Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Allergy Clin Immunol ; 145(1): 368-378.e13, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31194989

RESUMEN

BACKGROUND: Cryopyrin-associated periodic syndromes (CAPS) are a group of autoinflammatory diseases linked to gain-of-function mutations in the NOD-like receptor family, pyrin domain containing 3 (NLRP3) gene, which cause uncontrolled IL-1ß secretion. Proton pump inhibitors (PPIs), which are commonly used as inhibitors of gastric acid production, also have anti-inflammatory properties, protect mice from sepsis, and prevent IL-1ß secretion by monocytes from patients with CAPS. OBJECTIVE: We sought to develop a novel Nlrp3 knock-in (KI) mouse model of CAPS to study amyloidosis, a severe CAPS complication, and test novel therapeutic approaches. METHODS: We generated KI mice by engineering the N475K mutation, which is associated with the CAPS phenotype, into the mouse Nlrp3 gene. KI and wild-type mice received PPIs or PBS intraperitoneally and were analyzed for survival, inflammation, cytokine secretion, and amyloidosis development. RESULTS: Mutant Nlrp3 KI mice displayed features that recapitulate the immunologic and clinical phenotype of CAPS. They showed systemic inflammation with high levels of serum proinflammatory cytokines, inflammatory infiltrates in various organs, and amyloid deposits in the spleen, liver, and kidneys. Toll-like receptor stimulated macrophages from KI mice secreted high levels of IL-1ß, IL-18, and IL-1α but low amounts of IL-1 receptor antagonist. Treatment of KI mice with PPIs had a clear clinical effect, showing a reduction in inflammatory manifestations, regression of amyloid deposits, and normalization of proinflammatory and anti-inflammatory cytokine production by macrophages. CONCLUSION: Nlrp3 KI mice displayed a CAPS phenotype with many characteristics of autoinflammation, including amyloidosis. The therapeutic effectiveness of PPIs associated with a lack of toxicity indicates that these drugs could represent relevant adjuvants to the anti-IL-1 drugs in patients with CAPS and other IL-1-driven diseases.


Asunto(s)
Amiloidosis , Síndromes Periódicos Asociados a Criopirina , Proteína con Dominio Pirina 3 de la Familia NLR , Inhibidores de la Bomba de Protones/farmacología , Amiloidosis/tratamiento farmacológico , Amiloidosis/genética , Amiloidosis/inmunología , Animales , Síndromes Periódicos Asociados a Criopirina/tratamiento farmacológico , Síndromes Periódicos Asociados a Criopirina/genética , Síndromes Periódicos Asociados a Criopirina/inmunología , Síndromes Periódicos Asociados a Criopirina/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ratones , Ratones Mutantes , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
2.
Eur J Nucl Med Mol Imaging ; 46(5): 1184-1196, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30617965

RESUMEN

PURPOSE: The endoplasmic reticulum (ER) contains hexose-6P-dehydrogenase (H6PD). This enzyme competes with glucose-6P-phosphatase for processing a variety of phosphorylated hexoses including 2DG-6P. The present study aimed to verify whether this ER glucose-processing machinery contributes to brain FDG uptake. METHODS: Effect of the H6PD inhibitor metformin on brain 18F-FDG accumulation was studied, in vivo, by microPET imaging. These data were complemented with the in vitro estimation of the lumped constant (LC). Finally, reticular accumulation of the fluorescent 2DG analogue 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxyglucose (2NBDG) and its response to metformin was studied by confocal microscopy in cultured neurons and astrocytes. RESULTS: Metformin halved brain 18F-FDG accumulation without altering whole body tracer clearance. Ex vivo, this same response faced the doubling of both glucose consumption and lactate release. The consequent fall in LC was not explained by any change in expression or activity of its theoretical determinants (GLUTs, hexokinases, glucose-6P-phosphatase), while it agreed with the drug-induced inhibition of H6PD function. In vitro, 2NBDG accumulation selectively involved the ER lumen and correlated with H6PD activity being higher in neurons than in astrocytes, despite a lower glucose consumption. CONCLUSIONS: The activity of the reticular enzyme H6PD profoundly contributes to brain 18F-FDG uptake. These data challenge the current dogma linking 2DG/FDG uptake to the glycolytic rate and introduce a new model to explain the link between 18-FDG uptake and neuronal activity.


Asunto(s)
Encéfalo/citología , Encéfalo/metabolismo , Retículo Endoplásmico/metabolismo , Fluorodesoxiglucosa F18/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Transporte Biológico/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Deshidrogenasas de Carbohidratos/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Glucólisis/efectos de los fármacos , Metformina/farmacología , Ratones , Ratones Endogámicos BALB C , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidación-Reducción/efectos de los fármacos , Tomografía de Emisión de Positrones
3.
Eur J Immunol ; 47(4): 743-753, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28198545

RESUMEN

The fusion protein L19mTNF (mouse TNF and human antibody fragment L19 directed to fibronectin extra domain B) selectively targets the tumor vasculature, and in combination with melphalan induces a long-lasting T-cell therapeutic response and immune memory in murine models. Increasing evidence suggests that natural killer (NK) cells act to promote effective T-cell-based antitumor responses. We have analyzed the role of NK cells and dendritic cells (DCs) on two different murine tumor models: WEHI-164 fibrosarcoma and C51 colon carcinoma, in which the combined treatment induces high and low rejection rates, respectively. In vivo NK-cell depletion strongly reduced the rejection of WEHI-164 fibrosarcoma and correlated with a decrease in mature DCs, CD4+ , and CD8+ T cells in the tumor-draining LNs and mature DCs and CD4+ T cells in the tumor 40 h after initiation of the therapy. NK-cell depletion also resulted in the impairment of the stimulatory capability of DCs derived from tumor-draining LNs of WEHI-164-treated mice. Moreover, a significant reduction of M2-type infiltrating macrophages was detected in both tumors undergoing therapy. These results suggest that the efficacy of L19mTNF/melphalan therapy is strongly related to the early activation of NK cells and DCs, which are necessary for an effective T-cell response.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Neoplasias Colorrectales/tratamiento farmacológico , Células Dendríticas/inmunología , Quimioterapia Combinada , Fibrosarcoma/tratamiento farmacológico , Células Asesinas Naturales/inmunología , Melfalán/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular , Línea Celular Tumoral , Células Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Células Asesinas Naturales/efectos de los fármacos , Activación de Linfocitos , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Linfocitos T Citotóxicos/efectos de los fármacos , Carga Tumoral/efectos de los fármacos
4.
Carcinogenesis ; 34(3): 620-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23161574

RESUMEN

The chemopreventive and therapeutic efficacy of the cyclooxygenase (COX) inhibitor ibuprofen (IB) and of sulfasalazine (SASP), a drug that targets the antioxidant xc- system, were exploited in the experimental model of 3-methylcholantrene (3-MCA)-induced mouse sarcoma. The chemopreventive treatments gave unsatisfactory results because administration of IB one day after the 3-MCA injection only slightly delayed the tumor development, whereas SASP dispensed under the same conditions resulted in accelerated tumorigenesis. Similarly, the therapeutic treatment with either drug, administrated daily from the tumor detection, decreased the proliferation rate of tumor cells and increased the survival of treated mice only at a low extent. Remarkably, the combined chemopreventive treatment with IB and therapeutic treatment with SASP displayed a better efficacy, with strong delay of sarcoma growth, reduced tumor size and increased survival of treated mice. The two drugs target not only tumor cells but also tumor-associated macrophages that were dramatically decreased in the tumor infiltrate of mice subjected to the combined treatment. The synergistic effects of the association between a broad anti-inflammatory compound, such as IB, and a redox-directed drug, such as SASP, shed new light in the role of inflammation and of the redox response in chemical tumorigenesis and point to the combined chemopreventive plus therapeutic treatment with IB and SASP as a promising novel approach for antitumor therapy.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Anticarcinógenos/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Ibuprofeno/farmacología , Sarcoma Experimental/prevención & control , Sulfasalazina/farmacología , Adulto , Anciano , Sistema de Transporte de Aminoácidos y+/genética , Animales , Anticarcinógenos/uso terapéutico , Antioxidantes/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/uso terapéutico , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Ibuprofeno/uso terapéutico , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Metilcolantreno , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Sarcoma Experimental/inducido químicamente , Sarcoma Experimental/inmunología , Sarcoma Experimental/metabolismo , Sulfasalazina/uso terapéutico , Adulto Joven
5.
Cancer Med ; 11(1): 183-193, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34796694

RESUMEN

Triple negative breast cancers (TNBCs) are very aggressive and have a poor prognosis due to lack of efficacious therapies. The only effective treatment is chemotherapy that however is frequently hindered by the occurrence of drug resistance. We approached this problem in vitro and in vivo on a triple negative and a hormone sensitive breast cancer cell lines: 4T1 and TS/A. A main defense mechanism of tumors is the extrusion of intracellular protons derived from the metabolic shift to glycolysis, and necessary to maintain an intracellular pH compatible with life. The resulting acidic extracellular milieu bursts the malignant behavior of tumors and impairs chemotherapy. Therefore, we investigated the efficacy of combined therapies that associate cisplatin (Cis) with proton exchanger inhibitors, such as esomeprazole (ESO) and 5-(N-ethyl-N-isopropyl)amiloride (EIPA). Our results demonstrate that in the 4T1 triple negative model the combined therapy Cis plus EIPA is significantly more effective than the other treatments. Instead, in the TS/A tumor the best therapeutic result is obtained with ESO alone. Remarkably, in both 4T1 and TS/A tumors these treatments correlate with increase of CD8+  T lymphocytes and dendritic cells, and a dramatic reduction of M2 macrophages and other suppressor myeloid cells (MDSC) in the tumor infiltrates.


Asunto(s)
Amilorida/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/uso terapéutico , Esomeprazol/uso terapéutico , Inhibidores de la Bomba de Protones/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Amilorida/uso terapéutico , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones Endogámicos BALB C , Intercambiador 1 de Sodio-Hidrógeno/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Macrófagos Asociados a Tumores/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
6.
BMC Biotechnol ; 11: 104, 2011 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-22074550

RESUMEN

BACKGROUND: Ligand-targeted approaches have proven successful in improving the therapeutic index of a number of drugs. We hypothesized that the specific targeting of TNF-alpha antagonists to inflamed tissues could increase drug efficacy and reduce side effects. RESULTS: Using uteroglobin (UG), a potent anti-inflammatory protein, as a scaffold, we prepared a bispecific tetravalent molecule consisting of the extracellular ligand-binding portion of the human TNF-alpha receptor P75 (TNFRII) and the scFv L19. L19 binds to the ED-B containing fibronectin isoform (B-FN), which is expressed only during angiogenesis processes and during tissue remodeling. B-FN has also been demonstrated in the pannus in rheumatoid arthritis. L19-UG-TNFRII is a stable, soluble homodimeric protein that maintains the activities of both moieties: the immuno-reactivity of L19 and the capability of TNFRII to inhibit TNF-alpha. In vivo bio-distribution studies demonstrated that the molecule selectively accumulated on B-FN containing tissues, showing a very fast clearance from the blood but a very long residence time on B-FN containing tissues. Despite the very fast clearance from the blood, this fusion protein was able to significantly improve the severe symptomatology of arthritis in collagen antibody-induced arthritis (CAIA) mouse model. CONCLUSIONS: The recombinant protein described here, able to selectively deliver the TNF-alpha antagonist TNFRII to inflamed tissues, could yield important contributions for the therapy of degenerative inflammatory diseases.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Fibronectinas/metabolismo , Articulaciones/irrigación sanguínea , Neovascularización Patológica/tratamiento farmacológico , Receptores Tipo II del Factor de Necrosis Tumoral/farmacocinética , Proteínas Recombinantes de Fusión/farmacocinética , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Células CHO , Cricetinae , Dimerización , Fibronectinas/inmunología , Humanos , Radioisótopos de Yodo/análisis , Articulaciones/efectos de los fármacos , Articulaciones/inmunología , Articulaciones/patología , Ratones , Neovascularización Patológica/inmunología , Plásmidos , Unión Proteica , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Teratocarcinoma , Transfección , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Uteroglobina/química , Uteroglobina/genética
7.
J Biol Chem ; 284(39): 26646-54, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19632988

RESUMEN

We report a novel strategy to engineer and express stable and soluble human recombinant polyvalent/polyspecific fusion proteins. The procedure is based on the use of a central skeleton of uteroglobin, a small and very soluble covalently linked homodimeric protein that is very resistant to proteolytic enzymes and to pH variations. Using a human recombinant antibody (scFv) specific for the angiogenesis marker domain B of fibronectin, interleukin 2, and an scFv able to neutralize tumor necrosis factor-alpha, we expressed various biologically active uteroglobin fusion proteins. The results demonstrate the possibility to generate monospecific divalent and tetravalent antibodies, immunocytokines, and dual specificity tetravalent antibodies. Furthermore, compared with similar fusion proteins in which uteroglobin was not used, the use of uteroglobin improved properties of solubility and stability. Indeed, in the reported cases it was possible to vacuum dry and reconstitute the proteins without any aggregation or loss in protein and biological activity.


Asunto(s)
Interleucina-2/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Uteroglobina/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Humanos , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Ratones , Ratones Endogámicos , Modelos Moleculares , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Oxidación-Reducción , Plásmidos/genética , Multimerización de Proteína , Señales de Clasificación de Proteína/genética , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Uteroglobina/química , Uteroglobina/genética
8.
Int J Cancer ; 127(1): 101-10, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19877124

RESUMEN

L19-IL2 and L19TNFalpha are fusion proteins composed of L19(scFv), specific for the angiogenesis-associated ED-B containing fibronectin isoform and IL-2 or TNFalpha. Because of the tumor targeting properties of L19, IL-2 and TNFalpha concentrate at therapeutic doses at the tumor vascular level. To evaluate the therapeutic effects of L19-IL2 and L19mTNFalpha in neuroblastoma (NB)-bearing mice, A/J mice bearing Neuro2A or NIE115 NB were systemically treated with L19-IL2 and L19mTNFalpha, alone or in combination protocols. Seventy percent of Neuro2A- and 30% of NIE115-bearing mice were cured by the combined treatment with L19-IL2 and L19mTNFalpha, and further rejected a homologous tumor challenge, indicating specific antitumor immune memory. The immunological bases of tumor cure and rejection were studied. A highly efficient priming of CD4(+) T helper cells and CD8(+) CTL effectors was generated, paralleled by massive infiltration in the tumor tissue of CD4(+) and CD8(+) T cells at day 16 after tumor cell implantation, when, after therapy, tumor volume was drastically reduced and tumor necrosis reached about 80%. The curative treatment resulted in a long-lasting antitumor immune memory, accompanied by a mixed Th1/Th2 type of response. Concluding, L19-IL2 and L19mTNFalpha efficiently cooperate in determining a high percentage of NB cure that, in our experimental models, is strongly associated to the generation of adaptive immunity involving CD4(+) and CD8(+) T cells.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Interleucina-2/administración & dosificación , Neuroblastoma/terapia , Factor de Necrosis Tumoral alfa/administración & dosificación , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Ratones , Neuroblastoma/inmunología
9.
Curr Med Chem ; 27(25): 4233-4248, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30182839

RESUMEN

Tumor cells and tumor-associated stromal cells such as immune, endothelial and mesenchimal cells create a Tumor Microenvironment (TME) which allows tumor cell promotion, growth and dissemination while dampening the anti-tumor immune response. Efficient anti-tumor interventions have to keep into consideration the complexity of the TME and take advantage of immunotherapy and chemotherapy combined approaches. Thus, the aim of tumor therapy is to directly hit tumor cells and reverse endothelial and immune cell anergy. Selective targeting of tumor vasculature using TNFα-associated peptides or antibody fragments in association with chemotherapeutic agents, has been shown to exert a potent stimulatory effect on endothelial cells as well as on innate and adaptive immune responses. These drug combinations reducing the dose of single agents employed have led to minimize the associated side effects. In this review, we will analyze different TNFα-mediated tumor vesseltargeted therapies in both humans and tumor mouse models, with emphasis on the role played by the cross-talk between natural killer and dendritic cells and on the ability of TNFα to trigger tumor vessel activation and normalization. The improvement of the TNFα-based therapy with anti-angiogenic immunomodulatory drugs that may convert the TME from immunosuppressive to immunostimulant, will be discussed as well.


Asunto(s)
Neoplasias , Animales , Células Endoteliales , Humanos , Inmunoterapia , Microambiente Tumoral , Factor de Necrosis Tumoral alfa
10.
Cancers (Basel) ; 12(3)2020 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-32197460

RESUMEN

Human RNASET2 acts as a powerful oncosuppressor protein in in vivo xenograft-based murine models of human cancer. Secretion of RNASET2 in the tumor microenvironment seems involved in tumor suppression, following recruitment of M1-polarized macrophages. Here, we report a murine Rnaset2-based syngeneic in vivo assay. BALB/c mice were injected with parental, empty vector-transfected or murine Rnaset2-overexpressing mouse C51 or TS/A syngeneic cells and tumor growth pattern and immune cells distribution in tumor mass were investigated. Compared to control cells, mouse Rnaset2-expressing C51 cells showed strong delayed tumor growth. CD86+ M1 macrophages were massively recruited in Rnaset2-expressing C51-derived tumors, with concomitant inhibition of MDSCs and CD206+ M2 macrophages recruitment. At later times, a relevant expansion of intra-tumor CD8+ T cells was also observed. After re-challenge with C51 parental cells, most mice previously injected with Rnaset2-expressing C51 cells still rejected C51 tumor cells, suggesting a Rnaset2-mediated T cell adaptive immune memory response. These results point at T2 RNases as evolutionary conserved oncosuppressors endowed with the ability to inhibit cancer growth in vivo through rebalance of intra-tumor M1/M2 macrophage ratio and concomitant recruitment of adaptive anti-tumor CD8+ T cells.

11.
Transl Oncol ; 13(5): 100752, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32302773

RESUMEN

This study aims to verify in experimental models of hyperglycemia induced by streptozotocin (STZ-DM) to what degree the high competition between unlabeled glucose and metformin (MET) treatment might affect the accuracy of cancer FDG imaging. The study included 36 "control" and 36 "STZ-DM" Balb/c mice, undergoing intraperitoneal injection of saline or streptozotocin, respectively. Two-weeks later, mice were subcutaneously implanted with breast (4 T1) or colon (CT26) cancer cells and subdivided in three subgroups for treatment with water or with MET at 10 or 750 mg/Kg/day. Two weeks after, mice were submitted to micro-PET imaging. Enzymatic pathways and response to oxidative stress were evaluated in harvested tumors. Finally, competition by glucose, 2-deoxyglucose (2DG) and the fluorescent analog 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxyglucose (2-NBDG) on FDG uptake was studied in 4 T1 and CT26 cultured cells. STZ-DM slightly decreased cancer volume and FDG uptake rate (MRF). More importantly, it also abolished MET capability to decelerate lesion growth and MRF. This metabolic reprogramming closely agreed with the activity of hexose-6-phosphate dehydrogenase within the endoplasmic reticulum. Finally, co-incubation with 2DG virtually abolished FDG and 2-NBDG uptake within the endoplasmic reticulum in cultured cells. These data challenge the current dogma linking FDG uptake to glycolytic flux and introduce a new model to explain the relation between glucose analogue uptake and hexoses reticular metabolism. This selective fate of FDG contributes to the preserved sensitivity of PET imaging in oncology even in chronic moderate hyperglycemic conditions.

12.
Int J Cancer ; 125(4): 751-8, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19479996

RESUMEN

The angiogenesis-associated extra-domain B (EDB) of fibronectin (FN) is a complete type III repeat of 91 amino acids. Its expression is modulated by the alternative splicing pattern of the FN pre-mRNA. FN containing the EDB (B-FN) is undetectable in tissues of healthy adults, with rare exceptions such as the female reproductive system where tissue remodeling and angiogenesis are recurrent physiological processes. On the contrary, B-FN is expressed at high levels in neoplastic tissues and during angiogenesis; consequently, it is considered an excellent marker of angiogenesis. Here, we report on a novel FN cryptic sequence, localized on the FN type III repeat 8 (immediately downstream of the EDB) that is unmasked by the insertion of the EDB. This sequence is specifically recognized by the high-affinity monoclonal antibody, C6, that selectively recognizes B-FN by means of ELISA, immunohistochemical and Western blot assays. The variable regions of C6 were cloned and a divalent covalently linked mini-antibody was generated. Biodistribution studies using the radioiodinated C6 mini-antibody on tumor-bearing mice demonstrated an efficient tumor targeting. This antibody represents a new tool for the study of the potential biological functions of hindered sequences that the inclusion of the EDB renders accessible, and likewise makes its epitope an additional angiogenesis target.


Asunto(s)
Fibronectinas/metabolismo , Neoplasias/metabolismo , Animales , Western Blotting , Células CHO , Cricetinae , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Fibronectinas/química , Halogenación , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones SCID , Neoplasias Experimentales/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Uteroglobina/inmunología , Uteroglobina/metabolismo
13.
Cancers (Basel) ; 11(9)2019 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-31443604

RESUMEN

Epithelial ovarian cancer (EOC) is the fifth most common cancer affecting the female population. At present, different targeted treatment approaches may improve currently employed therapies leading either to the delay of tumor recurrence or to disease stabilization. In this study we show that syndecan-1 (SDC1) and tumor angiogenic-associated B-fibronectin isoform (B-FN) are involved in EOC progression and we describe the prominent role of SDC1 in the vasculogenic mimicry (VM) process. We also investigate a possible employment of L19-IL2, an immunocytokine specific for B-FN, and anti-SDC1 46F2SIP (small immuno protein) antibody in combination therapy in a human ovarian carcinoma model. A tumor growth reduction of 78% was obtained in the 46F2SIP/L19-IL2-treated group compared to the control group. We observed that combined treatment was effective in modulation of epithelial-mesenchymal transition (EMT) markers, loss of stemness properties of tumor cells, and in alleviating hypoxia. These effects correlated with reduction of VM structures in tumors from treated mice. Interestingly, the improved pericyte coverage in vascular structures suggested that combined therapy could be efficacious in induction of vessel normalization. These data could pave the way for a possible use of L19-IL2 combined with 46F2SIP antibody as a novel therapeutic strategy in EOC.

14.
Front Immunol ; 9: 2905, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619269

RESUMEN

Antibody-cytokine fusion proteins (immunocytokine) exert a potent anti-cancer effect; indeed, they target the immunosuppressive tumor microenvironment (TME) due to a specific anti-tumor antibody linked to immune activating cytokines. Once bound to the target tumor, the interleukin-2 (IL-2) immunocytokines composed of either full antibody or single chain Fv conjugated to IL-2 can promote the in situ recruitment and activation of natural killer (NK) cells and cytotoxic CD8+ T lymphocytes (CTL). This recruitment induces a TME switch toward a classical T helper 1 (Th1) anti-tumor immune response, supported by the cross-talk between NK and dendritic cells (DC). Furthermore, some IL-2 immunocytokines have been largely shown to trigger tumor cell killing by antibody dependent cellular cytotoxicity (ADCC), through Fcγ receptors engagement. The modulation of the TME can be also achieved with immunocytokines conjugated with a mutated form of IL-2 that impairs regulatory T (Treg) cell proliferation and activity. Preclinical animal models and more recently phase I/II clinical trials have shown that IL-2 immunocytokines can avoid the severe toxicities of the systemic administration of high doses of soluble IL-2 maintaining the potent anti-tumor effect of this cytokine. Also, very promising results have been reported using IL-2 immunocytokines delivered in combination with other immunocytokines, chemo-, radio-, anti-angiogenic therapies, and blockade of immune checkpoints. Here, we summarize and discuss the most relevant reported studies with a focus on: (a) the effects of IL-2 immunocytokines on innate and adaptive anti-tumor immune cell responses as well as immunosuppressive Treg cells and (b) the approaches to circumvent IL-2-mediated severe toxic side effects.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inmunoterapia/métodos , Neoplasias/terapia , Proteínas Recombinantes de Fusión/farmacología , Escape del Tumor/efectos de los fármacos , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioradioterapia/métodos , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Humanos , Interleucina-2/inmunología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Resultado del Tratamiento , Escape del Tumor/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
15.
Sci Rep ; 8(1): 11075, 2018 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30038406

RESUMEN

meso-(p-acetamidophenyl)-calix[4]pyrrole 3 was found to exhibit remarkable cytotoxicity towards A549 cancer cells. A comparative study including the isomer of 3 meso-(m-acetamidophenyl)-calix[4]pyrrole 5, as well as molecules containing 'fragments' of these structures, demonstrated that both the calix[4]pyrrole and the acetamidophenyl units are essential for high cytotoxicity. Although calix[4]pyrroles and other anion-complexing ionophores have recently been reported to induce apoptosis by perturbing cellular chloride concentrations, in our study an alternative mechanism has emerged, as proven by the isolation of covalent DNA adducts revealed by the 32P postlabelling technique. Preliminary pharmacokinetic studies indicate that 3 is able to cross the Blood-Brain-Barrier, therefore being a potential drug that could kill primary and brain metastatic cancer cells simultaneously.


Asunto(s)
Antineoplásicos/farmacología , Calixarenos/farmacología , Aductos de ADN/metabolismo , Mutágenos/toxicidad , Porfirinas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Calixarenos/química , Calixarenos/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Porfirinas/química , Porfirinas/farmacocinética , Distribución Tisular/efectos de los fármacos
16.
Clin Cancer Res ; 12(8): 2575-82, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16638868

RESUMEN

PURPOSE: We sought to demonstrate that a single systemic administration of L19mTNFalpha (a fusion protein constituted by the scFv L19 specific for the oncofetal ED-B domain of fibronectin and tumor necrosis factor alpha, TNFalpha) in combination with melphalan induced complete and long-lasting tumor eradication in tumor-bearing mice and triggered the generation of a specific T cell-based immune response that protects the animals from a second tumor challenge, as well as from challenges with syngeneic tumor cells of different histologic origin. EXPERIMENTAL DESIGN AND RESULTS: Treatment with L19mTNFalpha, in combination with melphalan, induced complete tumor regression in 83% of BALB/c mice with WEHI-164 fibrosarcoma and 33% of animals with C51 colon carcinoma. All cured mice rejected challenges with the same tumor cells and, in a very high percentage of animals, also rejected challenges with syngeneic tumor cells of different histologic origin. In adoptive immunity transfer experiments, the splenocytes from tumor-cured mice protected naive mice both from C51 colon carcinoma and from WEHI-164 fibrosarcoma. Similar results were also obtained in adoptive immunity transfer experiments using severely immunodepressed mice. Experiments using depleted splenocytes showed that T cells play a major role in tumor rejection. CONCLUSIONS: The results show that the selective targeting of mTNFalpha to the tumor enhances its immunostimulatory properties to the point of generating a therapeutic immune response against different histologically unrelated syngeneic tumors. These findings predicate treatment approaches for cancer patients based on the targeted delivery of TNFalpha to the tumor vasculature.


Asunto(s)
Inmunidad Celular/efectos de los fármacos , Neoplasias Experimentales/inmunología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citotoxicidad Inmunológica/inmunología , Relación Dosis-Respuesta a Droga , Fibronectinas/genética , Fibronectinas/inmunología , Inmunidad Celular/inmunología , Fragmentos de Inmunoglobulinas/genética , Inmunoterapia Adoptiva/métodos , Melfalán/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Neoplasias Experimentales/prevención & control , Neoplasias Experimentales/terapia , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Bazo/citología , Bazo/inmunología , Bazo/trasplante , Análisis de Supervivencia , Linfocitos T/citología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología
17.
Oncotarget ; 8(40): 67482-67496, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978047

RESUMEN

Neoplastic cells live in a stressful context and survive thanks to their ability to overcome stress. Thus, tumor cell responses to stress are potential therapeutic targets. We selected two such responses in melanoma and sarcoma cells: the xc- antioxidant system, that opposes oxidative stress, and surface v-ATPases that counteract the low pHi by extruding protons, and targeted them with the xc- blocker sulfasalazine and the proton pump inhibitor esomeprazole. Sulfasalazine inhibited the cystine/cysteine redox cycle and esomeprazole decreased pHi while increasing pHe in tumor cell lines. Although the single treatment with either drug slightly inhibited cell proliferation and motility, the association of sulfasalazine and esomeprazole powerfully decreased sarcoma and melanoma growth and migration. In the 3-methylcholanthrene (3-MCA)-induced sarcoma model, the combined therapy strongly reduced the tumor burden and increased the survival time: notably, 22 % of double-treated mice recovered and survived off therapy. Tumor-associated macrophages (TAM) displaying M2 markers, that abundantly infiltrate sarcoma and melanoma, overexpress xc- and membrane v-ATPases and were drastically decreased in tumors from mice undergone the combined therapy. Thus, the double targeting of tumor cells and macrophages by sulfasalazine and esomeprazole has a double therapeutic effect, as decreasing TAM infiltration deprives tumor cells of a crucial allied. Sulfasalazine and esomeprazole may therefore display unexpected therapeutic values, especially in case of hard-to-treat cancers.

18.
Oncotarget ; 6(35): 37426-42, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26460958

RESUMEN

Anti-angiogenic therapy of solid tumors has until now failed to produce the long lasting clinical benefits desired, possibly due to the complexity of the neoangiogenic process. Indeed, a prominent role is played by "vasculogenic" or "vascular" mimicry (VM), a phenomenon in which aggressive cancer cells form an alternative microvascular circulation, independently of endothelial cell angiogenesis. In this study we observed, in melanoma patient cell lines having vasculogenic/stem-cell like phenotype and in melanoma tumors, the syndecan-1 co-expression with VM markers, such as CD144 and VEGFR-2. We show that melanoma cells lose their ability to form tubule-like structures in vitro after blocking syndecan-1 activity by the specific human recombinant antibody, OC-46F2. Moreover, in a human melanoma xenograft model, the combined therapy using OC-46F2 and L19-IL2, an immunocytokine specific for the tumor angiogenic-associated B-fibronectin isoform(B-FN), led to a complete inhibition of tumor growth until day 90 from tumor implantation in 71% of treated mice, with statistically significant differences compared to groups treated with OC-46F2 or L19-IL2 as monotherapy. Furthermore, in the tumors recovered from mice treated with OC-46F2 either as monotherapy or in combination with L19-IL2, we observed a dramatic decrease of vascular density and loss of VM structures. These findings indicate for the first time a role of syndecan-1 in melanoma VM and that targeting syndecan-1, together with B-FN, could be promising in improving the treatment of metastatic melanoma.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Imitación Molecular , Neovascularización Patológica , Proteínas Recombinantes de Fusión/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Sindecano-1/antagonistas & inhibidores , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Masculino , Melanoma/irrigación sanguínea , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/secundario , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neovascularización Fisiológica/efectos de los fármacos , Fenotipo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Sindecano-1/inmunología , Sindecano-1/metabolismo , Factores de Tiempo , Carga Tumoral , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Antioxid Redox Signal ; 20(7): 1086-97, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23373831

RESUMEN

SIGNIFICANCE: Increasing evidence indicates that cancer development and progression are promoted by the joint action of redox distress and inflammation, supporting the potential impact of therapies aimed at restoring the redox homeostasis and fighting inflammation. RECENT ADVANCES: Most of the literature of the last 40 years converges to the view that continuous oxidative stress and chronic inflammation sustain each other, leads to transformation of a normal cell to a neoplastic cell, and promotes tumor progression. Some recent findings, however, support an alternative model whereby the increased production of reactive oxygen species (ROS) is an attempt to defend more than a pathogenetic factor in cancer. Rather, tumor development and progression may be promoted by an excess of antioxidants, induced in both transformed cells and recruited inflammatory cells as an adaptive response to ROS. CRITICAL ISSUES: Although the link among redox stress, chronic inflammation, and cancer is widely recognized, the underlying mechanisms are far to be understood. The redox unbalance of the microenvironment is likely to modulate the bioactivity of damage-associated molecular pattern molecules such as HMGB1, which are released by stressed tissues and play pleiotropic functions on tumor and inflammatory cells, but how this occur, and the relevant consequences, are still unclear. FUTURE DIRECTIONS: In vivo measurement of cell redox status is an important challenge for future investigations. The improvement of the methodologies for ROS and antioxidant detection will allow a better understanding of the redox-related events in the tumor microenvironment with tremendous application potential in the development of rational combination therapies for cancer treatment.


Asunto(s)
Carcinogénesis/patología , Inflamación/patología , Neoplasias/patología , Transducción de Señal/fisiología , Animales , Antioxidantes/metabolismo , Progresión de la Enfermedad , Humanos , Oxidación-Reducción , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral/fisiología
20.
Eur J Cancer ; 49(8): 2022-33, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23352437

RESUMEN

INTRODUCTION: Syndecan-1 is a cell membrane protein that, after its shedding by heparanase enzymes, is accumulated in the extracellular matrix of some tumours, e.g. myeloma and lung carcinoma, where it modulates several key processes of tumourigenesis such as cancer cell proliferation and apoptosis, angiogenesis and metastasis. Few studies have focused on syndecan-1 in malignant melanoma, a tumour for which new therapeutic targets are desperately needed. We aimed to investigate the role of syndecan-1 in melanoma and to evaluate the potential therapeutic efficacy of a novel fully human anti-syndecan-1 recombinant antibody in this deadly disease. METHODS: The OC-46F2 recombinant antibody was generated by selecting a human antibody phage display library on human melanoma cells and by its expression in mammalian cells. The specific antigen recognised by the antibody was identified by mass spectrometry. Murine models of human melanoma and ovarian carcinoma were used in the pre-clinical in vivo experiments. RESULTS: The fully human antibody OC-46F2, specific for the extracellular domain of syndecan-1, inhibited vascular maturation and tumour growth in an experimental human melanoma model. The therapeutic efficacy of this antibody was also demonstrated in an experimental ovarian carcinoma model. A co-distribution of syndecan-1 with vascular endothelial growth factor receptor 2 (VEGFR2) observed in the intratumour melanoma microenvironment was absent in the tumours from mice treated with OC-46F2 scFv. CONCLUSION: These findings highlight the role of syndecan-1 as a potential therapeutic target in melanoma and ovarian carcinoma and provide a new tool able to block vessel maturation, one of the mechanisms that underpin the angiogenic process essential for solid tumour growth.


Asunto(s)
Melanoma/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Anticuerpos de Cadena Única/farmacología , Sindecano-1/metabolismo , Carga Tumoral/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Células CHO , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Cricetinae , Cricetulus , Femenino , Células HEK293 , Humanos , Melanoma/irrigación sanguínea , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Datos de Secuencia Molecular , Neovascularización Patológica/patología , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Biblioteca de Péptidos , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Sindecano-1/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA