Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 303(12): L1046-56, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23043074

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease without effective therapeutics. Periostin has been reported to be elevated in IPF patients relative to controls, but its sources and mechanisms of action remain unclear. We confirm excess periostin in lungs of IPF patients and show that IPF fibroblasts produce periostin. Blood was obtained from 54 IPF patients (all but 1 with 48 wk of follow-up). We show that periostin levels predict clinical progression at 48 wk (hazard ratio = 1.47, 95% confidence interval = 1.03-2.10, P < 0.05). Monocytes and fibrocytes are sources of periostin in circulation in IPF patients. Previous studies suggest that periostin may regulate the inflammatory phase of bleomycin-induced lung injury, but periostin effects during the fibroproliferative phase of the disease are unknown. Wild-type and periostin-deficient (periostin(-/-)) mice were anesthetized and challenged with bleomycin. Wild-type mice were injected with bleomycin and then treated with OC-20 Ab (which blocks periostin and integrin interactions) or control Ab during the fibroproliferative phase of disease, and fibrosis and survival were assessed. Periostin expression was upregulated quickly after treatment with bleomycin and remained elevated. Periostin(-/-) mice were protected from bleomycin-induced fibrosis. Instillation of OC-20 during the fibroproliferative phase improved survival and limited collagen deposition. Chimeric mouse studies suggest that hematopoietic and structural sources of periostin contribute to lung fibrogenesis. Periostin was upregulated by transforming growth factor-ß in lung mesenchymal cells, and periostin promoted extracellular matrix deposition, mesenchymal cell proliferation, and wound closure. Thus periostin plays a vital role in late stages of pulmonary fibrosis and is a potential biomarker for disease progression and a target for therapeutic intervention.


Asunto(s)
Moléculas de Adhesión Celular/sangre , Fibrosis Pulmonar Idiopática/metabolismo , Anciano , Animales , Anticuerpos Neutralizantes/farmacología , Biomarcadores , Moléculas de Adhesión Celular/biosíntesis , Proliferación Celular , Colágeno/metabolismo , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Monocitos/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Cicatrización de Heridas
2.
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
3.
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
4.
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
5.
J Exp Med ; 198(4): 557-67, 2003 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-12913096

RESUMEN

Human natural killer (NK) cells express a series of activating receptors and coreceptors that are involved in recognition and killing of target cells. In this study, in an attempt to identify the cellular ligands for such triggering surface molecules, mice were immunized with NK-susceptible target cells. On the basis of a functional screening, four mAbs were selected that induced a partial down-regulation of the NK-mediated cytotoxicity against the immunizing target cells. As revealed by biochemical analysis, three of such mAbs recognized molecules of approximately 70 kD. The other mAb reacted with two distinct molecules of approximately 65 and 60 kD, respectively. Protein purification followed by tryptic digestion and mass spectra analysis, allowed the identification of the 70 kD and the 65/60 kD molecules as PVR (CD155) and Nectin-2 delta/alpha (CD112), respectively. PVR-Fc and Nectin-2-Fc soluble hybrid molecules brightly stained COS-7 cells transfected with the DNAM-1 (CD226) construct, thus providing direct evidence that both PVR and Nectin-2 represent specific ligands for the DNAM-1 triggering receptor. Finally, the surface expression of PVR or Nectin-2 in cell transfectants resulted in DNAM-1-dependent enhancement of NK-mediated lysis of these target cells. This lysis was inhibited or even virtually abrogated upon mAb-mediated masking of DNAM-1 (on NK cells) or PVR or Nectin-2 ligands (on cell transfectants).


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos de Diferenciación de Linfocitos T , Moléculas de Adhesión Celular/metabolismo , Proteínas de la Membrana , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos CD/metabolismo , Moléculas de Adhesión Celular/química , Línea Celular , Pruebas Inmunológicas de Citotoxicidad , Humanos , Células Asesinas Naturales/metabolismo , Ligandos , Ratones , Ratones Endogámicos BALB C , Nectinas , Péptidos/genética , Péptidos/metabolismo , Receptores Virales/química
6.
Exp Cell Res ; 315(2): 226-39, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19000672

RESUMEN

Tumor progression is characterized by definite changes in the protein composition of the nuclear matrix (NM). The interactions of chromatin with the NM occur via specific DNA sequences called MARs (matrix attachment regions). In the present study, we applied a proteomic approach along with a Southwestern assay to detect both differentially expressed and MAR-binding NM proteins, in persistent hepatocyte nodules (PHN) in respect with normal hepatocytes (NH). In PHN, the NM undergoes changes both in morphology and in protein composition. We detected over 500 protein spots in each two dimensional map and 44 spots were identified. Twenty-three proteins were differentially expressed; among these, 15 spots were under-expressed and 8 spots were over-expressed in PHN compared to NH. These changes were synchronous with several modifications in both NM morphology and the ability of NM proteins to bind nuclear RNA and/or DNA containing MARs sequences. In PHN, we observed a general decrease in the expression of the basic proteins that bound nuclear RNA and the over-expression of two species of Mw 135 kDa and 81 kDa and pI 6.7-7.0 and 6.2-7.4, respectively, which exclusively bind to MARs. These results suggest that the deregulated expression of these species might be related to large-scale chromatin reorganization observed in the process of carcinogenesis by modulating the interaction between MARs and the scaffold structure.


Asunto(s)
Neoplasias Hepáticas/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Matriz Nuclear/metabolismo , Proteómica/métodos , Animales , Western Blotting , Proteínas de Ciclo Celular , Electroforesis en Gel Bidimensional , Proteínas de Choque Térmico/análisis , Proteínas de Choque Térmico/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/análisis , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Queratinas Tipo II/análisis , Queratinas Tipo II/metabolismo , Laminas/análisis , Laminas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/ultraestructura , Masculino , Proteínas de Unión a la Región de Fijación a la Matriz/análisis , Microscopía Electrónica , Matriz Nuclear/química , Matriz Nuclear/ultraestructura , Proteínas Asociadas a Matriz Nuclear/análisis , Proteínas Nucleares/análisis , Proteínas Nucleares/metabolismo , Unión Proteica , ARN Nuclear/metabolismo , Proteínas de Unión al ARN/análisis , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Endogámicas F344 , Ribonucleósidos/química , Ribonucleósidos/metabolismo , Espectrometría de Masas en Tándem/métodos , Factores de Tiempo , Vanadatos/química , Vanadatos/metabolismo
7.
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
8.
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
9.
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.

10.
Cell Oncol ; 30(1): 13-26, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18219107

RESUMEN

INTRODUCTION: Although several molecular markers for bladder cancer have been identified, at present little information on prognostic biomarkers is available in the literature. Prognostication of this tumor is largely based on clinicopathological characteristics. Our aim was to identify nuclear matrix (NM) proteins that might serve to better characterize the phenotype of the invasive bladder cancer and to investigate their diagnostic and prognostic roles. METHODS: NM proteins expressed in normal (n=3) or non-tumoral (n=9) tissue specimens and muscle-invasive bladder cancer (n=21) specimens were analyzed by two dimensional (2D) gel electrophoresis. PDQuest image analysis software was used to generate a comparative NM proteome analysis. Selected spots were characterized by liquid chromatography coupled to tandem mass spectrometry and Western blot. RESULTS: We detected over 800 protein spots in each 2D map and 43 spots were identified. 30 proteins were differentially expressed by bladder tumor cells; among these, 19 proteins were detected in bladder tumoral tissues but not in normal and non-tumoral tissues and seven proteins correlated with tumor stage. One protein (p54nrb) was strongly correlated with vascular invasions and appeared to be also significantly (P<0.0001) associated with a decreased probability of survival. CONCLUSION: Important alterations in NM proteins occur in muscle-invasive bladder cancer. The differentially expressed proteins include biomarkers potentially useful for disease diagnosis, progression and prognosis. Our findings beyond improving the understanding of the biology of bladder cancer, could help to stratify patients into different prognostic subgroups and to select those who might be better candidate to multimodal therapeutic approaches.


Asunto(s)
Proteínas Asociadas a Matriz Nuclear/análisis , Proteoma/análisis , Neoplasias de la Vejiga Urinaria/química , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Western Blotting , Electroforesis en Gel Bidimensional , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/análisis , Pronóstico , Proteómica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/patología
11.
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
12.
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
13.
Mol Immunol ; 42(4): 463-9, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15607800

RESUMEN

The capability of NK lymphocytes to kill tumor cells depends on different receptors/ligands interactions. In order to identify the cellular ligands recognized by "orphan" triggering receptors, mice were immunized with NK susceptible target cells. mAbs were selected that inhibited NK cytotoxicity and recognized two different molecules of 70 and 60-65 kDa. Tryptic digestion and mass spectra analysis of purified proteins identified these molecules as PVR and Nectin-2, respectively. PVR-Fc and Nectin-2-Fc chimeric molecules stained COS-7 cells expressing the DNAM-1 activating receptor and conversely, PVR and Nectin-2 CHO-K cell transfectants were stained by DNAM-1-Fc. Thus, both PVR and Nectin-2 represent specific ligands for DNAM-1. Importantly, the specific interaction between DNAM-1 (in NK cells) and PVR or Nectin-2 (in target cells) enhanced the NK-mediated lysis of tumor cells that was downregulated by mAb-mediated masking of the receptor or its ligands.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/metabolismo , Neoplasias/inmunología , Receptores Virales/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Células COS , Moléculas de Adhesión Celular/aislamiento & purificación , Chlorocebus aethiops , Citotoxicidad Inmunológica , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Ligandos , Proteínas de la Membrana/aislamiento & purificación , Ratones , Nectinas , Mapeo Peptídico , Receptores Virales/aislamiento & purificación , Proteínas Recombinantes de Fusión/inmunología
14.
Anticancer Res ; 25(6B): 3999-4004, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16309191

RESUMEN

Different lines of evidence suggest that the nuclear matrix (NM), the protein scaffold of the nucleus, represents a functional unit playing a pivotal role in the spatial and temporal coordination of the events of gene activation. Any change in the gene expression pattern, which occurs during carcinogenesis, may partially depend on an impairment of the regulatory functions of the NM. Therefore, increasing interest has been addressed to the study of NM modifications associated with malignant transformations and to potential clinical applications. Here, recent results on the NM changes in prostate cancer are discussed. Tumor cells are characterized by a more complex NM protein pattern compared to normal tissue: the development of poorly-differentiated tumors is characterized by the expression of proteins that are not present in hyperplastic tissues or in more differentiated tumors. In addition, a few newly-expressed proteins are significantly correlated with the risk of biochemical progression. The potential application of these proteins at the diagnostic and prognostic levels calls for further studies.


Asunto(s)
Proteínas Asociadas a Matriz Nuclear/biosíntesis , Neoplasias de la Próstata/metabolismo , Animales , Humanos , Masculino , Pronóstico , Neoplasias de la Próstata/diagnóstico
15.
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
16.
Neuro Oncol ; 17(3): 372-82, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25140038

RESUMEN

BACKGROUND: Periostin is a secreted matricellular protein critical for epithelial-mesenchymal transition and carcinoma metastasis. In glioblastoma, it is highly upregulated compared with normal brain, and existing reports indicate potential prognostic and functional importance in glioma. However, the clinical implications of periostin expression and function related to its therapeutic potential have not been fully explored. METHODS: Periostin expression levels and patterns were examined in human glioma cells and tissues by quantitative real-time PCR and immunohistochemistry and correlated with glioma grade, type, recurrence, and survival. Functional assays determined the impact of altering periostin expression and function on cell invasion, migration, adhesion, and glioma stem cell activity and tumorigenicity. The prognostic and functional relevance of periostin and its associated genes were analyzed using the TCGA and REMBRANDT databases and paired recurrent glioma samples. RESULTS: Periostin expression levels correlated directly with tumor grade and recurrence, and inversely with survival, in all grades of adult human glioma. Stromal deposition of periostin was detected only in grade IV gliomas. Secreted periostin promoted glioma cell invasion and adhesion, and periostin knockdown markedly impaired survival of xenografted glioma stem cells. Interactions with αvß3 and αvß5 integrins promoted adhesion and migration, and periostin abrogated cytotoxicity of the αvß3/ß5 specific inhibitor cilengitide. Periostin-associated gene signatures, predominated by matrix and secreted proteins, corresponded to patient prognosis and functional motifs related to increased malignancy. CONCLUSION: Periostin is a robust marker of glioma malignancy and potential tumor recurrence. Abrogation of glioma stem cell tumorigenicity after periostin inhibition provides support for exploring the therapeutic impact of targeting periostin.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Glioma/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Adhesión Celular , Moléculas de Adhesión Celular/antagonistas & inhibidores , Línea Celular Tumoral , Glioma/mortalidad , Glioma/patología , Glioma/prevención & control , Humanos , Integrinas/metabolismo , Estimación de Kaplan-Meier , Clasificación del Tumor , Invasividad Neoplásica , Regulación hacia Arriba
17.
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
18.
Cancer Med ; 2(4): 478-87, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24156020

RESUMEN

L19-tumor necrosis factor alpha (L19mTNF-α; L), a fusion protein consisting of mouse TNFα and the human antibody fragment L19 directed to the extra domain-B (ED-B) of fibronectin, is able to selectively target tumor vasculature and to exert a long-lasting therapeutic activity in combination with melphalan (M) in syngeneic mouse tumor models. We have studied the antitumor activity of single L19mTNF-α treatment in combination with melphalan and gemcitabine (G) using different administration protocols in two histologically different murine tumor models: WEHI-164 fibrosarcoma and K7M2 osteosarcoma. All responding mice showed significant reduction in myeloid-derived suppressor cells (MDSCs) and an increase in CD4(+) and CD8(+) T cells in the tumor infiltrates, as well as significant reduction in regulatory T cells (Treg) at the level of draining lymph nodes. What is important is that all cured mice rejected tumor challenge up to 1 year after therapy. Targeted delivery of L19mTNF-α synergistically increases the antitumor activity of melphalan and gemcitabine, but optimal administration schedules are required. This study provides information for designing clinical studies using L19mTNF-α in combination with chemotherapeutic drugs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Modelos Animales de Enfermedad , Esquema de Medicación , Sinergismo Farmacológico , Memoria Inmunológica , Depleción Linfocítica , Linfocitos Infiltrantes de Tumor , Melfalán/administración & dosificación , Ratones , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/mortalidad , Proteínas Recombinantes de Fusión/administración & dosificación , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Gemcitabina
19.
Eur J Cancer ; 47(14): 2221-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21605971

RESUMEN

INTRODUCTION: Periostin (PN), a member of the fasciclin family of proteins, is a TGF-ß-induced extracellular matrix protein involved in cell survival, angiogenesis, invasion and metastasis. It is considered a potent angiogenic factor and a marker of tumour progression in many types of human cancer. Many different kinds of cells bind to PN by means of the integrins αvß3 and αvß5, but the periostin epitope recognised by these integrins is not formally demonstrated. The aim of our study was to identify which domain of PN could be involved in cell adhesion and its potential role in tumour growth. METHODS: We generated the monoclonal antibody OC-20 (mAb OC-20) by hybridoma technology. Different PN recombinant fragments were used to characterise the periostin epitope recognised by the mAb OC-20 and to localise a new cell binding site of the protein. A murine model of human melanoma was used in the preclinical in vivo experiments. RESULTS: We formally demonstrate that the periostin epitope recognised by OC-20 is a new binding site for the integrins αvß3 and αvß5, localised in the second FAS1 domain (FAS1-2) of the protein. Moreover the in vivo use of this antibody significantly inhibits tumour growth and angiogenesis. CONCLUSION: Our results show that the FAS1-2 domain of PN plays a role in tumour progression. Moreover this novel antibody may likewise prove to be very useful in clarifying the role of PN in angiogenesis and may contribute to the design of novel anti-angiogenesis drugs.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Moléculas de Adhesión Celular/metabolismo , Integrinas/metabolismo , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Secuencias de Aminoácidos , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Neoplasias Experimentales , Neuroblastoma/metabolismo , Neuroblastoma/patología , Estructura Terciaria de Proteína , Neoplasias Cutáneas/patología
20.
PLoS One ; 5(2): e9145, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-20161770

RESUMEN

BACKGROUND: Fibronectin (FN) is a multi-domain molecule involved in many cellular processes, including tissue repair, embryogenesis, blood clotting, and cell migration/adhesion. The biological activities of FN are mediated by exposed loops located mainly at the interdomain interfaces that interact with various molecules such as, but not only, integrins. Different FN isoforms arise from the alternative splicing of the pre-mRNA. In malignancies, the splicing pattern of FN pre-mRNA is altered; in particular, the FN isoform containing the extra-domain B (ED-B), a complete FN type III repeat constituted by 91 residues, is undetectable in normal adult tissues, but exhibits a much greater expression in fetal and tumor tissues, and is accumulated around neovasculature during angiogenic processes, thus making ED-B one of the best markers and targets of angiogenesis. The functions of ED-B are still unclear; however, it has been postulated that the insertion of an extra-domain such as ED-B modifies the domain-domain interface and may unmask loops that are otherwise cryptic, thus giving FN new potential activities. METHODOLOGY: We used the mAb C6, which reacts with ED-B containing FN, but not with ED-B-free FN and various recombinant FN fragments containing mutations, to precisely localize the epitopes recognized by the mAb C6. CONCLUSION: We formally demonstrated that the inclusion of the alternatively spliced angiogenesis-associated ED-B leads to the unmasking of the FNIII 8 B-C loop that is cryptic in FN molecules lacking ED-B. Thus, the mAb C6, in addition to providing a new reagent for angiogenesis targeting, represents a new tool for the study of the potential biological functions of the B-C loop of the repeat FNIII 8 that is unmasked during angiogenic processes.


Asunto(s)
Empalme Alternativo , Fibronectinas/genética , Mutación , Secuencias Repetitivas de Ácidos Nucleicos/genética , Animales , Anticuerpos Monoclonales/inmunología , Secuencia de Bases , Sitios de Unión/genética , Western Blotting , Mapeo Epitopo , Epítopos/química , Epítopos/genética , Epítopos/inmunología , Fibronectinas/química , Fibronectinas/inmunología , Humanos , Ratones , Modelos Moleculares , Neoplasias/irrigación sanguínea , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA