Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 31(12): 5356-5370, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28790175

RESUMO

JMJD6 is known to localize in the nucleus, exerting histone arginine demethylase and lysyl hydroxylase activities. A novel localization of JMJD6 in the extracellular matrix, resulting from its secretion as a soluble protein, was unveiled by a new anti-JMJD6 mAb called P4E11, which was developed to identify new targets in the stroma. Recombinant JMJD6 binds with collagen type I (Coll-I), and distinct JMJD6 peptides interfere with collagen fibrillogenesis, collagen-fibronectin interaction, and adhesion of human tumor cells to the collagen substrate. P4E11 and collagen binding to JMJD6 are mutually exclusive because the amino acid sequences of JMJD6 necessary for the interaction with Coll-I are part of the conformational epitope recognized by P4E11. In mice injected with mouse 4T1 breast carcinoma cells, treatment with P4E11 reduced fibrosis at the primary tumor and prevented lung metastases. Reduction of fibrosis has also been documented in human breast and ovarian tumors (MDA-MB-231 and IGROV1, respectively) xenotransplanted into immunodeficient mice treated with P4E11. In summary, this study uncovers a new localization and function for JMJD6 that is most likely independent from its canonical enzymatic activities, and demonstrates that JMJD6 can functionally interact with Coll-I. P4E11 mAb, inhibiting JMJD6/Coll-I interaction, represents a new opportunity to target fibrotic and tumor diseases.-Miotti, S., Gulino, A., Ferri, R., Parenza, M., Chronowska, A., Lecis, D., Sangaletti, S., Tagliabue, E., Tripodo, C., Colombo, M. P. Antibody-mediated blockade of JMJD6 interaction with collagen I exerts antifibrotic and antimetastatic activities.


Assuntos
Anticorpos Monoclonais/metabolismo , Colágeno Tipo I/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Colágeno Tipo I/genética , Ensaio de Imunoadsorção Enzimática , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Osteonectina/genética , Osteonectina/metabolismo , Biblioteca de Peptídeos , Ligação Proteica , Receptores de Superfície Celular/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Clin Cancer Res ; 23(17): 5149-5161, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28600479

RESUMO

Purpose: Osteosarcoma, the most common primary bone tumor, is characterized by an aggressive behavior with high tendency to develop lung metastases as well as by multiple genetic aberrations that have hindered the development of targeted therapies. New therapeutic approaches are urgently needed; however, novel combinations with immunotherapies and checkpoint inhibitors require suitable preclinical models with intact immune systems to be properly tested.Experimental Design: We have developed immunocompetent osteosarcoma models that grow orthotopically in the bone and spontaneously metastasize to the lungs, mimicking human osteosarcoma. These models have been used to test the efficacy of trabectedin, a chemotherapeutic drug utilized clinically for sarcomas and ovarian cancer.Results: Trabectedin, as monotherapy, significantly inhibited osteosarcoma primary tumor growth and lung metastases by both targeting neoplastic cells and reprogramming the tumor immune microenvironment. Specifically, trabectedin induced a striking differentiation of tumor cells by favoring the recruitment of Runx2, the master genetic regulator of osteoblastogenesis, on the promoter of genes involved in the physiologic process of terminal osteoblast differentiation. Differentiated neoplastic cells, as expected, showed reduced proliferation rate. Concomitantly, trabectedin enhanced the number of tumor-infiltrating T lymphocytes, with local CD8 T cells, however, likely post-activated or exhausted, as suggested by their high expression of the inhibitory checkpoint molecule PD-1. Accordingly, the combination with a PD-1-blocking antibody significantly increased trabectedin efficacy in controlling osteosarcoma progression.Conclusions: These results demonstrate the therapeutic efficacy of trabectedin in osteosarcoma treatment, unveiling its multiple activities and providing a solid rationale for its combination with immune checkpoint inhibitors. Clin Cancer Res; 23(17); 5149-61. ©2017 AACR.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Dioxóis/efeitos adversos , Neoplasias Pulmonares/tratamento farmacológico , Osteossarcoma/tratamento farmacológico , Tetra-Hidroisoquinolinas/efeitos adversos , Neoplasias Ósseas/genética , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Reprogramação Celular/efeitos dos fármacos , Reprogramação Celular/imunologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Dioxóis/administração & dosagem , Humanos , Imunoterapia/métodos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Osteossarcoma/genética , Osteossarcoma/imunologia , Osteossarcoma/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Tetra-Hidroisoquinolinas/administração & dosagem , Trabectedina , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Proteína Supressora de Tumor p53/genética
3.
Mol Cancer Ther ; 16(2): 365-375, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27980106

RESUMO

Prostate cancer is a leading cause of cancer-related death in males worldwide. Indeed, advanced and metastatic disease characterized by androgen resistance and often associated with neuroendocrine (NE) differentiation remains incurable. Using the spontaneous prostate cancer TRAMP model, we have shown that mast cells (MCs) support in vivo the growth of prostate adenocarcinoma, whereas their genetic or pharmacologic targeting favors prostate NE cancer arousal. Aiming at simultaneously targeting prostate NE tumor cells and MCs, both expressing the cKit tyrosine kinase receptor, we have tested the therapeutic effect of imatinib in TRAMP mice. Imatinib-treated TRAMP mice experience a partial benefit against prostate adenocarcinoma, because of inhibition of supportive MCs. However, they show an unexpected outgrowth of prostate NE tumors, likely because of defective signaling pathway downstream of cKit receptor. Also unexpected but very effective was the inhibition of epithelial-stromal tumors of the seminal vesicles achieved by imatinib treatment. These tumors normally arise in the seminal vesicles of TRAMP mice, independently of the degree of prostatic glandular lesions, and resemble phyllodes tumors found in human prostate and seminal vesicles, and in breast. In both mice and in patients, these tumors are negative for cKit but express PDGFR-ß, another tyrosine kinase receptor specifically inhibited by imatinib. Our results imply a possible detrimental effect of imatinib in prostate cancer patients but suggest a promising therapeutic application of imatinib in the treatment of recurrent or metastatic phyllodes tumors. Mol Cancer Ther; 16(2); 365-75. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Mesilato de Imatinib/farmacologia , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Animais , Biomarcadores , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/patologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Glândulas Seminais/metabolismo , Glândulas Seminais/patologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cell Rep ; 17(1): 233-248, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27681434

RESUMO

The extracellular matrix (ECM) contributes to the biological and clinical heterogeneity of breast cancer, and different prognostic groups can be identified according to specific ECM signatures. In high-grade, but not low-grade, tumors, an ECM signature characterized by high SPARC expression (ECM3) identifies tumors with increased epithelial-to-mesenchymal transition (EMT), reduced treatment response, and poor prognosis. To better understand how this ECM3 signature is contributing to tumorigenesis, we expressed SPARC in isogenic cell lines and found that SPARC overexpression in tumor cells reduces their growth rate and induces EMT. SPARC expression also results in the formation of a highly immunosuppressive microenvironment, composed by infiltrating T regulatory cells, mast cells, and myeloid-derived suppressor cells (MDSCs). The ability of SPARC to induce EMT depended on the localization and suppressive function of myeloid cells, and inhibition of the suppressive function MDSCs by administration of aminobisphosphonates could revert EMT, rendering SPARC-overexpressing tumor cells sensitive to Doxil. We conclude that that SPARC is regulating the interplay between MDSCs and the ECM to drive the induction of EMT in tumor cells.


Assuntos
Neoplasias da Mama/imunologia , Transição Epitelial-Mesenquimal/imunologia , Matriz Extracelular/imunologia , Células Mieloides/imunologia , Células Supressoras Mieloides/imunologia , Osteonectina/genética , Animais , Apresentação de Antígeno , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Celecoxib/farmacologia , Linhagem Celular Tumoral , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Transição Epitelial-Mesenquimal/genética , Matriz Extracelular/patologia , Feminino , Expressão Gênica , Humanos , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Mieloides/efeitos dos fármacos , Células Mieloides/patologia , Células Supressoras Mieloides/efeitos dos fármacos , Células Supressoras Mieloides/patologia , Gradação de Tumores , Osteonectina/deficiência , Polietilenoglicóis/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cancer Immunol Immunother ; 62(12): 1811-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24136641

RESUMO

Intra-tumor injection of immunotherapeutic agents is often the most effective, likely because of concomitant modification of tumor microenvironment. We tested an immunotherapeutic regimen consisting of CpG oligonucleotides and of adenovirus-mediated gene delivery of CCL16 chemokine directly into orthotopically implanted prostate tumors by ultrasound-guided injection, followed by systemic administration of an anti-IL-10R antibody. This combination treatment induced rapid stromal rearrangement, characterized by massive leukocyte infiltration and large areas of necrosis, a scenario that eventually led to complete tumor rejection and systemic immunity in 75 % of the treated mice. In vivo T lymphocyte depletion experiments demonstrated that the efficacy of CCL16/CpG/anti-IL-10R combination treatment relies upon CD8 T lymphocytes whereas CD4 T cells are dispensable. The results underlie the feasibility of echo-guided local immunotherapy of tumors located in visceral organs that are not easily accessible.


Assuntos
Quimiocinas CC/genética , Terapia Genética , Imunoterapia , Interleucina-10/antagonistas & inibidores , Oligodesoxirribonucleotídeos/genética , Neoplasias da Próstata/terapia , Ultrassonografia , Adenoviridae/genética , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Terapia Combinada , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Injeções Intralesionais , Interleucina-10/imunologia , Interleucina-10/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/imunologia , Células Tumorais Cultivadas
6.
Blood ; 120(15): 3007-18, 2012 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-22932797

RESUMO

Antineutrophil cytoplasmic antibodies (ANCAs) target proteins normally retained within neutrophils, indicating that cell death is involved in the autoimmunity process. Still, ANCA pathogenesis remains obscure. ANCAs activate neutrophils inducing their respiratory burst and a peculiar form of cell death, named NETosis, characterized by formation of neutrophil extracellular traps (NETs), decondensed chromatin threads decorated with cytoplasmic proteins endorsed with antimicrobial activity. NETs have been consistently detected in ANCA-associated small-vessel vasculitis, and this association prompted us to test whether the peculiar structure of NET favors neutrophil proteins uploading into myeloid dendritic cells and the induction of ANCAs and associated autoimmunity. Here we show that myeloid DCs uploaded with and activated by NET components induce ANCA and autoimmunity when injected into naive mice. DC uploading and autoimmunity induction are prevented by NET treatment with DNAse, indicating that NET structural integrity is needed to maintain the antigenicity of cytoplasmic proteins. We found NET intermingling with myeloid dendritic cells also positive for neutrophil myeloperoxidase in myeloperoxidase-ANCA-associated microscopic poliangiitis providing a potential correlative picture in human pathology. These data provide the first demonstration that NET structures are highly immunogenic such to trigger adaptive immune response relevant for autoimmunity.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/etiologia , Anticorpos Anticitoplasma de Neutrófilos/sangue , Anticorpos Anticitoplasma de Neutrófilos/imunologia , Autoimunidade/imunologia , Citosol/imunologia , Células Dendríticas/imunologia , Células Mieloides/imunologia , Neutrófilos/imunologia , Animais , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/patologia , Apoptose , Autoantígenos , Western Blotting , Diferenciação Celular , Proliferação de Células , Citosol/metabolismo , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Imunização , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Neutrófilos/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/imunologia , Pele/metabolismo , Pele/patologia
7.
Am J Pathol ; 179(6): 3000-10, 2011 12.
Artigo em Inglês | MEDLINE | ID: mdl-22001347

RESUMO

Fibrosis results from inflammatory tissue damage and impaired regeneration. In the context of bleomycin-induced pulmonary fibrosis, we demonstrated that the matricellular protein termed secreted protein acidic and rich in cysteine (SPARC) distinctly regulates inflammation and collagen deposition, depending on its cellular origin. Reciprocal Sparc(-/-) and wild-type (WT) bone marrow chimeras revealed that SPARC expression in host fibroblasts is required and sufficient to induce collagen fibrosis in a proper inflammatory environment. Accordingly, Sparc(-/-) >WT chimeras showed exacerbated inflammation and fibrosis due to the inability of Sparc(-/-) macrophages to down-regulate tumor necrosis factor production because of impaired responses to tumor growth factor-ß. Hence, the use of bone marrow cells expressing a dominant-negative form of tumor growth factor-ß receptor type II under the monocyte-specific CD68 promoter, as a decoy, phenocopied Sparc(-/-) donor chimeras. Our results point to an unexpected dual role of SPARC in oppositely influencing the outcome of fibrosis.


Assuntos
Fibroblastos/metabolismo , Macrófagos/fisiologia , Osteonectina/fisiologia , Pneumonia/induzido quimicamente , Fibrose Pulmonar/induzido quimicamente , Fator de Crescimento Transformador beta/fisiologia , Animais , Bleomicina/toxicidade , Células da Medula Óssea/metabolismo , Quimera , Colágeno/metabolismo , Regulação para Baixo , Leucócitos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Osteonectina/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
8.
Cancer Res ; 71(18): 5987-97, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21896641

RESUMO

Mast cells (MC) are c-Kit-expressing cells, best known for their primary involvement in allergic reactions, but recently reappraised as important players in either cancer promotion or inhibition. Here, we assessed the role of MCs in prostate tumor development. In prostate tumors from both tumor-prone transgenic adenocarcinoma of the mouse prostate (TRAMP) mice and human patients, MCs are specifically enriched and degranulated in areas of well-differentiated (WD) adenocarcinoma but not around poorly differentiated (PD) foci that coexist in the same tumors. We derived novel TRAMP tumor cell lines, representative of WD and PD variants, and through pharmacologic stabilization or genetic ablation of MCs in recipients mice, we showed that MCs promote WD adenocarcinoma growth but are dispensable for PD tumors. WD tumors rely on MCs for matrix metalloprotease 9 (MMP-9) provision, as reconstitution of MC-deficient mice with wild-type but not MMP-9(-/-) MCs was sufficient to promote their growth. In contrast, PD tumors are MMP-9 self-competent, consistently with epithelial-to-mesenchymal transition. Such a dual source of MMP-9 was confirmed in human tumors, suggesting that MCs could be a good target for early-stage prostate cancer. Interestingly, in testing whether MC targeting could block or delay tumorigenesis in tumor-prone TRAMP mice, we observed a high incidence of early and aggressive tumors, characterized by a neuroendocrine (NE) signature and c-Kit expression. Taken together, these data underscore the contribution of MCs in tumor progression and uncover a new, opposite role of MCs in protecting against the occurrence of aggressive NE variants in prostate cancer.


Assuntos
Adenocarcinoma/patologia , Carcinoma Neuroendócrino/patologia , Mastócitos/patologia , Neoplasias da Próstata/patologia , Adenocarcinoma/enzimologia , Animais , Carcinoma Neuroendócrino/enzimologia , Degranulação Celular/fisiologia , Linhagem Celular Tumoral , Progressão da Doença , Transição Epitelial-Mesenquimal , Humanos , Masculino , Mastócitos/enzimologia , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias da Próstata/enzimologia , Proteínas Proto-Oncogênicas c-kit/biossíntese , Membro 25 de Receptores de Fatores de Necrose Tumoral , Microambiente Tumoral
9.
Cancer Res ; 68(21): 9050-9, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18974151

RESUMO

Other than genetic imprinting and epithelial to mesenchymal transition, cancer cells need interaction with the nearby stroma toward metastasis. Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein known to regulate extracellular matrix (ECM) deposition and cell-ECM interaction. Gene expression profiles associate SPARC to malignant progression. Using reciprocal bone marrow chimeras between SPARC knockout and wild-type mice, we show that SPARC produced by inflammatory cells is necessary for spontaneous, but not experimental, i.v. metastasis. Macrophage-derived SPARC induces cancer cell migration and enhances their migration to other ECM proteins at least through alpha(v)beta(5) integrin. Indeed, RNA interference knockdown of beta(5) integrin expression reduces cell migration in vitro and metastasis in vivo. Together these results show that macrophage-derived SPARC takes part in metastasis, acting at the step of integrin-mediated migration of invasive cells.


Assuntos
Matriz Extracelular/metabolismo , Macrófagos/metabolismo , Metástase Neoplásica , Osteonectina/fisiologia , Animais , Sequência de Bases , Primers do DNA , Fibronectinas/metabolismo , Citometria de Fluxo , Inativação Gênica , Imuno-Histoquímica , Cadeias beta de Integrinas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Osteonectina/genética , RNA Interferente Pequeno
10.
Oncogene ; 24(27): 4349-61, 2005 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-15824736

RESUMO

Altered expression of CCN3 has been observed in a variety of musculoskeletal tumours, including Ewing's sarcoma (ES). Despite its widespread distribution, very little is known about its biological functions and molecular mechanisms of action. We transfected CCN3 gene into a CCN3-negative ES cell line and analysed the in vitro and in vivo behaviours of stably transfected clones. Forced expression of CCN3 significantly reduced cell proliferation in vitro, growth in anchorage-independent conditions, and tumorigenicity in nude mice. Despite the antiproliferative effect, CCN3-transfected ES cells displayed increased migration and invasion of Matrigel. The decreased expression of alpha2beta1 integrin receptor and the increased amount of cell surface-associated matrix metalloproteinase (MMP)-9 following the expression of CCN3 may be the basis for the increased migratory abilities of transfected cells. Cells lacking alpha2beta1 are less facilitated to have stable anchorage since the predominant collagen extracted from ES tissue is indeed type I collagen, and proMMP-9 was recently found to provide a cellular switch between stationary and migratory ES cell phase. Our findings are in line with those recently obtained in glioblastoma. However, the underlying molecular mechanisms appear to be different, further highlighting the importance of the cellular context in the regulation of function of CCN proteins.


Assuntos
Movimento Celular , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Animais , Adesão Celular , Linhagem Celular , Proliferação de Células , Células Clonais/metabolismo , Células Clonais/patologia , Fator de Crescimento do Tecido Conjuntivo , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Nus , Invasividade Neoplásica , Proteína Sobre-Expressa em Nefroblastoma , Receptor IGF Tipo 1/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Sarcoma de Ewing/genética , Transfecção
11.
Cancer Res ; 65(6): 2321-9, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15781646

RESUMO

Although in vitro establishment of new colorectal carcinoma (CRC) cell lines is an infrequent event, we have observed that primary cultures of CRC can be repeatedly and reproducibly initiated following in vitro plating of tumor-derived epithelial cells. These cultures, however, usually display a short life span as they undergo a limited number of cell passages before entering a state of irreversible growth arrest. In this study, we show that short-lived CRC primary cultures lack constitutive telomerase activity and undergo a senescence process characterized by progressive telomere shortening. Moreover, transduction of these cells with a retroviral vector encoding human telomerase reverse transcriptase (hTERT) is sufficient to reconstitute telomerase activity and allow immortalization. Detailed molecular characterization of hTERT-immortalized CRC cell lines confirms their individual tumor origin by showing expression of colonic epithelial differentiation markers, such as cytokeratin-20 (CK20), full match with class I and class II human leukocyte antigen genotyping of autologous B-lymphoblastoid cells, and presence of somatic mutations in key cancer genes (KRAS2, APC) identical to those of the corresponding autologous original tumor tissues. Moreover, functional characterization of hTERT-immortalized CRC cell lines shows that they have a transformed phenotype, being able to form colonies in soft agar and tumors in severe combined immunodeficient mice. Most interestingly, immunohistochemical analysis of original tumor tissues indicates that short-lived CRC primary cultures, although hTERT-negative in vitro, derive from hTERT-positive tumors. Taken together, our data show that, in a least subset of CRC, biochemical pathways involved in maintenance of telomere length, such as telomerase, are not activated in a constitutive way in all tumor cells.


Assuntos
Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Telomerase/biossíntese , Animais , Senescência Celular/fisiologia , Neoplasias Colorretais/genética , Proteínas de Ligação a DNA , Feminino , Vetores Genéticos , Humanos , Camundongos , Camundongos SCID , Transplante de Neoplasias , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Retroviridae/genética , Telomerase/genética , Telomerase/metabolismo , Telômero/fisiologia , Transdução Genética , Transplante Heterólogo , Células Tumorais Cultivadas
12.
J Immunol ; 172(7): 4026-36, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15034014

RESUMO

The CC chemokine ligand (CCL)16 exerts chemotactic activity on human monocytes and lymphocytes. Although no murine homologous has been defined, the TSA mouse adenocarcinoma cells engineered to express human CCL16 are rapidly rejected by syngenic mice. An adenovirus encoding CCL16 (AdCCL16) was generated using a Cre-Lox-based system and was used to determine whether this chemokine might also block pre-existing tumors. Both recombinant and viral CCL16 showed in vitro chemotactic activity for murine CD4(+) and CD8(+) lymphocytes and dendritic cells (DC). AdCCL16, but not the control empty vector, when injected in established nodules significantly delayed tumor growth. Immunohistochemistry revealed accumulation of CD4(+) and CD8(+) T cells and DC in the treated tumors as well as in draining lymph nodes. DC from such lymph nodes stimulated IFN-gamma by a T cell clone specific for the known TSA tumor-associated Ag (TAA), suggesting the tumor origin of these cells. Lymphocytes from the same nodes showed specific CTL activity against TSA tumor cells and their immunodominant TAA peptide. Antitumor activity required CD4, CD8, and IFN-gamma production, as shown using subset-depleted and knockout mice. Despite the robust and rapid immune response triggered by intratumoral injection of AdCCL16, the lesions were not completely rejected; however, the same treatment given before surgical excision of primary lesions prevented metastatic spread and cured 63% of mice bearing the 4T1 mammary adenocarcinoma, which is perhaps the most compelling model of spontaneous metastasis.


Assuntos
Adenocarcinoma/mortalidade , Adenocarcinoma/cirurgia , Adenoviridae/genética , Antineoplásicos/administração & dosagem , Quimiocinas CC/administração & dosagem , Inibidores do Crescimento/administração & dosagem , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/cirurgia , Adenocarcinoma/imunologia , Adenocarcinoma/secundário , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Quimiocinas CC/genética , Terapia Combinada/métodos , Terapia Combinada/mortalidade , Células Dendríticas/imunologia , Células Dendríticas/patologia , Epitopos de Linfócito T/imunologia , Feminino , Vetores Genéticos , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/mortalidade , Inibidores do Crescimento/genética , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Injeções Intralesionais , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/prevenção & controle , Linfonodos/imunologia , Linfonodos/patologia , Metástase Linfática/imunologia , Metástase Linfática/prevenção & controle , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/mortalidade , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia
13.
Cancer Res ; 64(4): 1502-8, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14973071

RESUMO

Tumor immunotherapy has exploited the ability of heat shock proteins to chaperone precursors of antigenic peptides to antigen-presenting cells and to activate efficiently an immune response against tumor-associated antigens. The most common strategy is based on the purification of heat shock protein-peptide complexes from tumor cell lines or from tumor surgical samples for in vivo administration. In this article, we have modified the murine-inducible hsp70 into a secreted protein and engineered tumor cells to secrete constitutively their antigenic repertoire associated with the hsp70 protein. In vitro studies showed that the relocalization of hsp70 from the cytoplasm to the secretory pathway did not modify the ability of hsp70 to interact with peptides derived either from natural tumor-associated antigens or model antigens, and that antigen-presenting cells specifically took up the secreted hsp70 and presented the chaperoned epitopes to T cells. In vivo studies showed that tumors secreting hsp70 displayed increased immunogenicity, with induction of a strong and specific CTL response. Mice injected with hsp70-secreting tumors showed increased survival and impaired tumor take compared with mice bearing parental tumors. More than 70% of mice rejected tumor cells secreting hsp70 through mechanisms that involve T lymphocytes and natural killer cells, with the induction of a memory response in the case of T lymphocytes. Moreover, hsp70 secretion increased the immunogenic potential of tumor cell vaccines.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Imunoterapia Adotiva , Neoplasias/terapia , Animais , Células Apresentadoras de Antígenos/fisiologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Feminino , Proteínas de Choque Térmico HSP70/genética , Memória Imunológica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Transfecção
14.
Blood ; 101(2): 568-75, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12393660

RESUMO

Cytokine gene-modified tumor cells have increased immunogenicity and retain the antigenic repertoire of a particular neoplasia. However, practical concerns have led to an increased interest in allogeneic gene-transduced bystander cells as a broader source of cytokines for autologous tumor cell-based vaccines. Here, we show that allogeneic B78H1 major histocompatibility complex (MHC) class I-negative and -positive (H-2K(b)- and D(b)-transfected) cells induced cytotoxic T lymphocytes (CTLs) and protection in BALB/c mice at comparable levels in response to a challenge with C26 (H-2(d)) colon carcinoma cells sharing the tumor-associated antigen envelope glycoprotein 70 (env-gp70) with both cell lines. Class I-negative B78H1 cells transduced to express interleukin-12 (IL-12) and mixed with autologous A20 tumor cells led to eradication of preestablished A20 lymphoma in 50% or 100% of treated mice after 3 or 4 vaccinations, respectively, whereas A20 cells alone or mixed with nontransduced B78H1 cured none or 50% of mice after 3 or 4 vaccinations, respectively. Immunization with the IL-12-producing bystander cell line increased tumor-specific proliferation and type 1 cytokine production by CD4(+) T cells. By contrast, CD4 T-cell function appeared impaired after immunization with A20 cells alone or mixed with B78H1 cells. Indeed, only CD4(+) T cells from IL-12-treated mice could be restimulated with anti-OX40 monoclonal antibody (mAb) in place of a fourth cellular boost. Moreover, the IL-12-based tumor vaccine induced expansion of tumor-specific interferon-gamma (IFN-gamma)-producing CD8(+) T cells. These results are clinically relevant for the development of feasible IL-12 cancer vaccines based on engineered class I-negative bystander cells.


Assuntos
Vacinas Anticâncer , Antígenos de Histocompatibilidade Classe I/imunologia , Linfoma/patologia , Linfoma/terapia , Receptores do Fator de Necrose Tumoral , Animais , Efeito Espectador/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacologia , Citocinas/biossíntese , Citocinas/metabolismo , Imunidade Celular , Interleucina-12/biossíntese , Interleucina-12/genética , Interleucina-12/metabolismo , Ativação Linfocitária , Linfoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Receptores OX40 , Linfócitos T Citotóxicos/imunologia , Transdução Genética , Resultado do Tratamento , Células Tumorais Cultivadas , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...