Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Cell Death Dis ; 14(2): 129, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792589

RESUMEN

Lipid and cholesterol metabolism play a crucial role in tumor cell behavior and in shaping the tumor microenvironment. In particular, enzymatic and non-enzymatic cholesterol metabolism, and derived metabolites control dendritic cell (DC) functions, ultimately impacting tumor antigen presentation within and outside the tumor mass, dampening tumor immunity and immunotherapeutic attempts. The mechanisms accounting for such events remain largely to be defined. Here we perturbed (oxy)sterol metabolism genetically and pharmacologically and analyzed the tumor lipidome landscape in relation to the tumor-infiltrating immune cells. We report that perturbing the lipidome of tumor microenvironment by the expression of sulfotransferase 2B1b crucial in cholesterol and oxysterol sulfate synthesis, favored intratumoral representation of monocyte-derived antigen-presenting cells, including monocyte-DCs. We also found that treating mice with a newly developed antagonist of the oxysterol receptors Liver X Receptors (LXRs), promoted intratumoral monocyte-DC differentiation, delayed tumor growth and synergized with anti-PD-1 immunotherapy and adoptive T cell therapy. Of note, looking at LXR/cholesterol gene signature in melanoma patients treated with anti-PD-1-based immunotherapy predicted diverse clinical outcomes. Indeed, patients whose tumors were poorly infiltrated by monocytes/macrophages expressing LXR target genes showed improved survival over the course of therapy. Thus, our data support a role for (oxy)sterol metabolism in shaping monocyte-to-DC differentiation, and in tumor antigen presentation critical for responsiveness to immunotherapy. The identification of a new LXR antagonist opens new treatment avenues for cancer patients.


Asunto(s)
Melanoma , Monocitos , Ratones , Animales , Monocitos/metabolismo , Diferenciación Celular , Colesterol/metabolismo , Presentación de Antígeno , Células Dendríticas/metabolismo , Microambiente Tumoral
2.
Hum Gene Ther ; 32(13-14): 744-760, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33554732

RESUMEN

Effectiveness of adoptively transferred chimeric antigen receptor (CAR) T cells strongly depends on the quality of CAR-mediated interaction of the effector cells with the target antigen on tumor cells. A major role in this interaction is played by the affinity of the single-chain variable fragment (scFv) for the antigen, and by the CAR design. In particular, the spacer domain may impact on the CAR T cell function by affecting the length and flexibility of the resulting CAR. This study addresses the need to improve the manufacturing process and the antitumor activity of CD44v6-specific CAR T cells by defining the optimal structure of a spacer region derived from the extracellular domain of the human low-affinity nerve growth factor receptor (LNGFR). We tailored the LNGFR spacer to modulate CAR length to efficiently recognize distal or proximal epitopes and to allow selection of transduced CAR T cells by the use of clinical-grade validated manufacturing systems. The different LNGFR spacers investigated in this study are responsible for the generation of CAR T cells with a different memory phenotype, which is mainly related to the level of CAR expression and the extent of the associated tonic signaling. In particular, the CD44v6-NWN2.CAR T cells are enriched in central memory cells and show improved in vitro functions in terms of killing capability, and in vivo antitumor activity against hematological and solid tumors. Clinical Trial Registration numbers: clinicaltrial.gov NCT04097301; ClinicalTrials.gov, NCT00423124.


Asunto(s)
Receptores Quiméricos de Antígenos , Línea Celular Tumoral , Humanos , Inmunoterapia Adoptiva , Receptor de Factor de Crecimiento Nervioso , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Receptores de Factor de Crecimiento Nervioso , Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Front Immunol ; 11: 99, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117253

RESUMEN

The main challenge of adoptive therapy with Chimeric Antigen Receptor modified T cells (CAR T) is the application to the field of solid tumors, where the identification of a proper antigen has emerged as one of the major drawbacks to CAR T cell treatment success. CD44 is a glycoprotein involved in cell-cell and cell-matrix interactions. The isoform containing the variant domain 6 of CD44 gene (CD44v6) has been implicated in tumorigenesis, tumor cell invasion and metastasis and represents an attractive target for CAR T cell therapies. Targeting CD44v6 antigen has been shown to control tumor growth in acute myeloid leukemia and multiple myeloma mouse models. While CAR T approach for the treatment of B cell malignancies has shown great success, response rates among patients with solid cancer are less favorable. The purpose of our study was to test the efficacy of CD44v6.CAR T cells, produced in compliance with Good Manufacturing Practice (GMP), in adenocarcinoma tumor models. We generated a bicistronic retroviral vector containing the CD44v6 CAR and the HSV-TK Mut2 suicide gene to enhance the safety of the proposed CAR T cell therapy. CD44v6 transduced CAR T cells were homogeneously positive for ΔLNGFR selection marker, were enriched in T central memory (TCM) and T memory stem cells (TSCM) and displayed a highly activated phenotype. In vitro assays revealed antigen-specific activation and cytotoxicity of human CD44v6.CAR T cells against CD44v6 expressing tumor cell lines. When infused in immunodeficient tumor bearing mice, human CD44v6.CAR T cells were able to reach, infiltrate and proliferate at tumor sites, finally resulting in tumor growth control. Next, we checked if cells produced in compliance with GMP grade standards retained the same antitumor activity of those produced with research grade materials and protocols. Noteworthy, no differences in the potency of the CAR T obtained with the two manufacturing processes were observed. In conclusion, our preclinical results suggest that CD44v6.CAR T based adoptive therapy could be a promising strategy in solid cancer treatment.


Asunto(s)
Adenocarcinoma/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/uso terapéutico , Adenocarcinoma/inmunología , Animales , Antígenos CD19 , Línea Celular Tumoral , Proliferación Celular , Femenino , Genes Transgénicos Suicidas , Humanos , Receptores de Hialuranos/genética , Inmunoterapia Adoptiva , Pulmón/patología , Ratones , Ratones Transgénicos , Terapia Molecular Dirigida , Ovario/metabolismo , Ovario/patología , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología
4.
Int J Mol Sci ; 20(18)2019 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-31547231

RESUMEN

NGR-hTNF is a therapeutic agent for a solid tumor that specifically targets angiogenic tumor blood vessels, through the NGR motif. Its activity has been assessed in several clinical studies encompassing tumors of different histological types. The drug's activity is based on an improved permeabilization of newly formed tumor vasculature, which favors intratumor penetration of chemotherapeutic agents and leukocyte trafficking. This work investigated the binding and the signaling properties of the NGR-hTNF, to elucidate its mechanism of action. The crystal structure of NGR-hTNF and modeling of its interaction with TNFR suggested that the NGR region is available for binding to a specific receptor. Using 2D TR-NOESY experiments, this study confirmed that the NGR-peptides binds to a specific CD13 isoform, whose expression is restricted to tumor vasculature cells, and to some tumor cell lines. The interaction between hTNF or NGR-hTNF with immobilized TNFRs showed similar kinetic parameters, whereas the competition experiments performed on the cells expressing both TNFR and CD13 showed that NGR-hTNF had a higher binding affinity than hTNF. The analysis of the NGR-hTNF-triggered signal transduction events showed a specific impairment in the activation of pro-survival pathways (Ras, Erk and Akt), compared to hTNF. Since a signaling pattern identical to NGR-hTNF was obtained with hTNF and NGR-sequence given as distinct molecules, the inhibition observed on the survival pathways was presumably due to a direct effect of the NGR-CD13 engagement on the TNFR signaling pathway. The reduced activation of the pro survival pathways induced by NGR-hTNF correlated with the increased caspases activation and reduced cell survival. This study demonstrates that the binding of the NGR-motif to CD13 determines not only the homing of NGR-hTNF to tumor vessels, but also the increase in its antiangiogenic activity.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias/irrigación sanguínea , Oligopéptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Inhibidores de la Angiogénesis/química , Línea Celular Tumoral , Cristalografía por Rayos X , Células Endoteliales de la Vena Umbilical Humana , Humanos , Modelos Moleculares , Oligopéptidos/química , Proteínas Recombinantes de Fusión/química , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/química
5.
J Med Chem ; 61(17): 7474-7485, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-29883545

RESUMEN

The isoDGR sequence is an integrin-binding motif that has been successfully employed as a tumor-vasculature-homing molecule or for the targeted delivery of drugs and diagnostic agents to tumors. In this context, we previously demonstrated that cyclopeptide 2, the product of the conjugation of c(CGisoDGRG) (1) to 4-( N-maleimidomethyl)cyclohexane-1-carboxamide, can be successfully used as a tumor-homing ligand for nanodrug delivery to neoplastic tissues. Here, combining NMR, computational, and biochemical methods, we show that the succinimide ring contained in 2 contributes to stabilizing interactions with αvß3, an integrin overexpressed in the tumor vasculature. Furthermore, we demonstrate that various cyclopeptides containing the isoDGR sequence embedded in different molecular scaffolds do not induce αvß3 allosteric activation and work as pure integrin antagonists. These results could be profitably exploited for the rational design of novel isoDGR-based ligands and tumor-targeting molecules with improved αvß3-binding properties and devoid of adverse integrin-activating effects.


Asunto(s)
Integrina alfaVbeta3/metabolismo , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Succinimidas/química , Regulación Alostérica , Unión Competitiva , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células HEK293 , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/química , Espectroscopía de Resonancia Magnética , Melanoma/patología , Simulación del Acoplamiento Molecular , Péptidos Cíclicos/farmacología , Conformación Proteica , Venenos de Serpiente/farmacología , Relación Estructura-Actividad , Tirosina/metabolismo
6.
Nat Med ; 24(6): 739-748, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29808007

RESUMEN

In the clinic, chimeric antigen receptor-modified T (CAR T) cell therapy is frequently associated with life-threatening cytokine-release syndrome (CRS) and neurotoxicity. Understanding the nature of these pathologies and developing treatments for them are hampered by the lack of appropriate animal models. Herein, we describe a mouse model recapitulating key features of CRS and neurotoxicity. In humanized mice with high leukemia burden, CAR T cell-mediated clearance of cancer triggered high fever and elevated IL-6 levels, which are hallmarks of CRS. Human monocytes were the major source of IL-1 and IL-6 during CRS. Accordingly, the syndrome was prevented by monocyte depletion or by blocking IL-6 receptor with tocilizumab. Nonetheless, tocilizumab failed to protect mice from delayed lethal neurotoxicity, characterized by meningeal inflammation. Instead, the IL-1 receptor antagonist anakinra abolished both CRS and neurotoxicity, resulting in substantially extended leukemia-free survival. These findings offer a therapeutic strategy to tackle neurotoxicity and open new avenues to safer CAR T cell therapies.


Asunto(s)
Inmunoterapia Adoptiva/efectos adversos , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Monocitos/metabolismo , Neurotoxinas/toxicidad , Receptores Quiméricos de Antígenos/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Línea Celular Tumoral , Células Madre Hematopoyéticas/metabolismo , Humanos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Leucemia/inmunología , Leucemia/patología , Ratones , Síndrome
7.
Front Immunol ; 9: 507, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29619024

RESUMEN

Chimeric antigen receptor (CAR)-T cell immunotherapy is at the forefront of innovative cancer therapeutics. However, lack of standardization of cellular products within the same clinical trial and lack of harmonization between different trials have hindered the clear identification of efficacy and safety determinants that should be unveiled in order to advance the field. With the aim of facilitating the isolation and in vivo tracking of CAR-T cells, we here propose the inclusion within the CAR molecule of a novel extracellular spacer based on the low-affinity nerve-growth-factor receptor (NGFR). We screened four different spacer designs using as target antigen the CD44 isoform variant 6 (CD44v6). We successfully generated NGFR-spaced CD44v6 CAR-T cells that could be efficiently enriched with clinical-grade immuno-magnetic beads without negative consequences on subsequent expansion, immuno-phenotype, in vitro antitumor reactivity, and conditional ablation when co-expressing a suicide gene. Most importantly, these cells could be tracked with anti-NGFR monoclonal antibodies in NSG mice, where they expanded, persisted, and exerted potent antitumor effects against both high leukemia and myeloma burdens. Similar results were obtained with NGFR-enriched CAR-T cells specific for CD19 or CEA, suggesting the universality of this strategy. In conclusion, we have demonstrated that the incorporation of the NGFR marker gene within the CAR sequence allows for a single molecule to simultaneously work as a therapeutic and selection/tracking gene. Looking ahead, NGFR spacer enrichment might allow good manufacturing procedures-manufacturing of standardized CAR-T cell products with high therapeutic potential, which could be harmonized in different clinical trials and used in combination with a suicide gene for future application in the allogeneic setting.


Asunto(s)
Inmunoterapia Adoptiva , Proteínas del Tejido Nervioso/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores de Factor de Crecimiento Nervioso/inmunología , Linfocitos T/inmunología , Timidina Quinasa/genética , Animales , Línea Celular Tumoral , Genes Transgénicos Suicidas , Receptores de Hialuranos/inmunología , Leucemia/terapia , Ratones , Mieloma Múltiple/terapia , Proteínas del Tejido Nervioso/genética , Receptores Quiméricos de Antígenos/genética , Receptores de Factor de Crecimiento Nervioso/genética
8.
Proc Natl Acad Sci U S A ; 113(41): E6219-E6227, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27671648

RESUMEN

Cells in the tumor microenvironment may be reprogrammed by tumor-derived metabolites. Cholesterol-oxidized products, namely oxysterols, have been shown to favor tumor growth directly by promoting tumor cell growth and indirectly by dampening antitumor immune responses. However, the cellular and molecular mechanisms governing oxysterol generation within tumor microenvironments remain elusive. We recently showed that tumor-derived oxysterols recruit neutrophils endowed with protumoral activities, such as neoangiogenesis. Here, we show that hypoxia inducible factor-1a (HIF-1α) controls the overexpression of the enzyme Cyp46a1, which generates the oxysterol 24-hydroxycholesterol (24S-HC) in a pancreatic neuroendocrine tumor (pNET) model commonly used to study neoangiogenesis. The activation of the HIF-1α-24S-HC axis ultimately leads to the induction of the angiogenic switch through the positioning of proangiogenic neutrophils in proximity to Cyp46a1+ islets. Pharmacologic blockade or genetic inactivation of oxysterols controls pNET tumorigenesis by dampening the 24S-HC-neutrophil axis. Finally, we show that in some human pNET samples Cyp46a1 transcripts are overexpressed, which correlate with the HIF-1α target VEGF and with tumor diameter. This study reveals a layer in the angiogenic switch of pNETs and identifies a therapeutic target for pNET patients.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Hidroxicolesteroles/metabolismo , Tumores Neuroendocrinos/etiología , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/metabolismo , Animales , Transformación Celular Neoplásica/genética , Colestanotriol 26-Monooxigenasa/genética , Colestanotriol 26-Monooxigenasa/metabolismo , Colesterol 24-Hidroxilasa , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Activación Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Neovascularización Patológica/genética , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Cancer Immunol Immunother ; 65(11): 1303-1315, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27520505

RESUMEN

Tumor-derived metabolites dampen tumor-infiltrating immune cells and antitumor immune responses. Among the various metabolites produced by tumors, we recently showed that cholesterol oxidized products, namely oxysterols, favor tumor growth through the inhibition of DC migration toward lymphoid organs and by promoting the recruitment of pro-tumor neutrophils within the tumor microenvironment. Here, we tested different drugs capable of blocking cholesterol/oxysterol formation. In particular, we tested efficacy and safety of different administration schedules, and of immunotherapy-based combination of a class of compounds, namely zaragozic acids, which inhibit cholesterol pathway downstream of mevalonate formation, thus leaving intact the formation of the isoprenoids, which are required for the maturation of proteins involved in the immune cell function. We show that zaragozic acids inhibit the in vivo growth of the RMA lymphoma and the Lewis lung carcinoma (LLC) without inducing side effects. Tumor growth inhibition requires an intact immune system, as immunodeficient tumor-bearing mice do not respond to zaragozic acid treatment. Of note, the effect of zaragozic acids is accompanied by a marked reduction in the LXR target genes Abcg1, Mertk, Scd1 and Srebp-1c in the tumor microenvironment. On the other hand, zoledronate, which blocks also isoprenoid formation, did not control the LLC tumor growth. Finally, we show that zaragozic acids potentiate the antitumor effects of active and adoptive immunotherapy, significantly prolonging the overall survival of tumor-bearing mice treated with the combo zaragozic acids and TAA-loaded DCs. This study identifies zaragozic acids as new antitumor compounds exploitable for the treatment of cancer patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Carcinoma Pulmonar de Lewis/terapia , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Linfoma de Células T/terapia , Ácidos Tricarboxílicos/uso terapéutico , Animales , Carcinoma Pulmonar de Lewis/inmunología , Colesterol/metabolismo , Terapia Combinada , Células Dendríticas/trasplante , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfoma de Células T/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Oxiesteroles/metabolismo , Escape del Tumor , Microambiente Tumoral
10.
Methods Mol Biol ; 1393: 97-104, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27033219

RESUMEN

The exploitation of the physiologic processing and presenting machinery of dendritic cells (DCs) by in vivo loading of tumor-associated antigens may improve the immunogenic potential and clinical efficacy of DC-based cancer vaccines. The approach developed by our group was based on the clinical observation that some patients treated with the infusion of donor lymphocytes transduced to express the HSV-TK suicide gene for relapse of hematologic malignancies, after allogeneic hematopoietic stem cell transplantation, developed a T cell-mediated immune response specifically directed against the HSV-TK gene product.We demonstrated that lymphocytes genetically modified to express HSV-TK as well as self/tumor antigens, acting as antigen carriers, efficiently target DCs in vivo in tumor-bearing mice. The infusion of TRP-2-transduced lymphocytes induced the establishment of protective immunity and long-term memory in tumor-bearing mice by cross-presentation of the antigen mediated by the CD11c(+)CD8a(+) DCs subset. A similar approach was applied in a clinical setting. Ten patients affected by MAGE-3(+) metastatic melanoma were treated with autologous lymphocytes retrovirally transduced to express the MAGE-3 tumor antigen. In three patients, the treatment led to the increase of MAGE-3 specific CD8+ and CD4+ effectors and the development of long-term memory, which ultimately correlated with a favorable clinical outcome. Transduced lymphocytes represent an efficient way for in vivo loading of tumor-associated antigens of DCs.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Proteínas de Neoplasias/inmunología , Vacunación , Animales , Reactividad Cruzada , Células Dendríticas/inmunología , Humanos , Memoria Inmunológica , Ratones
11.
Sci Transl Med ; 7(317): 317ra198, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26659572

RESUMEN

Long-lasting immune protection from pathogens and cancer requires the generation of memory T cells able to survive long-term. To unravel the immunological requirements for long-term persistence of human memory T cells, we characterized and traced, over several years, T lymphocytes genetically modified to express the thymidine kinase (TK) suicide gene that were infused in 10 patients after haploidentical hematopoietic stem cell transplantation (HSCT). At 2 to 14 years after infusion and in the presence of a broad and resting immune system, we could still detect effectors/effector memory (TEM/EFF), central memory (TCM), and stem memory (TSCM) TK(+) cells, circulating at low but stable levels in all patients. Longitudinal analysis of cytomegalovirus (CMV)- and Flu-specific TK(+) cells indicated that antigen recognition was dominant in driving in vivo expansion and persistence at detectable levels. The amount of infused TSCM cells positively correlated with early expansion and with the absolute counts of long-term persisting gene-marked cells. By combining T cell sorting with sequencing of integration (IS), TCRα and TCRß clonal markers, we showed that T cells retrieved long-term were enriched in clones originally shared in different memory T cell subsets, whereas dominant long-term clonotypes appeared to preferentially originate from infused TSCM and TCM clones. Together, these results indicate that long-term persistence of gene-modified memory T cells after haploidentical HSCT is influenced by antigen exposure and by the original phenotype of infused cells. Cancer adoptive immunotherapy might thus benefit from cellular products enriched in lymphocytes with an early-differentiated phenotype.


Asunto(s)
Rastreo Celular , Ingeniería Genética , Memoria Inmunológica , Linfocitos T/inmunología , Adulto , Anciano , Antígenos/inmunología , Proliferación Celular , Células Clonales , Femenino , Genes Transgénicos Suicidas , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Recuento de Linfocitos , Depleción Linfocítica , Masculino , Persona de Mediana Edad , Fenotipo , Timidina Quinasa/metabolismo , Factores de Tiempo , Donantes de Tejidos , Adulto Joven
12.
Oncoimmunology ; 4(10): e1041700, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26451306

RESUMEN

NGR-TNF is a vascular targeting agent in advanced clinical development, coupling tumor necrosis factor-α (TNF) with the CNGRCG peptide, which targets a CD13 isoform specifically expressed by angiogenic vessels. Antitumor efficacy of NGR-TNF has been described in different transplantation tumor models. Nevertheless, the mechanism underlying its activity is not fully understood. In the wild type and in the immunodeficient (RAG-/-) RIP1-Tag2 models of multistage pancreatic carcinogenesis, we demonstrate that CD13 is highly expressed on endothelial cells of hyperplastic and angiogenic islets, whereas its expression is down regulated in tumors where it partially colocalize with pericytes. In vivo CNGRCG peptides coupled to fluorescent nanoparticles (quantum dots) bind to CD13 and colocalize with anti-CD31, in pancreatic islets. At early stage, low doses of NGR-murine (m)TNF have a direct cytotoxic effect inducing endothelial cell apoptosis, reducing vessel density and eventually inhibiting the development of angiogenic islets. At a later stage, NGR-mTNF is able to reduce tumor growth inducing vascular normalization, exclusively when treatment is carried out in the immunocompetent mice. Interestingly, NGR-mTNF-treated tumors from these mice are characterized by CD8+ T cell infiltration. At molecular level, overexpression of genes involved in vessels normalization was detected only in NGR-mTNF-treated tumors from immunocompetent mice. These findings identified a new mechanism of action of NGR-mTNF, providing support for the development of new therapeutic strategies combining chemotherapy or active/adoptive immunotherapies to low dose NGR-TNF treatment.

13.
Clin Exp Metastasis ; 32(3): 289-300, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25648442

RESUMEN

Tumor vessels are an attractive target for cancer therapy, including metastasis treatment. Angiogenesis inhibitors targeting the VEGF signalling pathway have proven to be efficacious in preclinical cancer models and in clinical trials. However, angiogenesis inhibition concomitantly elicits tumor adaptation and progression to stages of greater malignancy, with heightened invasiveness and in some cases increased distant metastasis. Here, we investigated whether NGR-TNF, a vascular targeting agent in phase III clinical development, coupling the CNGRCG angiogenic vessel-homing peptide with TNF-α, has an effect on metastasis in a model of murine breast cancer, which spontaneously metastasize to lungs, and on the growth of experimental melanoma lung metastasis. We report that NGR-TNF does not increase cancer invasiveness, as other antiangiogenics agents do, but controls metastatic growth in both models, both when administered as primary treatment and in adjuvant settings, improving the overall survival of metastasis-bearing mice.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Mamarias Animales/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Proteínas Recombinantes de Fusión/uso terapéutico , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Femenino , Citometría de Flujo , Técnicas para Inmunoenzimas , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Animales/mortalidad , Neoplasias Mamarias Animales/patología , Melanoma Experimental/mortalidad , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Tasa de Supervivencia , Células Tumorales Cultivadas
14.
Eur J Immunol ; 44(7): 1896-903, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24777958

RESUMEN

Oxysterols are involved in maintaining cellular cholesterol levels. Recently, oxysterols have been demonstrated to modulate the function of immune cells and tumor growth. These effects can be dependent on the activation of the oxysterol-binding liver X receptors (LXRs) or, as recently demonstrated for T and B cells, DCs and neutrophils, can be independent of LXR activation. LXR-dependent oxysterol effects can be ascribed to the activation of LXRα, LXRß or LXRαß isoforms, which induces transcriptional activation or trans-repression of target genes. The prevalent activation of one isoform seems to be cell-, tissue-, or context-specific, as shown in some pathologic processes, i.e., infectious diseases, atherosclerosis, and autoimmunity. Oxysterol-LXR signaling has recently been shown to inhibit antitumor immune responses, as well as to modulate tumor cell growth. Here, we review the mechanisms that link oxysterols to tumor growth, and discuss possible networks at the basis of LXR-dependent and -independent oxysterol effects on immune cells and tumor development.


Asunto(s)
Colesterol/metabolismo , Hidroxicolesteroles/metabolismo , Inmunidad , Neoplasias/patología , Receptores Nucleares Huérfanos/fisiología , Animales , Células Dendríticas/inmunología , Humanos , Receptores X del Hígado , Linfocitos/inmunología , Macrófagos/fisiología , Monocitos/fisiología , Receptores CCR7/fisiología , Microambiente Tumoral
15.
J Exp Med ; 210(9): 1711-28, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23897983

RESUMEN

Tumor-infiltrating immune cells can be conditioned by molecules released within the microenvironment to thwart antitumor immune responses, thereby facilitating tumor growth. Among immune cells, neutrophils play an important protumorigenic role by favoring neoangiogenesis and/or by suppressing antitumor immune responses. Tumor-derived oxysterols have recently been shown to favor tumor growth by inhibiting dendritic cell migration toward lymphoid organs. We report that tumor-derived oxysterols recruit protumor neutrophils in a liver X receptor (LXR)-independent, CXCR2-dependent manner, thus favoring tumor growth by promoting neoangiogenesis and immunosuppression. We demonstrate that interfering with the oxysterol-CXCR2 axis delays tumor growth and prolongs the overall survival of tumor-bearing mice. These results identify an unanticipated protumor function of the oxysterol-CXCR2 axis and a possible target for cancer therapy.


Asunto(s)
Neoplasias/metabolismo , Neutrófilos/metabolismo , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Esteroles/metabolismo , Animales , Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Proliferación Celular , Quimiotaxis , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Hidroxicolesteroles/metabolismo , Terapia de Inmunosupresión , Ligandos , Receptores X del Hígado , Espectrometría de Masas , Ratones , Células Mieloides/metabolismo , Células Mieloides/patología , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Receptores Nucleares Huérfanos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
16.
Cancer Res ; 73(2): 804-12, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23151901

RESUMEN

Poor penetration of antitumor drugs into the extravascular tumor tissue is often a major factor limiting the efficacy of cancer treatments. Our group has recently described a strategy to enhance tumor penetration of chemotherapeutic drugs through use of iRGD peptide (CRGDK/RGPDC). This peptide comprises two sequence motifs: RGD, which binds to αvß3/5 integrins on tumor endothelia and tumor cells, and a cryptic CendR motif (R/KXXR/K-OH). Once integrin binding has brought iRGD to the tumor, the peptide is proteolytically cleaved to expose the cryptic CendR motif. The truncated peptide loses affinity for its primary receptor and binds to neuropilin-1, activating a tissue penetration pathway that delivers the peptide along with attached or co-administered payload into the tumor mass. Here, we describe the design of a new tumor-penetrating peptide based on the current knowledge of homing sequences and internalizing receptors. The tumor-homing motif in the new peptide is the NGR sequence, which binds to endothelial CD13. The NGR sequence was placed in the context of a CendR motif (RNGR), and this sequence was embedded in the iRGD framework. The resulting peptide (CRNGRGPDC, iNGR) homed to tumor vessels and penetrated into tumor tissue more effectively than the standard NGR peptide. iNGR induced greater tumor penetration of coupled nanoparticles and co-administered compounds than NGR. Doxorubicin given together with iNGR was significantly more efficacious than the drug alone. These results show that a tumor-specific, tissue-penetrating peptide can be constructed from known sequence elements. This principle may be useful in designing tissue-penetrating peptides for other diseases.


Asunto(s)
Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Ratones , Unión Proteica
17.
Int J Cancer ; 132(11): 2557-66, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23151995

RESUMEN

Cancer vaccines have recently been shown to induce some clinical benefits. The relationship between clinical activity and anti-vaccine T cell responses is somewhat controversial. Indeed, in many trials it has been documented that the induction of vaccine-specific T cells exceeds the clinical responses observed. Here, we evaluate immunological and clinical responses in 23 MAGE-A3(+) melanoma patients treated with autologous lymphocytes genetically engineered to express the tumor antigen MAGE-A3 and the viral gene product thymidine kinase of the herpes simplex virus (HSV-TK). HSV-TK was used as safety system in case of adverse events and as tracer antigen to monitor the immune competence of treated patients. The increase of anti-TK and anti-MAGE-A3 T-cells after vaccination was observed in 90 and 27% of patients, respectively. Among 19 patients with measurable disease, we observed a disease control rate of 26.3%, with one objective clinical response, and four durable, stable diseases. Three patients out of five with no evidence of disease (NED) at the time of vaccination remained NED after 73+, 70+ and 50+ months. Notably, we report that only patients experiencing MAGE-A3-specific immune responses showed a clinical benefit. Additionally, we report that responder and non-responder patients activate and expand T cells against the tracer antigen TK in a similar way, suggesting that local rather than systemic immune suppression might be involved in limiting clinically relevant antitumor immune responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Terapia Genética , Melanoma/inmunología , Proteínas de Neoplasias/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Neoplasias Óseas/inmunología , Neoplasias Óseas/mortalidad , Neoplasias Óseas/terapia , Ensayos Clínicos Fase II como Asunto , Femenino , Estudios de Seguimiento , Humanos , Hipersensibilidad Tardía , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/terapia , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Melanoma/mortalidad , Melanoma/terapia , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/terapia , Linfocitos T/metabolismo , Timidina Quinasa/inmunología , Timidina Quinasa/metabolismo
18.
Oncoimmunology ; 2(11): e26469, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24490125

RESUMEN

By binding to the liver X receptor (LXR), oxysterols inhibit the expression of chemokine (C-C motif) receptor 7 (CCR7), hence impairing the migration of dendritic cells to secondary lymphoid organs and inhibiting antitumor immune responses. We have recently identified a new tumor-supporting activity of oxysterols, which recruit neutrophils within tumor microenvironment by a chemokine (C-X-C motif) receptor 2 (CXCR2)-dependent, LXR-independent mechanism.

19.
Mol Ther ; 20(10): 1932-43, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22910293

RESUMEN

Despite the genotoxic complications encountered in clinical gene therapy trials for primary immunodeficiency diseases targeting hematopoietic cells with integrating vectors; this strategy holds promise for the cure of several monogenic blood, metabolic and neurodegenerative diseases. In this study, we asked whether the inclusion of a suicide gene in a standard retrovirus vector would allow elimination of vector-containing stem and progenitor cells and their progeny in vivo following transplantation, using our rhesus macaque transplantation model. Following stable engraftment with autologous CD34(+) cells transduced with a retrovirus vector encoding a highly sensitive modified Herpes simplex virus thymidine kinase SR39, the administration of the antiviral prodrug ganciclovir (GCV) was effective in completely eliminating vector-containing cells in all hematopoietic lineages in vivo. The sustained absence of vector-containing cells over time, without additional GCV administration, suggests that the ablation of TkSR39 GCV-sensitive cells occurred in the most primitive hematopoietic long-term repopulating stem or progenitor cell compartment. These results are a proof-of-concept that the inclusion of a suicide gene in integrating vectors, in addition to a therapeutic gene, can provide a mechanism for later elimination of vector-containing cells, thereby increasing the safety of gene transfer.


Asunto(s)
Ganciclovir/uso terapéutico , Genes Transgénicos Suicidas , Vectores Genéticos , Hematopoyesis/genética , Timidina Quinasa/genética , Animales , Antivirales/uso terapéutico , Replicación del ADN , Terapia Genética/métodos , Células Madre Hematopoyéticas/citología , Macaca mulatta , Retroviridae/genética , Transducción Genética
20.
Curr Opin Pharmacol ; 12(6): 729-35, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22832233

RESUMEN

Oxysterols/oxysterol receptors have been shown to modulate several immune cell subsets, such as macrophages, T-cells and B-cells, neutrophils and dendritic cells (DCs). They participate in the control of several pathologic processes, that is, infectious diseases, atherosclerosis and autoimmunity. Moreover, some oxysterols have also been shown to favor tumor progression by dampening the antitumor immune response. The cellular responses generated by oxysterols depend on the engagement of Liver X Receptor (LXR) α and/or ß isoforms, which induce activation of target genes or trans-repression of pro-inflammatory gene transcription. Recently, some reports have described a different mechanism of action of oxysterols, mediated by the engagement of G-Protein Coupled Receptors. Here, we summarize LXR-dependent and LXR-independent responses of oxysterols on immune cells with possible effects on tumor development.


Asunto(s)
Sistema Inmunológico/metabolismo , Neoplasias/patología , Receptores Nucleares Huérfanos/inmunología , Animales , Progresión de la Enfermedad , Humanos , Sistema Inmunológico/citología , Enfermedades del Sistema Inmune/fisiopatología , Receptores X del Hígado , Neoplasias/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Esteroles/inmunología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...