factor-α (TNF-α) or by drugs blocking immunosuppressive escape pathways, with the potential to obtain durable responses.


Asunto(s)
Leucemia , Neoplasias , Animales , Presentación de Antígeno , Interferón gamma , Ratones , Células Mieloides , Microambiente Tumoral , Factor de Necrosis Tumoral alfa
10.
Sci Immunol ; 5(53)2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33158974

RESUMEN

Recent advances have redefined a role for T cell factor 1 (TCF1) that goes beyond T cell development and T memory formation and encompasses new functions in the regulation of T cell biology. Here, we discuss the multifaceted and context-dependent role of TCF1 in peripheral T cells, particularly during disease-induced inflammatory states such as autoimmunity, cancer, and chronic infections. Understanding how TCF1 fine-tunes peripheral T cell biology holds the potential to tailor improved immune-targeted therapies.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Neoplasias/inmunología , Factor 1 de Transcripción de Linfocitos T/metabolismo , Linfocitos T/inmunología , Virosis/inmunología , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Comunicación Celular/efectos de los fármacos , Comunicación Celular/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/inmunología , Enfermedad Crónica/tratamiento farmacológico , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Memoria Inmunológica/efectos de los fármacos , Memoria Inmunológica/genética , Neoplasias/tratamiento farmacológico , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Virosis/tratamiento farmacológico , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/inmunología
12.
Nat Commun ; 9(1): 2896, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30042420

RESUMEN

Immunotherapy is emerging as a new pillar of cancer treatment with potential to cure. However, many patients still fail to respond to these therapies. Among the underlying factors, an immunosuppressive tumor microenvironment (TME) plays a major role. Here we show that monocyte-mediated gene delivery of IFNα inhibits leukemia in a mouse model. IFN gene therapy counteracts leukemia-induced expansion of immunosuppressive myeloid cells and imposes an immunostimulatory program to the TME, as shown by bulk and single-cell transcriptome analyses. This reprogramming promotes T-cell priming and effector function against multiple surrogate tumor-specific antigens, inhibiting leukemia growth in our experimental model. Durable responses are observed in a fraction of mice and are further increased combining gene therapy with checkpoint blockers. Furthermore, IFN gene therapy strongly enhances anti-tumor activity of adoptively transferred T cells engineered with tumor-specific TCR or CAR, overcoming suppressive signals in the leukemia TME. These findings warrant further investigations on the potential development of our gene therapy strategy towards clinical testing.


Asunto(s)
Antígenos de Neoplasias/inmunología , Terapia Genética/métodos , Inmunidad/inmunología , Interferones/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Células Cultivadas , Femenino , Regulación Leucémica de la Expresión Génica , Inmunidad/genética , Inmunoterapia Adoptiva/métodos , Interferones/genética , Interferones/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Microambiente Tumoral/genética
13.
EMBO Mol Med ; 8(2): 155-70, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26769348

RESUMEN

Colorectal cancer (CRC) metastatic dissemination to the liver is one of the most life-threatening malignancies in humans and represents the leading cause of CRC-related mortality. Herein, we adopted a gene transfer strategy into mouse hematopoietic stem/progenitor cells to generate immune-competent mice in which TEMs-a subset of Tie2(+) monocytes/macrophages found at peritumoral sites-express interferon-alpha (IFNα), a pleiotropic cytokine with anti-tumor effects. Utilizing this strategy in mouse models of CRC liver metastasis, we show that TEMs accumulate in the proximity of hepatic metastatic areas and that TEM-mediated delivery of IFNα inhibits tumor growth when administered prior to metastasis challenge as well as on established hepatic lesions, improving overall survival. Further analyses unveiled that local delivery of IFNα does not inhibit homing but limits the early phases of hepatic CRC cell expansion by acting on the radio-resistant hepatic microenvironment. TEM-mediated IFNα expression was not associated with systemic side effects, hematopoietic toxicity, or inability to respond to a virus challenge. Along with the notion that TEMs were detected in the proximity of CRC metastases in human livers, these results raise the possibility to employ similar gene/cell therapies as tumor site-specific drug-delivery strategies in patients with CRC.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Neoplasias Colorrectales/complicaciones , Terapia Genética/métodos , Interferón-alfa/metabolismo , Neoplasias Hepáticas/prevención & control , Neoplasias Hepáticas/terapia , Metástasis de la Neoplasia/prevención & control , Animales , Neoplasias Colorrectales/prevención & control , Neoplasias Colorrectales/terapia , Modelos Animales de Enfermedad , Humanos , Ratones , Metástasis de la Neoplasia/terapia , Análisis de Supervivencia
14.
Cancer Cell ; 28(2): 225-39, 2015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26267536

RESUMEN

The fibroblast growth factor (FGF)/FGF receptor (FGFR) system plays a crucial role in cancer by affecting tumor growth, angiogenesis, drug resistance, and escape from anti-angiogenic anti-vascular endothelial growth factor therapy. The soluble pattern recognition receptor long-pentraxin 3 (PTX3) acts as a multi-FGF antagonist. Here we demonstrate that human PTX3 overexpression in transgenic mice driven by the Tie2 promoter inhibits tumor growth, angiogenesis, and metastasis in heterotopic, orthotopic, and autochthonous FGF-dependent tumor models. Using pharmacophore modeling of the interaction of a minimal PTX3-derived FGF-binding pentapeptide with FGF2, we identified a small-molecule chemical (NSC12) that acts as an extracellular FGF trap with significant implications in cancer therapy.


Asunto(s)
Proteína C-Reactiva/genética , Factores de Crecimiento de Fibroblastos/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Componente Amiloide P Sérico/genética , Animales , Western Blotting , Proteína C-Reactiva/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Estructura Molecular , Neoplasias/metabolismo , Neoplasias/terapia , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Componente Amiloide P Sérico/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
15.
Oncoimmunology ; 3: e28696, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25097805

RESUMEN

An immunosuppressive tumor microenvironment is a cancer hallmark and a major impediment to successful immunotherapy. We engineered hematopoietic progenitors to target expression of an interferon-α (IFNα) transgene specifically to their monocytic progeny, including tumor-infiltrating macrophages. Mice chimeric for these IFNα-expressing macrophages showed activation of innate and adaptive immune cells against breast cancer and inhibited disease progression.

16.
Sci Transl Med ; 6(217): 217ra3, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24382895

RESUMEN

The immunosuppressive tumor microenvironment represents a major hurdle to cancer therapy. We developed a gene transfer strategy into hematopoietic stem cells (HSCs) to target transgene expression to tumor-infiltrating monocytes/macrophages. Using a combination of transcriptional and microRNA-mediated control, we achieved selective expression of an interferon-α (IFN-α) transgene in differentiated monocytes of human hematochimeric mice. We show that IFN-α transgene expression does not impair engraftment and long-term multilineage repopulation of NSG (NOD/LtSz-scidIL2Rγ(null)) mice by transplanted human HSCs. By providing a source of human cytokines in the mice, we improved the functional reconstitution of human myeloid, natural killer, and T cell lineages, and achieved enhanced immune-mediated clearance of transplanted human breast tumors when hematopoiesis was engineered for tumor-targeted IFN-α expression. By applying our strategy to mouse breast cancer models, we achieved inhibition of tumor progression and experimental metastases in an autologous setting, likely through enhanced generation of effector T cells and their recruitment to the neoplastic tissues. By forcing IFN-α expression in tumor-infiltrating macrophages, we blunted their innate protumoral activity and reprogrammed the tumor microenvironment toward more effective dendritic cell activation and immune effector cell cytotoxicity. Overall, our studies validate the feasibility, safety, and therapeutic potential of a new cancer gene therapy strategy, and open the way to test this approach as adjuvant therapy in advanced breast cancer patients.


Asunto(s)
Neoplasias de la Mama/patología , Ingeniería Genética , Hematopoyesis/genética , Interferón-alfa/administración & dosificación , Animales , Neoplasias de la Mama/terapia , Progresión de la Enfermedad , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA