Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
1.
ESMO Open ; 8(4): 101590, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37393630

RESUMO

BACKGROUND: Gene expression profiling (GEP)-based prognostic signatures are being rapidly integrated into clinical decision making for systemic management of breast cancer patients. However, GEP remains relatively underdeveloped for locoregional risk assessment. Yet, locoregional recurrence (LRR), especially early after surgery, is associated with poor survival. PATIENTS AND METHODS: GEP was carried out on two independent luminal-like breast cancer cohorts of patients developing early (≤5 years after surgery) or late (>5 years) LRR and used, by a training and testing approach, to build a gene signature able to intercept women at risk of developing early LRR. The GEP data of two in silico datasets and of a third independent cohort were used to explore its prognostic value. RESULTS: Analysis of the first two cohorts led to the identification of three genes, CSTB, CCDC91 and ITGB1, whose expression, derived by principal component analysis, generated a three-gene signature significantly associated with early LRR in both cohorts (P value <0.001 and 0.005, respectively), overcoming the discriminatory capability of age, hormone receptor status and therapy. Remarkably, the integration of the signature with these clinical variables led to an area under the curve of 0.878 [95% confidence interval (CI) 0.810-0.945]. In in silico datasets we found that the three-gene signature retained its association, showing higher values in the early relapsed patients. Moreover, in the third additional cohort, the signature significantly associated with relapse-free survival (hazard ratio 1.56, 95% CI 1.04-2.35). CONCLUSIONS: Our three-gene signature represents a new exploitable tool to aid treatment choice in patients with luminal-like breast cancer at risk of developing early recurrence.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Neoplasias da Mama/genética , Neoplasias da Mama/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Prognóstico , Transcriptoma , Medição de Risco
3.
Physiol Rev ; 100(4): 1839-1850, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32721181

RESUMO

Cancer patients appear to be more likely to be diagnosed with coronavirus disease 2019 (COVID-19). This is supported by the understanding of immunometabolic pathways that intersect patients with infection and cancer. However, data derived by case series and retrospective studies do not offer a coherent interpretation, since data from China suggest an increased risk of COVID-19, while data from the United States and Italy show a prevalence of COVID-19 in cancer patients comparable with the general population. Noteworthy, cancer and COVID-19 exploit distinct patterns of macrophage activation that promote disease progression in the most severe forms. In particular, the alternative activation of M2-polarized macrophages plays a crucial role in cancer progression. In contrast, the macrophage-activation syndrome appears as the source of M1-related cytokine storm in severe COVID-19 disease, thus indicating macrophages as a source of distinct inflammatory states in the two diseases, nonetheless as a common therapeutic target. New evidence indicates that NAMPT/NAD metabolism can direct both innate immune cell effector functions and the homeostatic robustness, in both cancer and infection. Moreover, a bidirectional relationship exists between the metabolism of NAD and the protective role that angiotensin converting enzyme 2, the COVID-19 receptor, can play against hyperinflammation. Within this immunometabolic framework, the review considers possible interference mechanisms that viral infections and tumors elicit on therapies and provides an overview for the management of patients with cancer affected by COVID-19, particularly for the balance of risk and benefit when planning normally routine cancer treatments and follow-up appointments.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/imunologia , Neoplasias/imunologia , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/imunologia , Enzima de Conversão de Angiotensina 2 , Animais , Betacoronavirus/imunologia , COVID-19 , Infecções por Coronavirus/complicações , Citocinas/metabolismo , Humanos , Neoplasias/complicações , Nicotinamida Fosforribosiltransferase/metabolismo , Pandemias , Peptidil Dipeptidase A/imunologia , Pneumonia Viral/complicações , SARS-CoV-2
4.
Cancer Treat Rev ; 75: 39-51, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30954906

RESUMO

Immunotherapy has dramatically changed the therapeutic scenario in treatment naïve advanced non-small cell lung cancer (NSCLC). While single agent pembrolizumab has become the standard therapy in patients with PD-L1 expression on tumor cells ≥ 50%, the combination of pembrolizumab or atezolizumab and platinum-based chemotherapy has emerged as an effective first line treatment regardless of PD-L1 expression both in squamous and non-squamous NSCLC without oncogenic drivers. Furthermore, double immune checkpoint inhibition has shown promising results in treatment naïve patients with high tumor mutational burden (TMB). Of note, the presence of both negative PD-L1 expression and low TMB may identify a subgroup of patients who has little benefit from immunotherapy combinations and for whom the best treatment option may still be platinum-based chemotherapy. To date, first-line single agent immune checkpoint blockade has demonstrated limited activity in EGFR mutated NSCLC and the combination of immunotherapy and targeted agents has raised safety concerns in both EGFR and ALK positive NSCLC patients. Finally, in EGFR mutated or ALK rearranged NSCLC, atezolizumab in combination with platinum-based chemotherapy and bevacizumab is emerging as a potential treatment option upon progression to first line tyrosine kinase inhibitors.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígeno B7-H1/metabolismo , Bevacizumab/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Humanos , Fatores Imunológicos/metabolismo , Imunoterapia/métodos , Neoplasias Pulmonares/metabolismo
5.
Semin Immunol ; 35: 29-34, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29428698

RESUMO

Basophils, eosinophils and mast cells were first recognized by Paul Ehrlich in the late 19th century. These cells have common, but non-redundant roles, in the pathogenesis of allergic diseases and in the protection against parasites. Nevertheless, in virtue of their shared-adeptness to produce a huge variety of immunological mediators and express membrane-bound receptors, they are able to interact with immune and non-immune components of the tissue microenvironment, contributing to the regulation of tissue homeostasis and immune response while participating to further deregulation of tissues transforming into neoplasia.


Assuntos
Basófilos/imunologia , Eosinófilos/imunologia , Hipersensibilidade/imunologia , Mastócitos/imunologia , Neoplasias/imunologia , Animais , Transformação Celular Neoplásica , Microambiente Celular , Homeostase , Humanos
6.
Cytokine Growth Factor Rev ; 38: 1-9, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29029813

RESUMO

The seventh Edition of "Innovative Therapy, Monoclonal Antibodies and Beyond" Meeting took place in Milan, Italy, on January 27, 2017. The two sessions of the meeting were focused on: 1) Preclinical assays and novel biotargets; and 2) monoclonal antibodies, cell therapies and targeted molecules. Between these two sessions, a lecture entitled "HLA-antigens modulation and response to immune checkpoint inhibitor immunotherapy" was also presented. Despite the impressive successes in cancer immunotherapy in recent years, the response to immune based interventions occurs only in a minority of patients (∼20%). Several basic and translational mechanisms of resistance to immune checkpoint blockers (ICBs) were discussed during the meeting: 1. the impact of tumor microenvironment on the activity of immune system; 2. strategies to inhibit the cross-talk between extracellular matrix and myeloid-derived suppressor cells (MDSC) in the preclinical setting; 3. microRNA expression as a biomarker and as a target of therapy in non-small cell lung cancer (NSCLC); 4. the significance of complement activation pathways in response to immune checkpoint inhibitors; 5. the immunosuppressive activity of the microbiota by inducing IL-17 producing cells; and 6. modulation of HLA antigens as possible markers of response to ICB therapy. In order to overcome the deficiency in active anti-tumor T cells, several clinically applicable combination strategies were also discussed: 1. strategies to enhance the anticancer effects of immunogenic cell death inducing-chemotherapy; 2. the use of CAR T-cells in solid tumors; 3. the use of combination strategies involving oncolytic viruses and ICBs; 4. combinations of new ICBs with anti-PD-1/CTLA-4 therapy; and 4. combinations of targeted therapies and ICBs in melanoma. Overall, this conference emphasized the many novel strategies that are being investigated to improve the overall patient response to cancer immunotherapy. Optimization of biomarkers to accurately select patients who will respond to immunotherapy, coupled with combination strategies to improve long term patient survival remain critical challenges in the immuno-oncology field.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoterapia , Neoplasias/terapia , Animais , Humanos , Neoplasias/tratamento farmacológico
7.
Med Biol Eng Comput ; 54(12): 1949-1957, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27099155

RESUMO

Despite the technological improvement of radiologic, endoscopic and nuclear imaging, the accuracy of diagnostic procedures for tumors can be limited whenever a mass-forming lesion is identified. This is true also because bioptical sampling cannot be properly guided into the lesions so as to puncture neoplastic tissue and to avoid necrotic areas. Under these circumstances, invasive and expensive procedures are still required to obtain diagnosis which is mandatory to plan the most appropriate therapeutic strategy. In order to test if electrical impedance spectroscopy may be helpful in providing further evidence for cancer detection, resistivity measurements were taken on 22 mice, 11 wild-type and 11 sparc-/- (knock out for the protein SPARC: secreted protein acidic and rich in cysteine), bearing mammary carcinomas, by placing a needle-probe into tumor, peritumoral and contralateral healthy fat areas. Tumor resistivity was significantly lower than both peritumoral fat and contralateral fat tissues. Resistivity in sparc-/- mice was lower than wild-type animals. A significant frequency dependence of resistivity was present in tissues analyzed. We conclude that accurate measurements of resistivity may allow to discriminate between tissues with different pathological and/or structural characteristics. Therefore, resistivity measurements could be considered for in vivo detection and differential diagnosis of tumor masses.


Assuntos
Neoplasias Mamárias Experimentais/patologia , Osteonectina/deficiência , Animais , Modelos Animais de Doenças , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Camundongos Endogâmicos BALB C , Agulhas , Osteonectina/metabolismo , Ultrassom
8.
Oncogene ; 35(30): 3944-54, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-26616853

RESUMO

Sarcomas are mesenchymal tumors characterized by blocked differentiation process. In Ewing sarcoma (EWS) both CD99 and EWS-FLI1 concur to oncogenesis and inhibition of differentiation. Here, we demonstrate that uncoupling CD99 from EWS-FLI1 by silencing the former, nuclear factor-κB (NF-κB) signaling is inhibited and the neural differentiation program is re-established. NF-κB inhibition passes through miR-34a-mediated repression of Notch pathway. CD99 counteracts EWS-FLI1 in controlling NF-κB signaling through the miR-34a, which is increased and secreted into exosomes released by CD99-silenced EWS cells. Delivery of exosomes from CD99-silenced cells was sufficient to induce neural differentiation in recipient EWS cells through miR-34a inhibition of Notch-NF-κB signaling. Notably, even the partial delivery of CD99 small interfering RNA may have a broad effect on the entire tumor cell population owing to the spread operated by their miR-34a-enriched exosomes, a feature opening to a new therapeutic option.


Assuntos
Antígeno 12E7/fisiologia , MicroRNAs/fisiologia , NF-kappa B/fisiologia , Receptores Notch/fisiologia , Sarcoma de Ewing/patologia , Transdução de Sinais/fisiologia , Diferenciação Celular , Humanos , Proteínas de Fusão Oncogênica/fisiologia , Proteína Proto-Oncogênica c-fli-1/fisiologia , RNA Interferente Pequeno/genética , Proteína EWS de Ligação a RNA/fisiologia
9.
Cancer Microenviron ; 8(3): 167-76, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25194694

RESUMO

Mast cells (MCs) are granulocytic immune cells that reside in tissues exposed to the external environment. MCs are best known for their activity in allergic reactions, but they have been involved in different physiological and pathological conditions. In particular, MC infiltration has been shown in several types of human tumors and in animal cancer models. Nevertheless, the role of MCs in the tumor microenvironment is still debated because they have been associated either to good or poor prognosis depending on tumor type and tissue localization. This dichotomous role relies on MC capacity to secrete a broad spectrum of molecules with modulatory functions, which may condition the final tumor outcome also promoting angiogenesis and tissue remodeling. In this review, we analyze the multifaceted role of mast cell in tumor progression and inhibition considering their ability to interact with: i) immune cells, ii) tumor cells and iii) the extracellular matrix. Eventually, the current MC targeting strategies to treat cancer patients are discussed. Deciphering the actual role of MCs in tumor onset and progression is crucial to identify MC-targeted treatments aimed at killing cancer cells or at making the tumor vulnerable to selected anti-cancer drugs.

10.
Cell Death Dis ; 4: e920, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24232096

RESUMO

Smac mimetics (SMs) comprise a class of small molecules that target members of the inhibitor of apoptosis family of pro-survival proteins, whose expression in cancer cells hinders the action of conventional chemotherapeutics. Herein, we describe the activity of SM83, a newly synthesised dimeric SM, in two cancer ascites models: athymic nude mice injected intraperitoneally with IGROV-1 human ovarian carcinoma cells and immunocompetent BALB/c mice injected with murine Meth A sarcoma cells. SM83 rapidly killed ascitic IGROV-1 and Meth A cells in vivo (prolonging mouse survival), but was ineffective against the same cells in vitro. IGROV-1 cells in nude mice were killed within the ascites by a non-apoptotic, tumour necrosis factor (TNF)-dependent mechanism. SM83 administration triggered a rapid inflammatory event characterised by host secretion of TNF, interleukin-1ß and interferon-γ. This inflammatory response was associated with the reversion of the phenotype of tumour-associated macrophages from a pro-tumoural M2- to a pro-inflammatory M1-like state. SM83 treatment was also associated with a massive recruitment of neutrophils that, however, was not essential for the antitumoural activity of this compound. In BALB/c mice bearing Meth A ascites, SM83 treatment was in some cases curative, and these mice became resistant to a second injection of cancer cells, suggesting that they had developed an adaptive immune response. Altogether, these results indicate that, in vivo, SM83 modulates the immune system within the tumour microenvironment and, through its pro-inflammatory action, leads cancer cells to die by necrosis with the release of high-mobility group box-1. In conclusion, our work provides evidence that SMs could be more therapeutically active than expected by stimulating the immune system.


Assuntos
Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Necrose/induzido quimicamente , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Materiais Biomiméticos/uso terapêutico , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Células HCT116 , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamação/induzido quimicamente , Proteínas Inibidoras de Apoptose , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Oncogene ; 30(7): 757-69, 2011 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-21057534

RESUMO

c-Kit tyrosine kinase receptor and its ligand stem cell factor have multiple functions during development, whereas in adulthood they are mostly needed for stem cell (SC) maintenance and mast cell (MC) biology. c-Kit plays an essential tumor-cell-intrinsic role in many types of cancer, either providing the tumorigenic force when aberrantly activated or conferring stem-like features characterizing the most aggressive variants. A tumor-cell-extrinsic role occurs through c-Kit-dependent accessory cells (such as MCs) that infiltrate tumors and deeply influence their progression. c-Kit-targeted therapy with tyrosine kinase inhibitors (TKIs) may ideally work against both tumor and stromal cells. Here, we summarize the tumor-intrinsic and -extrinsic roles of c-Kit in cancer and discuss TKIs with their on- and off-targets, with a special emphasis on MCs as paradigmatic c-Kit-dependent accomplices for tumor progression.


Assuntos
Antineoplásicos/uso terapêutico , Mastócitos/metabolismo , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/metabolismo , Animais , Humanos , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Mastocitose/irrigação sanguínea , Mastocitose/tratamento farmacológico , Mastocitose/imunologia , Camundongos , Invasividade Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/enzimologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/enzimologia , Ratos
12.
Ann Oncol ; 18(2): 226-32, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17116643

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been and is still widely used as an adjuvant in clinical trials of vaccination with autologous tumor cells, peptides and/or dendritic cells in a variety of human neoplasms. This cytokine was administered either as product of gene-transduced tumor cells or as recombinant protein together with the vaccine given subcutaneously or intradermally. Results of these trials were heterogeneous in terms of induction of vaccine-specific immune response and of clinical response. Though in some of these studies GM-CSF appeared to help in generating an immune response, in others no effect or even a suppressive effect was reported. Here, we review the literature dealing with the immune adjuvant activity of GM-CSF both in animal models and clinical trials. As a consequence of such analysis, we conclude that GM-CSF may increase the vaccine-induced immune response when administered repeatedly at relatively low doses (range 40-80 microg for 1-5 days) whereas an opposite effect was often reported at dosages of 100-500 microg. The potential mechanisms of the GM-CSF-mediated immune suppression are discussed at the light of studies describing the activation and expansion of myeloid suppressor cells by endogenous tumor-derived or exogenous GM-CSF.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Neoplasias/imunologia , Animais , Humanos , Neoplasias/terapia
14.
J Exp Med ; 194(9): 1195-205, 2001 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-11696586

RESUMO

Transgenic Balb/c mice expressing the transforming rat HER-2/neu oncogene develop early and multifocal mammary carcinomas. Within the first 5 months of life the tissue-specific expression of HER-2/neu causes a progression in all their 10 mammary glands from atypical hyperplasia to invasive carcinoma. It was previously observed that chronic administration of interleukin (IL)-12 increased tumor latency, but every mouse eventually succumbed to multiple carcinomas. A significant improvement in tumor prevention was sought by administering allogeneic mammary carcinoma cells expressing HER-2/neu combined with systemic IL-12. This treatment reduced tumor incidence by 90% and more than doubled mouse lifetime. For the maximum prevention p185(neu) antigen must be expressed by allogeneic cells. IL-12 treatment strongly increased the cell vaccine efficacy. The mammary glands of mice receiving the combined treatment displayed a markedly reduced epithelial cell proliferation, angiogenesis, and HER-2/neu expression, while the few hyperplastic foci were heavily infiltrated by granulocytes, macrophages, and CD8(+) lymphocytes. Specific anti-HER-2/neu antibodies were produced and a nonpolarized activation of CD4(+) and CD8(+) cells secreting IL-4 and interferon (IFN)-gamma were evident. A central role for IFN-gamma in the preventive effect was proven by the lack of efficacy of vaccination in IFN-gamma gene knockout HER-2/neu transgenic Balb/c mice. A possible requirement for IFN-gamma is related to its effect on antibody production, in particular on IgG2a and IgG2b subclasses, that were not induced in IFN-gamma knockout HER-2/neu mice. In conclusion, our data show that an allogeneic HER-2/neu-expressing cell vaccine combined with IL-12 systemic treatment can prevent the onset of genetically determined tumors.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/imunologia , Interleucina-12/imunologia , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/fisiologia , Animais , Mama/patologia , Linfócitos T CD4-Positivos/imunologia , Transplante de Células , Feminino , Imunidade Celular/imunologia , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/administração & dosagem , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Ratos , Receptor ErbB-2/genética , Transplante Homólogo , Células Tumorais Cultivadas , Vacinação/métodos
15.
Eur J Immunol ; 31(10): 3101-10, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11592087

RESUMO

The glycolipid alpha-galactosylceramide (alpha-GalCer), ligand of NKT cells, has been recently shown to induce antitumor immunity in mice through the induction of IL-12 production by dendritic cells. In the present study we compared alpha-GalCer and rIL-12 antitumor activities in the treatment of hepatic metastases of the C-26 murine colon carcinoma. We show that in immunocompetent mice the two molecules display similar efficacy, whereas in mice knockout (KO) for beta2-microglobulin (beta2m), IFN-gamma or IL-12p40, alpha-GalCer antitumor activity is severely impaired. Conversely,in all such KO mice, rIL-12 retains its efficacy. In this context, the IL-12 effect relies on NK cell function since it is abrogated by antibodies to NK1.1, expressed by both NK and NKT cells, but not in beta2m KO mice that lack NKT and CD8 T cells, but have a perfectly functional NK cell population. Furthermore, in IFN-gamma and IL-12p40 double KO mice, exogenous rIL-12 completely loses antitumor efficacy, suggesting the existence of an IFN-gamma-independent IL-12 effect that does require the presence of endogenous IL-12p40 chain.


Assuntos
Neoplasias do Colo/terapia , Galactosilceramidas/uso terapêutico , Interleucina-12/uso terapêutico , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Animais , Feminino , Interferon gama/biossíntese , Interleucina-12/biossíntese , Neoplasias Hepáticas Experimentais/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/uso terapêutico , Microglobulina beta-2/fisiologia
16.
Cancer Res ; 61(17): 6532-9, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11522651

RESUMO

We had demonstrated previously a functional bridge between altered homebox (HOX) gene expression and tumor progression through HOXB7 transactivation of basic fibroblast growth factor. Here, we have studied whether HOXB7, in addition to basic fibroblast growth factor, may induce other genes directly or indirectly related to neoangiogenesis and tumor invasion. Parental, beta-galactosidase-transduced, and HOXB7-transduced SkBr3 cell lines were examined for the expression of several growth factors and growth factor receptors involved in the proliferative and angiogenic processes. Vascular endothelial growth factor, melanoma growth-stimulatory activity/growth-related oncogenene alpha, interleukin-8, and angiopoietin-2 were up-regulated by HOXB7 transduction. The exception was angiopoietin-1 expression that was abrogated. Additional analyses included the expression levels of enzymes such as matrix metalloprotease (MMP)-2 and MMP-9 and heparanase, capable of proteolytic degradation of extracellular matrix and basement membranes. Results showed an induction of only MMP-9. The functional implication of such a finding was tested using an in vitro coculture assay in a three-dimensional matrix. A delay of differentiation with persistent nests of proliferating cells was found in endothelial cells cocultured with HOXB7-transduced SkBr3 cells. Tumorigenicity of these cells has been evaluated in vivo. Xenograft into athymic nude mice showed that SkBr3/HOXB7 cells developed tumors in mice, either irradiated or not, whereas parental SkBr3 cells did not show any tumor take unless mice were sublethally irradiated. Comparison of tumor nodules for vascularization by CD-31 and CD-34 immunostaining revealed an increased number of blood vessels in tumors expressing HOXB7. Together, the results indicate HOXB7 as a key factor up-regulating a variety of proangiogenic stimuli. Thus, HOXB7 gene or protein is a target to aim at to inhibit tumor-associated neoangiogenesis, considering the number and the redundancy of proangiogenic molecules that should be targeted one by one to theoretically achieve the same effect.


Assuntos
Adenocarcinoma/irrigação sanguínea , Neoplasias da Mama/irrigação sanguínea , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Neovascularização Patológica/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Angiopoietina-1 , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Técnicas de Cocultura , Fatores de Crescimento Endotelial/biossíntese , Fatores de Crescimento Endotelial/genética , Endotélio Vascular/citologia , Regulação Enzimológica da Expressão Gênica , Glucuronidase/biossíntese , Glucuronidase/genética , Humanos , Linfocinas/biossíntese , Linfocinas/genética , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica/metabolismo , Isoformas de Proteínas , Receptores de Fatores de Crescimento/biossíntese , Receptores de Fatores de Crescimento/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Transplante Heterólogo , Células Tumorais Cultivadas , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
17.
Cancer Immunol Immunother ; 50(4): 199-211, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11459172

RESUMO

In order to construct an immunogenic cellular vaccine, we transduced three HLA-A*0201 human melanoma lines, selected for expression of classes I and II HLA, adhesion molecules and the T cell-defined melanoma antigens Melan/MART-1, gp100 and tyrosinase, with both interleukin-2 (IL-2) and B7-1 genes by the use of a polycistronic retroviral vector. The lines were selected to share only the HLA-A*0201 allele to avoid generation of strong alloreactivity in case of their multiple in vivo use in HLA-A*0201 + patients. Phenotypic and functional analysis of B7-1-IL2 transduced melanoma lines in comparison with B7-1 transduced and/or parental untransduced counterparts were then carried out. Tumor cells expressing either B7-1 or both genes did not change their original antigenic profile. From a functional point of view, expression of both genes in melanoma lines: (1) improved the response of anti-melanoma cytotoxic T lymphocytes (CTL) over singly transduced or untransduced melanoma cells when subthreshold levels of MHC-peptide complexes were expressed by melanoma cells; (2) conferred a distinct advantage in the ability to stimulate cytotoxicity and interferon-gamma release by autologous and/or HLA-A*0201-compatible allogeneic lymphocytes; (3) allowed the generation of a high number of specific CTL by in vitro stimulation of lymphocytes of HLA-A*0201-melanoma patients. Thus, B7-IL2 gene-transduced melanoma lines appear to display a high immunogenicity and could be used as vaccine in melanoma patients.


Assuntos
Antígeno B7-1/genética , Vacinas Anticâncer/imunologia , Interleucina-2/genética , Ativação Linfocitária , Melanoma/imunologia , Transdução Genética , Antígeno B7-1/metabolismo , Citotoxicidade Imunológica , Expressão Gênica , Terapia Genética , Antígenos HLA/metabolismo , Antígeno HLA-A2/metabolismo , Humanos , Imunofenotipagem , Interferon gama/biossíntese , Interleucina-2/metabolismo , Melanoma/genética , Linfócitos T Citotóxicos/imunologia
18.
J Immunol ; 166(6): 3890-9, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11238633

RESUMO

In vivo IL-12-dependent tumor inhibition rests on the ability of IL-12 to activate a CD8-mediated cytotoxicity, inhibit angiogenesis, and cause vascular injury. Although in vivo studies have shown that such inhibition stems from complex interactions of immune cells and the production of IFN-gamma and other downstream angiostatic chemokines, the mechanisms involved are still poorly defined. Here we show that IL-12 activates an anti-angiogenic program in Con A-activated mouse spleen cells (activated spc) or human PBMC (activated PBMC). The soluble factors they release in its presence arrest the cycle of endothelial cells (EC), inhibit in vitro angiogenesis, negatively modulate the production of matrix metalloproteinase-9, and the ability of EC to adhere to vitronectin and up-regulate ICAM-1 and VCAM-1 expression. These effects do not require direct cell-cell contact, yet result from continuous interaction between activated lymphoid cells and EC. We used neutralizing Abs to show that the IFN-inducible protein-10 and monokine-induced by IFN-gamma chemokines are pivotal in inducing these effects. Experiments with nu/nu mice, nonobese diabetic-SCID mice, or activated spc enriched in specific cell subpopulations demonstrated that CD4(+), CD8(+), and NK cells are all needed to mediate the full anti-angiogenetic effect of IL-12.


Assuntos
Inibidores da Angiogênese/fisiologia , Comunicação Celular/imunologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Interleucina-12/fisiologia , Subpopulações de Linfócitos/imunologia , Neovascularização Fisiológica/imunologia , Inibidores da Angiogênese/metabolismo , Animais , Apoptose/imunologia , Adesão Celular/imunologia , Ciclo Celular/imunologia , Linhagem Celular Transformada , Células Cultivadas , Técnicas de Cocultura , Selectina E/biossíntese , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Inibidores do Crescimento/fisiologia , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Interleucina-12/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária , Subpopulações de Linfócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Baço/citologia , Baço/imunologia , Baço/metabolismo , Molécula 1 de Adesão de Célula Vascular/biossíntese
20.
J Biol Regul Homeost Agents ; 15(4): 351-8, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11860223

RESUMO

As a natural consequence of the expression of the activated transforming rat Her-2/neu oncogene all mammary glands of female transgenic BALB/c (BALB-neuT) mice develop atypical epithelial hyperplasia which progresses to invasive carcinoma. A lobular carcinoma is palpable in all mammary glands of 33-week-old BALB-neuT mice. This progression is markedly delayed by systemic administration of IL-12. In a series of studies the best administration schedule, the lowest dose and the most effective administration time have been defined. The cellular and molecular mechanisms resulting in the delay of carcinogenesis have been established. By means of a series of downstream mediators IL-12 inhibits the angiogenic burst that goes along with the passage from preneoplastic to neoplastic and invasive lesions; it also recruits lymphoid cells in the mammary pad and activates their cytotoxicity towards neoplastic cells and newly formed vessels; and furthermore, it induces lymphoid cells to trigger antiangiogenic activities in neoplastic epithelial cells. Effective, low-dose and non-toxic IL-12 treatments may thus be envisaged as a possible option in the management of preneoplastic mammary lesions and in mammary cancer prevention.


Assuntos
Interleucina-12/metabolismo , Neoplasias Mamárias Animais/metabolismo , Receptor ErbB-2/metabolismo , Animais , Células Cultivadas , Feminino , Humanos , Interferon gama/metabolismo , Neoplasias Mamárias Animais/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Neovascularização Patológica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...