Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Med ; 11(1): 183-193, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34796694

RESUMO

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


Assuntos
Amilorida/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cisplatino/uso terapêutico , Esomeprazol/uso terapêutico , Inibidores da Bomba de Prótons/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Amilorida/uso terapêutico , Animais , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Concentração de Íons de Hidrogênio , Camundongos Endogâmicos BALB C , Trocador 1 de Sódio-Hidrogênio/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Macrófagos Associados a Tumor/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
2.
Transl Oncol ; 13(5): 100752, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32302773

RESUMO

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

3.
Cancers (Basel) ; 12(3)2020 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-32197460

RESUMO

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

4.
Curr Med Chem ; 27(25): 4233-4248, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-30182839

RESUMO

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.


Assuntos
Neoplasias , Animais , Células Endoteliais , Humanos , Imunoterapia , Microambiente Tumoral , Fator de Necrose Tumoral alfa
5.
J Allergy Clin Immunol ; 145(1): 368-378.e13, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31194989

RESUMO

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


Assuntos
Amiloidose , Síndromes Periódicas Associadas à Criopirina , Proteína 3 que Contém Domínio de Pirina da Família NLR , Inibidores da Bomba de Prótons/farmacologia , Amiloidose/tratamento farmacológico , Amiloidose/genética , Amiloidose/imunologia , Animais , Síndromes Periódicas Associadas à Criopirina/tratamento farmacológico , Síndromes Periódicas Associadas à Criopirina/genética , Síndromes Periódicas Associadas à Criopirina/imunologia , Síndromes Periódicas Associadas à Criopirina/patologia , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Camundongos , Camundongos Mutantes , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia
6.
Cancers (Basel) ; 11(9)2019 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-31443604

RESUMO

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.

7.
Eur J Nucl Med Mol Imaging ; 46(5): 1184-1196, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30617965

RESUMO

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


Assuntos
Encéfalo/citologia , Encéfalo/metabolismo , Retículo Endoplasmático/metabolismo , Fluordesoxiglucose F18/metabolismo , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Transporte Biológico/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Desidrogenases de Carboidrato/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Metformina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oxirredução/efeitos dos fármacos , Tomografia por Emissão de Pósitrons
8.
Sci Rep ; 8(1): 11075, 2018 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-30038406

RESUMO

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


Assuntos
Antineoplásicos/farmacologia , Calixarenos/farmacologia , Adutos de DNA/metabolismo , Mutagênicos/toxicidade , Porfirinas/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Calixarenos/química , Calixarenos/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Porfirinas/química , Porfirinas/farmacocinética , Distribuição Tecidual/efeitos dos fármacos
9.
Front Immunol ; 9: 2905, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619269

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Imunoterapia/métodos , Neoplasias/terapia , Proteínas Recombinantes de Fusão/farmacologia , Evasão Tumoral/efeitos dos fármacos , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimiorradioterapia/métodos , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Humanos , Interleucina-2/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Resultado do Tratamento , Evasão Tumoral/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
10.
Oncotarget ; 8(40): 67482-67496, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28978047

RESUMO

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

11.
Eur J Immunol ; 47(4): 743-753, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28198545

RESUMO

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


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Neoplasias Colorretais/tratamento farmacológico , Células Dendríticas/imunologia , Quimioterapia Combinada , Fibrossarcoma/tratamento farmacológico , Células Matadoras Naturais/imunologia , Melfalan/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Diferenciação Celular , Linhagem Celular Tumoral , Células Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Citotóxicos/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos
12.
Oncotarget ; 6(35): 37426-42, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26460958

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Melanoma/tratamento farmacológico , Mimetismo Molecular , Neovascularização Patológica , Proteínas Recombinantes de Fusão/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Sindecana-1/antagonistas & inibidores , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Masculino , Melanoma/irrigação sanguínea , Melanoma/imunologia , Melanoma/metabolismo , Melanoma/secundário , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Terapia de Alvo Molecular , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Fenótipo , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Sindecana-1/imunologia , Sindecana-1/metabolismo , Fatores de Tempo , Carga Tumoral , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Antioxid Redox Signal ; 20(7): 1086-97, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23373831

RESUMO

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


Assuntos
Carcinogênese/patologia , Inflamação/patologia , Neoplasias/patologia , Transdução de Sinais/fisiologia , Animais , Antioxidantes/metabolismo , Progressão da Doença , Humanos , Oxirredução , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral/fisiologia
14.
Cancer Med ; 2(4): 478-87, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24156020

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Modelos Animais de Doenças , Esquema de Medicação , Sinergismo Farmacológico , Memória Imunológica , Depleção Linfocítica , Linfócitos do Interstício Tumoral , Melfalan/administração & dosagem , Camundongos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/mortalidade , Proteínas Recombinantes de Fusão/administração & dosagem , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Gencitabina
15.
Eur J Cancer ; 49(8): 2022-33, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23352437

RESUMO

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.


Assuntos
Melanoma/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Anticorpos de Cadeia Única/farmacologia , Sindecana-1/metabolismo , Carga Tumoral/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Células CHO , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Cricetinae , Cricetulus , Feminino , Células HEK293 , Humanos , Melanoma/irrigação sanguínea , Melanoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Dados de Sequência Molecular , Neovascularização Patológica/patologia , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Biblioteca de Peptídeos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Sindecana-1/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Carcinogenesis ; 34(3): 620-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23161574

RESUMO

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


Assuntos
Sistema y+ de Transporte de Aminoácidos/metabolismo , Anticarcinógenos/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Ibuprofeno/farmacologia , Sarcoma Experimental/prevenção & controle , Sulfassalazina/farmacologia , Adulto , Idoso , Sistema y+ de Transporte de Aminoácidos/genética , Animais , Anticarcinógenos/uso terapêutico , Antioxidantes/metabolismo , Transformação Celular Neoplásica/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/uso terapêutico , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Ibuprofeno/uso terapêutico , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Metilcolantreno , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Sarcoma Experimental/induzido quimicamente , Sarcoma Experimental/imunologia , Sarcoma Experimental/metabolismo , Sulfassalazina/uso terapêutico , Adulto Jovem
17.
BMC Biotechnol ; 11: 104, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-22074550

RESUMO

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.


Assuntos
Artrite Experimental/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Fibronectinas/metabolismo , Articulações/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Receptores Tipo II do Fator de Necrose Tumoral/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Células CHO , Cricetinae , Dimerização , Fibronectinas/imunologia , Humanos , Radioisótopos do Iodo/análise , Articulações/efeitos dos fármacos , Articulações/imunologia , Articulações/patologia , Camundongos , Neovascularização Patológica/imunologia , Plasmídeos , Ligação Proteica , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Teratocarcinoma , Transfecção , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Uteroglobina/química , Uteroglobina/genética
18.
Eur J Cancer ; 47(14): 2221-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21605971

RESUMO

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.


Assuntos
Anticorpos Monoclonais/farmacologia , Moléculas de Adesão Celular/metabolismo , Integrinas/metabolismo , Melanoma/metabolismo , Neoplasias Cutâneas/metabolismo , Motivos de Aminoácidos , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Melanoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Neoplasias Experimentais , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Estrutura Terciária de Proteína , Neoplasias Cutâneas/patologia
19.
Clin Cancer Res ; 17(3): 447-58, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21106730

RESUMO

PURPOSE: Targeting the tumor vasculature by vascular disrupting agents (VDAs) has shown therapeutic activity in mouse models. In most cases, however, VDA efficacy is substantially compromised by the inability of these drugs to completely kill tumor cells located at the periphery of the tumor mass. In this study, we investigated anti-tumor effects of L19mTNFα, a fusion protein composed of L19 (scFv), specific for the angiogenesis-associated ED-B containing fibronectin isoform, and murine TNFα, in xenograft models of esophageal cancer. EXPERIMENTAL DESIGN: We evaluated ED-B expression in esophageal cancer samples. Subsequently, we generated subcutaneous xenografts from primary tumors, treated them with the L19mTNFα scFv, and determined effects on tumor vasculature, viability and proliferation, and VEGF expression and infiltration by hematopoietic cells. To overcome tumor resistance, L19mTNFα scFv was combined with vandetanib, a tyrosine kinase inhibitor of VEGF receptor, epidermal growth factor receptor, and RET signaling. RESULTS: ED-B was broadly expressed by esophageal cancer cell lines, as well as xenografts and primary surgical samples of esophageal cancer. Administration of L19mTNFα acutely damaged tumor vasculature and increased necrosis, indicating a VDA-like activity of this immunoconjugate. This event was followed, however, by rapid tumor growth recovery associated with increased expression of VEGF and recruitment of CD11b+Gr1+ myeloid cells into tumors. Combination of L19mTNFα with vandetanib severely impaired vascular functions in tumors, leading to a reduction of cell proliferation and increased necrosis, without apparent signs of toxicity. CONCLUSION: These findings indicate that a combination of vascular damaging agents with anti-angiogenic drugs could represent a promising therapeutic strategy for esophageal cancer.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Esofágicas/tratamento farmacológico , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Anticorpos de Cadeia Única/uso terapêutico , Fator de Necrose Tumoral alfa/uso terapêutico , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fibronectinas/antagonistas & inibidores , Fibronectinas/metabolismo , Humanos , Camundongos , Camundongos SCID , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Antioxid Redox Signal ; 13(8): 1145-55, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20367281

RESUMO

During their differentiation to antibody-secreting plasma cells, B lymphocytes undergo dramatic changes in metabolism, structure, and function. Here we show that this transition entails extensive intra- and extracellular redox changes. Lipopolysaccharide (LPS)-driven activation and differentiation of naïve murine B splenocytes is paralleled by increased production of reactive oxygen species (ROS) from different sources, followed by a strong antioxidant response. This response includes upregulation of thioredoxin and of the cystine transporter xCT, and increased production and extracellular release of nonprotein thiols, mainly glutathione (GSH) and cysteine. Although ROS levels are higher in late-differentiating B cells, an early oxidative step is likely required to start the differentiation program, because inhibition of NADPH oxidase-dependent early ROS production impairs B-cell activation and differentiation. Addition of reducing agents such as 2-ME results in increased IgM secretion per cell, suggesting that the antioxidant response not only is aimed at restoring the redox homeostasis but also plays a functional role. A highly reduced environment coincident with the presence of large ROS-producing cells is observed in histologic sections of spleens from immunized mice, indicating that the redox modifications observed in LPS-induced B-cell differentiation in vitro occur also in vivo during physiologic immune responses.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Diferenciação Celular , Ativação Linfocitária , Modelos Imunológicos , Animais , Antioxidantes/metabolismo , Linfócitos B/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Cultivadas , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/imunologia , Espécies Reativas de Oxigênio/imunologia , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...