Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurooncol ; 140(3): 519-527, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30238350

RESUMO

PURPOSE: A major challenge to developing new therapies for patients with malignant brain tumors is that relatively few small molecule anticancer drugs penetrate the blood-brain barrier (BBB) well enough to provide therapeutically effective concentrations in brain tissue before drug exposure in non-CNS tissues results in unacceptable toxicity. METHODS: KX2-361, a member of a novel family of compounds with Src-kinase and tubulin polymerization inhibitory activity, demonstrates good oral bioavailability and readily crosses the BBB in mice. The objective of this study was to investigate the activity of KX2-361 against human and murine glioma cells and assess its therapeutic effect in a syngeneic orthotopic model of glioblastoma. RESULTS: In addition to reducing the level of Src autophosphorylation in the GL261 murine glioblastoma cell line, KX2-361 binds directly to tubulin and disrupts microtubule architecture in glioma cells maintained in culture. CONCLUSIONS: The drug is active in vivo against orthotopic GL261 gliomas in syngeneic C57BL/6 mice. Long term survival is not observed in mice lacking an adaptive immune system, indicating that KX2-361 works in concert with the host immune system to control tumor growth and promote long-term survival in the GL261 glioma model.


Assuntos
Acetamidas/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Morfolinas/administração & dosagem , Piridinas/administração & dosagem , Moduladores de Tubulina/administração & dosagem , Quinases da Família src/antagonistas & inibidores , Animais , Apoptose , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glioblastoma/tratamento farmacológico , Humanos , Camundongos Endogâmicos C57BL , Fosforilação , Inibidores de Proteínas Quinases/administração & dosagem
2.
Cancer Immunol Immunother ; 65(11): 1339-1352, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27576783

RESUMO

Survivin is an anti-apoptotic protein that is highly expressed in many cancers, including malignant gliomas. Preclinical studies established that the conjugated survivin peptide mimic SurVaxM (SVN53-67/M57-KLH) could stimulate an anti-tumor immune response against murine glioma in vivo, as well as human glioma cells ex vivo. The current clinical study was conducted to test safety, immunogenicity and clinical effects of the vaccine. Recurrent malignant glioma patients whose tumors were survivin-positive, and who had either HLA-A*02 or HLA-A*03 MHC class I allele-positivity, were given subcutaneous injections of SurVaxM (500 µg) in Montanide ISA 51 with sargramostim (100 µg) at 2-week intervals. SurVaxM was well tolerated with mostly grade one adverse events (AE) and no serious adverse events (SAE) attributable to the study drug. Six patients experienced local injection site reactions; three patients reported fatigue (grades 1 and 2), and 2 patients experienced myalgia (grade 1). Six of eight immunologically evaluable patients developed both cellular and humoral immune responses to vaccine. The vaccine also stimulated HLA-A*02, HLA-A*03 and HLA-A*24 restricted T cell responses. Three patients maintained a partial clinical response or stable disease for more than 6 months. Median progression-free survival was 17.6 weeks, and median overall survival was 86.6 weeks from study entry with seven of nine patients surviving more than 12 months.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer/imunologia , Glioma/terapia , Imunoterapia Ativa/métodos , Proteínas Inibidoras de Apoptose/imunologia , Peptídeos/imunologia , Linfócitos T/imunologia , Adulto , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Feminino , Glioma/imunologia , Glioma/mortalidade , Antígeno HLA-A2/metabolismo , Antígeno HLA-A3/metabolismo , Humanos , Imunidade Humoral , Proteínas Inibidoras de Apoptose/genética , Interferon gama/metabolismo , Masculino , Pessoa de Meia-Idade , Peptídeos/genética , Recidiva , Análise de Sobrevida , Survivina , Resultado do Tratamento , Vacinas de Subunidades Antigênicas
3.
Clin Cancer Res ; 30(4): 865-876, 2024 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-38060213

RESUMO

PURPOSE: The abundance and biological contribution of cancer-associated fibroblasts (CAF) in glioblastoma (GBM) are poorly understood. Here, we aim to uncover its molecular signature, cellular roles, and potential tumorigenesis implications. EXPERIMENTAL DESIGN: We first applied single-cell RNA sequencing (RNA-seq) and bioinformatics analysis to identify and characterize stromal cells with CAF transcriptomic features in human GBM tumors. Then, we performed functional enrichment analysis and in vitro assays to investigate their interactions with malignant GBM cells. RESULTS: We found that CAF abundance was low but significantly correlated with tumor grade, poor clinical outcome, and activation of extracellular matrix remodeling using three large cohorts containing bulk RNA-seq data and clinical information. Proteomic analysis of a GBM-derived CAF line and its secretome revealed fibronectin (FN1) as a critical candidate factor mediating CAF functions. This was validated using in vitro cellular models, which demonstrated that CAF-conditioned media and recombinant FN1 could facilitate the migration and invasion of GBM cells. In addition, we showed that CAFs were more abundant in the mesenchymal-like state (or subtype) than in other states of GBMs. Interestingly, cell lines resembling the proneural state responded to the CAF signaling better for the migratory and invasive phenotypes. CONCLUSIONS: Overall, this study characterized the molecular features and functional impacts of CAFs in GBM, alluding to novel cell interactions mediated by CAFs in the GBM microenvironment.


Assuntos
Fibroblastos Associados a Câncer , Glioblastoma , Humanos , Fibroblastos Associados a Câncer/metabolismo , Glioblastoma/patologia , Linhagem Celular Tumoral , Proteômica , Movimento Celular/genética , Microambiente Tumoral/genética , Fibroblastos/metabolismo
4.
J Clin Oncol ; 41(7): 1453-1465, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36521103

RESUMO

PURPOSE: Despite intensive treatment with surgery, radiation therapy, temozolomide (TMZ) chemotherapy, and tumor-treating fields, mortality of newly diagnosed glioblastoma (nGBM) remains very high. SurVaxM is a peptide vaccine conjugate that has been shown to activate the immune system against its target molecule survivin, which is highly expressed by glioblastoma cells. We conducted a phase IIa, open-label, multicenter trial evaluating the safety, immunologic effects, and survival of patients with nGBM receiving SurVaxM plus adjuvant TMZ following surgery and chemoradiation (ClinicalTrials.gov identifier: NCT02455557). METHODS: Sixty-four patients with resected nGBM were enrolled including 38 men and 26 women, in the age range of 20-82 years. Following craniotomy and fractionated radiation therapy with concurrent TMZ, patients received four doses of SurVaxM (500 µg once every 2 weeks) in Montanide ISA-51 plus sargramostim (granulocyte macrophage colony-stimulating factor) subcutaneously. Patients subsequently received adjuvant TMZ and maintenance SurVaxM concurrently until progression. Progression-free survival (PFS) and overall survival (OS) were reported. Immunologic responses to SurVaxM were assessed. RESULTS: SurVaxM plus TMZ was well tolerated with no serious adverse events attributable to SurVaxM. Of the 63 patients who were evaluable for outcome, 60 (95.2%) remained progression-free 6 months after diagnosis (prespecified primary end point). Median PFS was 11.4 months and median OS was 25.9 months measured from first dose of SurVaxM. SurVaxM produced survivin-specific CD8+ T cells and antibody/immunoglobulin G titers. Apparent clinical benefit of SurVaxM was observed in both methylated and unmethylated patients. CONCLUSION: SurVaxM appeared to be safe and well tolerated. The combination represents a promising therapy for nGBM. For patients with nGBM treated in this manner, PFS may be an acceptable surrogate for OS. A large randomized clinical trial of SurVaxM for nGBM is in progress.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Masculino , Humanos , Feminino , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Survivina/uso terapêutico , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Adjuvantes Imunológicos/uso terapêutico , Vacinas de Subunidades Antigênicas/uso terapêutico
5.
Cancer Immunol Immunother ; 59(8): 1211-21, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20422411

RESUMO

Survivin is a tumor-associated antigen with significant potential as a cancer vaccine target. We have identified a survivin peptide mimic containing human MHC class I epitopes and a potential class II ligand that induces a potent antitumor response in C57BL/6 mice with GL261 cerebral gliomas. This peptide is able to elicit both CD8+ CTL and T helper cell responses in C57BL/6 mice. The corresponding region of the human survivin molecule represented by peptide SVN53-67 is 100% homologous to the murine protein, but SVN53-67 is weakly immunogenic in man. We evaluated several amino acid substitutions in putative human MHC I anchor positions in SVN53-67 to identify potential peptide mimics that could provide an enhanced antitumor immune response against human glioma and primary central nervous system lymphoma (PCNSL) cells in culture. We evaluated survivin peptides with predicted binding to human HLA-A*0201 antigen using peptide-loaded dendritic cells from PBMC of patients with these malignancies. One alteration (M57) led to binding to HLA-A*0201 with significantly higher affinity. We compared the ability of autologous dendritic cells loaded with SVN53-67 peptide and SVN53-67/M57 in CTL assays against allomatched and autologous, survivin-expressing, human malignant glioma and PCNSL cells. Both SVN53-67 and SVN53-67/M57 produced CTL-mediated killing of malignant target cells; however, SVN53-67/M57 was significantly more effective than SVN53-67. Thus, SVN53-67/M57 may act as a peptide mimic to induce an enhanced antitumor CTL response in tumor patients. The use of SVN53-67/M57 as a cancer vaccine might have application for cancer vaccine therapy.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/imunologia , Vacinas Anticâncer , Glioma/imunologia , Proteínas Associadas aos Microtúbulos/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Glioma/patologia , Glioma/terapia , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Proteínas Inibidoras de Apoptose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Proteínas Mutantes/imunologia , Fragmentos de Peptídeos/imunologia , Ligação Proteica , Engenharia de Proteínas , Proteínas Repressoras , Homologia de Sequência de Aminoácidos , Survivina , Linfócitos T Citotóxicos/patologia
6.
J Neurooncol ; 94(1): 21-30, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19152070

RESUMO

Human melanoma proteoglycan (HMP), a melanoma-associated antigen, is expressed in both human melanomas and gliomas. We used HMP-specific monoclonal antibody (mAb) VT68.2 to investigate whether murine GL261 cerebral gliomas express the HMP homologue AN2 and to determine whether AN2 could be targeted for selective delivery of radiation in vivo. HMP-specific mAb VT68.2 stained murine GL261 glioma cells grown in culture and intracerebrally in syngeneic C57BL/6 mice. Positron emission tomography with radiolabeled mAb VT68.2 showed high-contrast, positive images of gliomas against a negative background. At 96 h after injection, glioma uptake of radiolabeled mAb VT68.2 was 10x greater than that of the isotype control mAb and 20x greater than that detected in normal cerebral tissue. Our results show murine GL261 cerebral gliomas express AN2 and HMP-specific mAb VT68.2 accumulates selectively and specifically at high concentration and is retained within murine cerebral gliomas. Thus, HMP is a potential target for antibody-mediated selective delivery of radiation to cerebral gliomas in vivo.


Assuntos
Antígenos/metabolismo , Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Proteoglicanas/metabolismo , Animais , Anticorpos Monoclonais , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/radioterapia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Citometria de Fluxo , Glioma/diagnóstico por imagem , Glioma/radioterapia , Humanos , Radioisótopos do Iodo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Transplante de Neoplasias , Tomografia por Emissão de Pósitrons , Fatores de Tempo , Distribuição Tecidual
7.
Clin Cancer Res ; 14(3): 883-91, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18245552

RESUMO

PURPOSE: The purpose of the present study was to evaluate the anti-epidermal growth factor receptor (EGFR) monoclonal antibody (mAb), cetuximab, (IMC-C225) and the anti-EGFRvIII mAb, L8A4, used in combination as delivery agents for boron neutron capture therapy (BNCT) of a rat glioma composed of a mixture of cells expressing either wild-type (F98(EGFR)) or mutant receptors(F98(npEGFRvIII)). EXPERIMENTAL DESIGN: A heavily boronated polyamidoamine dendrimer (BD) was linked by heterobifunctional reagents to produce the boronated mAbs, BD-C225 and BD-L8A4. For in vivo biodistribution and therapy studies, a mixture of tumor cells were implanted intracerebrally into Fischer rats. Biodistribution studies were carried out by administering (125)I-labeled bioconjugates via convection-enhanced delivery (CED), and for therapy studies, nonradiolabeled bioconjugates were used for BNCT. This was carried out 14 days after tumor implantation and 24 h after CED at the Massachusetts Institute of Technology nuclear reactor. RESULTS: Following CED of a mixture of (125)I-BD-C225 and (125)I-BD-L8A4 to rats bearing composite tumors, 61.4% of the injected dose per gram (ID/g) was localized in the tumor compared with 30.8% ID/g for (125)I-BD-L8A4 and 34.7% ID/g for (125)I-BD-C225 alone. The corresponding calculated tumor boron values were 24.4 mug/g for rats that received both mAbs, and 12.3 and 13.8 mug/g, respectively, for BD-L8A4 or BD-C225 alone. The mean survival time of animals bearing composite tumors, which received both mAbs, was 55 days (P < 0.0001) compared with 36 days for BD-L8A4 and 38 days for BD-C225 alone, which were not significantly different from irradiated controls. CONCLUSIONS: Both EGFRvIII and wild-type EGFR tumor cell populations must be targeted using a combination of BD-cetuximab and BD-L8A4. Although in vitro C225 recognized both receptors, in vivo it was incapable of delivering the requisite amount of (10)B for BNCT of EGFRvIII-expressing gliomas.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Boro/análise , Neoplasias Encefálicas/radioterapia , Receptores ErbB/análise , Glioma/radioterapia , Animais , Sítios de Ligação de Anticorpos , Boro/imunologia , Modelos Animais de Doenças
8.
Sci Rep ; 9(1): 4959, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30874569

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

9.
Clin Cancer Res ; 13(4): 1260-8, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17317838

RESUMO

PURPOSE: The purpose of the present study was to evaluate the anti-epidermal growth factor monoclonal antibody (mAb) cetuximab (IMC-C225) as a delivery agent for boron neutron capture therapy (BNCT) of a human epidermal growth factor receptor (EGFR) gene-transfected rat glioma, designated as F98(EGFR). EXPERIMENTAL DESIGN: A heavily boronated polyamidoamine dendrimer was chemically linked to cetuximab by means of the heterobifunctional reagents N-succinimidyl 3-(2-pyridyldithio)-propionate and N-(k-maleimido undecanoic acid)-hydrazide. The bioconjugate, designated as BD-C225, was specifically taken up by F98(EGFR) glioma cells in vitro compared with receptor-negative F98 wild-type cells (41.8 versus 9.1 microg/g). For in vivo biodistribution studies, F98(EGFR) cells were implanted stereotactically into the brains of Fischer rats, and 14 days later, BD-C225 was given intracerebrally by either convection enhanced delivery (CED) or direct intratumoral (i.t.) injection. RESULTS: The amount of boron retained by F98(EGFR) gliomas 24 h following CED or i.t. injection was 77.2 and 50.8 microg/g, respectively, with normal brain and blood boron values <0.05 mug/g. Boron neutron capture therapy was carried out at the Massachusetts Institute of Technology Research Reactor 24 h after CED of BD-C225, either alone or in combination with i.v. boronophenylalanine (BPA). The corresponding mean survival times (MST) were 54.5 and 70.9 days (P = 0.017), respectively, with one long-term survivor (more than 180 days). In contrast, the MSTs of irradiated and untreated controls, respectively, were 30.3 and 26.3 days. In a second study, the combination of BD-C225 and BPA plus sodium borocaptate, given by either i.v. or intracarotid injection, was evaluated and the MSTs were equivalent to that obtained with BD-C225 plus i.v. BPA. CONCLUSIONS: The survival data obtained with BD-C225 are comparable with those recently reported by us using boronated mAb L8A4 as the delivery agent. This mAb recognizes the mutant receptor, EGFRvIII. Taken together, these data convincingly show the therapeutic efficacy of molecular targeting of EGFR using a boronated mAb either alone or in combination with BPA and provide a platform for the future development of combinations of high and low molecular weight delivery agents for BNCT of brain tumors.


Assuntos
Anticorpos Monoclonais/farmacologia , Compostos de Boro/administração & dosagem , Terapia por Captura de Nêutron de Boro/métodos , Receptores ErbB/biossíntese , Glioma/tratamento farmacológico , Glioma/radioterapia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Compostos de Boro/química , Compostos de Boro/farmacocinética , Cetuximab , Terapia Combinada , Dendrímeros/administração & dosagem , Dendrímeros/química , Dendrímeros/farmacocinética , Receptores ErbB/imunologia , Receptores ErbB/metabolismo , Glioma/metabolismo , Humanos , Imunoconjugados/farmacocinética , Imunoconjugados/farmacologia , Imageamento por Ressonância Magnética/métodos , Ratos , Ratos Endogâmicos F344 , Distribuição Tecidual
10.
Clin Cancer Res ; 24(11): 2642-2652, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29540489

RESUMO

Purpose: Survivin is an inhibitor of apoptosis protein (IAP) that is highly expressed in many cancers and represents an attractive molecule for targeted cancer therapy. Although primarily regarded as an intracellular protein with diverse actions, survivin has also been identified in association with circulating tumor exosomes.Experimental Design: We have reported that active, specific vaccination with a long peptide survivin immunogen leads to the development of survivin-specific CD8-mediated tumor cell lysis and prolongation of survival in tumor-bearing mice. In addition to cellular antitumor responses, circulating anti-survivin antibodies are detected in the serum of mice and human glioblastoma patients following vaccination with the survivin immunogen.Results: Here we demonstrate that survivin is present on the outer cell membrane of a wide variety of cancer cell types, including both murine and human glioma cells. In addition, antibodies to survivin that are derived from the immunogen display antitumor activity against murine GL261 gliomas in both flank and intracranial tumor models and against B16 melanoma as well.Conclusions: In addition to immunogen-induced, CD8-mediated tumor cell lysis, antibodies to the survivin immunogen have antitumor activity in vivo Cell-surface survivin could provide a specific target for antibody-mediated tumor immunotherapeutic approaches. Clin Cancer Res; 24(11); 2642-52. ©2018 AACR.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/farmacologia , Biomarcadores Tumorais , Membrana Celular/metabolismo , Survivina/antagonistas & inibidores , Animais , Afinidade de Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Modelos Animais de Doenças , Expressão Gênica , Humanos , Masculino , Melanoma Experimental , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Camundongos , Peptídeos/antagonistas & inibidores , Peptídeos/imunologia , Proteínas Recombinantes de Fusão , Survivina/química , Survivina/genética , Survivina/metabolismo
11.
Sci Rep ; 8(1): 12905, 2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150674

RESUMO

Local acidification of stroma is proposed to favour pre-metastatic niche formation but the mechanism of initiation is unclear. We investigated whether Human Melanoma-derived exosomes (HMEX) could reprogram human adult dermal fibroblasts (HADF) and cause extracellular acidification. HMEX were isolated from supernatants of six melanoma cell lines (3 BRAF V600E mutant cell lines and 3 BRAF wild-type cell lines) using ultracentrifugation or Size Exclusion Chromatography (SEC). Rapid uptake of exosomes by HADF was demonstrated following 18 hours co-incubation. Exposure of HDAF to HMEX leads to an increase in aerobic glycolysis and decrease in oxidative phosphorylation (OXPHOS) in HADF, consequently increasing extracellular acidification. Using a novel immuno-biochip, exosomal miR-155 and miR-210 were detected in HMEX. These miRNAs were present in HMEX from all six melanoma cell lines and were instrumental in promoting glycolysis and inhibiting OXPHOS in tumour cells. Inhibition of miR-155 and miR-210 activity by transfection of miRNA inhibitors into HMEX reversed the exosome-induced metabolic reprogramming of HADF. The data indicate that melanoma-derived exosomes modulate stromal cell metabolism and may contribute to the creation of a pre-metastatic niche that promotes the development of metastasis.


Assuntos
Reprogramação Celular/fisiologia , Exossomos/metabolismo , Melanoma/metabolismo , MicroRNAs/metabolismo , Aerobiose/genética , Aerobiose/fisiologia , Linhagem Celular Tumoral , Reprogramação Celular/genética , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Glicólise/genética , Glicólise/fisiologia , Humanos , Melanoma/genética , MicroRNAs/genética , Microambiente Tumoral/genética , Microambiente Tumoral/fisiologia
12.
Clin Cancer Res ; 12(12): 3792-802, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778107

RESUMO

PURPOSE: The purpose of the present study was to evaluate a boronated EGFRvIII-specific monoclonal antibody, L8A4, for boron neutron capture therapy (BNCT) of the receptor-positive rat glioma, F98(npEGFRvIII). EXPERIMENTAL DESIGN: A heavily boronated polyamido amine (PAMAM) dendrimer (BD) was chemically linked to L8A4 by two heterobifunctional reagents, N-succinimidyl 3-(2-pyridyldithio)propionate and N-(k-maleimidoundecanoic acid)hydrazide. For in vivo studies, F98 wild-type receptor-negative or EGFRvIII human gene-transfected receptor-positive F98(npEGFRvIII) glioma cells were implanted i.c. into the brains of Fischer rats. Biodistribution studies were initiated 14 days later. Animals received [(125)I]BD-L8A4 by either convection enhanced delivery (CED) or direct i.t. injection and were euthanized 6, 12, 24, or 48 hours later. RESULTS: At 6 hours, equivalent amounts of the bioconjugate were detected in receptor-positive and receptor-negative tumors, but by 24 hours the amounts retained by receptor-positive gliomas were 60.1% following CED and 43.7% following i.t. injection compared with 14.6% ID/g by receptor-negative tumors. Boron concentrations in normal brain, blood, liver, kidneys, and spleen all were at nondetectable levels (<0.5 microg/g) at the corresponding times. Based on these favorable biodistribution data, BNCT studies were initiated at the Massachusetts Institute of Technology Research Reactor-II. Rats received BD-L8A4 ( approximately 40 microg (10)B/ approximately 750 mug protein) by CED either alone or in combination with i.v. boronophenylalanine (BPA; 500 mg/kg). BNCT was carried out 24 hours after administration of the bioconjugate and 2.5 hours after i.v. injection of BPA for those animals that received both agents. Rats that received BD-L8A4 by CED in combination with i.v. BPA had a mean +/- SE survival time of 85.5 +/- 15.5 days with 20% long-term survivors (>6 months) and those that received BD-L8A4 alone had a mean +/- SE survival time of 70.4 +/- 11.1 days with 10% long-term survivors compared with 40.1 +/- 2.2 days for i.v. BPA and 30.3 +/- 1.6 and 26.3 +/- 1.1 days for irradiated and untreated controls, respectively. CONCLUSIONS: These data convincingly show the therapeutic efficacy of molecular targeting of EGFRvIII using either boronated monoclonal antibody L8A4 alone or in combination with BPA and should provide a platform for the future development of combinations of high and low molecular weight delivery agents for BNCT of brain tumors.


Assuntos
Receptores ErbB/análise , Glioma/radioterapia , Animais , Anticorpos Monoclonais/uso terapêutico , Compostos de Boro/uso terapêutico , Terapia por Captura de Nêutron de Boro , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioma/patologia , Humanos , Injeções , Radioisótopos do Iodo/farmacocinética , Radioisótopos do Iodo/uso terapêutico , Ratos , Proteínas Recombinantes/metabolismo , Transfecção
13.
Curr Genomics ; 8(3): 163-70, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-18645600

RESUMO

The epidermal growth factor receptor (EGFR) is a membrane-bound, 170 kDa, protein tyrosine kinase that plays an important role in tumorigenesis. The EGFR gene, which is composed of over 168 kb of sequence, including a 123-kb first intron, is frequently amplified and rearranged in malignant gliomas leading to the expression of oncogenic deletion (DM) and tandem duplication (TDM) mutants. The most common DM in gliomas is EGFRvIII, which arises from recombination between introns 1 and 7 with deletion of exons 2 through 7 and intervening introns. In addition, some human gliomas express 180- to 190-kDa TDM, which are constitutively active and highly oncogenic. Both DM and TDM arise by recombination of introns that contain sequences with homology to the recombination signal sequence (RSS) heptamers and nonamers present in the V(D)J region of the immunoglobin and T lymphocyte antigen receptor genes. V(D)J RSS have also been identified in certain proto-oncogenes like bcl-2 that are involved in translocations associated with the development of human lymphomas and in other genes such as hypoxanthine-guainine phosphoribosyl transferase (HPRT) in which deletion mutations and intron rearrangements are a common phenomenon. Together with the expression of recombination associated gene (RAG) and nonhomologous end-joining (NHEJ) proteins in gliomas, these observation suggest that aberrant activity of the V(D)J recombinase may be involved in the activation of proto-oncogenes in both liquid and solid tumors.

14.
Oncotarget ; 8(70): 114722-114735, 2017 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-29383115

RESUMO

Glioma cells release exosomes in culture and into the extracellular matrix in vivo. These nanobodies transport an array of biomolecules and are capable of mediating cell-cell communication. Circulating exosomes in cancer patients may be indicative of disease status and response to therapy. The inhibitor of apoptosis protein (IAP) survivin (SVN) promotes cancer cell proliferation, local immune suppression and resistance to chemotherapy and it is a potential cancer biomarker. We used imaging flow cytometry to perform quantitative measurements of circulating SVN+ exosomes in the serum of malignant glioma patients undergoing investigational treatment with an anti-survivin vaccine (SurVaxM). Serum from glioma patients contained abundant CD9+ exosomes with both SVN and glial fibrillary acidic protein (GFAP) on their surface. Survivin and GFAP were evaluated both independently and together as possible tumor markers on CD9+ exosomes. Patients with longer time to tumor progression generally exhibited a decrease in circulating CD9+/SVN+ and CD9+/GFAP+/SVN+ exosomes immediately following survivin vaccination; whereas, those with early tumor progression had an increase in exosomes, despite anti-survivin immunotherapy. Serum from non-cancer healthy control individuals had very few detectable CD9+/GFAP+/SVN+ exosomes, although CD9+/GFAP+ exosomes were detectable in small numbers. This study demonstrates that patients with malignant gliomas have CD9+/GFAP+/SVN+ and CD9+/SVN+ exosomes that are released into the circulation and that early reductions in their numbers following anti-survivin immunotherapy might be associated with longer progression-free survival.

15.
Clin Cancer Res ; 11(1): 341-50, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15671565

RESUMO

PURPOSE: The goals of the present study were 2-fold: (a) to develop and characterize a rat brain tumor model that could be used for studies of molecular targeting of EGFRvIII and (b) to study the tumor localizing properties of radiolabeled monoclonal antibody (mAb) L8A4, specifically directed against EGFRvIII, following systemic, i.t., and convection enhanced delivery to brain tumor-bearing rats. EXPERIMENTAL DESIGN AND RESULTS: F98 wild-type (F98WT) rat glioma cells were transfected with a gene encoding human EGFRvIII, and following selection and cloning, a cell line, designated F98(npEGFRvIII), was identified, which expressed a nonconstitutively phosphorylated form of the receptor. As determined by a radioligand binding assay, there were 1.2 x 10(5) EGFRvIII sites per cell compared with an undetectable number on F98WT cells. The tumorigenicity of the F98(npEGFRvIII) glioma was studied following i.c. implantation of 10(3), 10(4), or 10(5) cells into CD-Fischer rats. Mean survival times were 23, 17, and 13 days, respectively, which were equivalent to those obtained with F98EGFR and F98WT cells. As determined by magnetic resonance imaging, the mean doubling times for the F98WT and F98(npEGFRvIII) gliomas were similar (59.8 +/- 4.8 versus 52 +/- 3.3 hours). Following i.v. administration to glioma-bearing rats, mAb L8A4 specifically targeted the F98(npEGFRvIII) glioma, and at 24 hours, 7.7% of the injected dose per gram (ID/g) localized in the tumor. This increased 5-fold to 39.5% ID/g following i.t. injection and 7-fold to 59.8% ID/g at 24 hours following convection enhanced delivery. CONCLUSIONS: Based on these data, we have concluded that the F98(npEGFRvIII) glioma should be a valuable animal model for therapy studies focusing on molecular targeting of EGFRvIII by receptor specific mAbs.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias Encefálicas/metabolismo , Modelos Animais de Doenças , Receptores ErbB/biossíntese , Animais , Anticorpos Monoclonais/química , Western Blotting , Encéfalo/metabolismo , Linhagem Celular Tumoral , Separação Celular , Clonagem Molecular , Relação Dose-Resposta a Droga , Receptores ErbB/metabolismo , Citometria de Fluxo , Vetores Genéticos , Glioblastoma/patologia , Glioma/metabolismo , Humanos , Imuno-Histoquímica , Cinética , Imageamento por Ressonância Magnética , Fosforilação , RNA/metabolismo , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo , Transfecção
16.
Cancer Res ; 62(22): 6552-8, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12438250

RESUMO

Convection enhanced delivery (CED) is potentially a powerful method to improvethe targeting of macromolecules to the central nervous system by applying a pressure gradient to establish bulk flow through the brain interstitium during infusion. The purpose of the present study was to evaluate CED as a means to improve the intracerebral and intratumoral (i.t.) uptake of a heavily boronated macromolecule (dendrimer; BD) linked to epidermal growth factor (EGF) for neutron capture therapy in rats bearing a syngeneic epidermal growth factor receptor (EGFR) + glioma. Boronated EGF was radiolabeled with 125I and administered by CED at a rate of 0.33 micro l/min for 15, 30, and 60 min [infusion volumes (V(I)) of 5, 10, and 20 micro l, respectively], using a syringe pump connected to an indwelling cannula implanted into the right caudate nucleus of normal rats or i.t. in rats bearing either F98(EGFR) or F98 wild-type (F98(WT)) gliomas. After infusion, rats were euthanized, and their brains were removed and serially sectioned. The uptake and biodistribution of (125)I-boronated EGF in tumor or brain was studied by quantitative autoradiography and gamma-scintillation counting. The volume of distribution (V(d)) in brain was assessed using a computer interfaced image analysis system. After CED, the V(d) increased from 34.4 to 123.5 micro l with corresponding V(i) ranging from 5 to 20 micro l. The V(d) of BD-EGF in the brain was 64.8 +/- 13.4 micro l with CED (V(i) 10 micro ), and the V(d):V(i) ratio was 6.5 compared with a V(d) of 9.4 +/- 1.6 micro l and a V(d):V(i) ratio of 0.9 after direct intracerebral injection. As determined by quantitative autoradiography and gamma-scintillation counting at 24 h after CED, 47.4% of the injected dose per gram tissue (%ID/g) was localized in F98(EGFR) gliomas compared with 33.2%ID/g after direct i.t. injection and 12.3%ID/g in F98(WT) gliomas. On the basis of these observations, we have concluded that CED is more effective than i.t. injection as a way to deliver boronated EGF to EGFR (+) gliomas for boron neutron capture therapy.


Assuntos
Boranos/farmacocinética , Terapia por Captura de Nêutron de Boro/métodos , Neoplasias Encefálicas/metabolismo , Fator de Crescimento Epidérmico/farmacocinética , Receptores ErbB/metabolismo , Glioma/metabolismo , Animais , Autorradiografia , Boranos/administração & dosagem , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/radioterapia , Dendrímeros , Fator de Crescimento Epidérmico/administração & dosagem , Glioma/diagnóstico por imagem , Glioma/radioterapia , Radioisótopos do Iodo , Poliaminas/administração & dosagem , Poliaminas/farmacocinética , Cintilografia , Ratos , Ratos Endogâmicos F344 , Distribuição Tecidual
17.
Immunotherapy ; 8(11): 1293-1308, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27993092

RESUMO

Glioblastoma is the most common primary brain cancer. Aggressive treatment with surgery, radiation therapy and chemotherapy provides limited overall survival benefit. Glioblastomas have a formidable tumor microenvironment that is hostile to immunological effector cells and these cancers produce profound systemic immunosuppression. However, surgical resection of these tumors creates conditions that favor the use of immunotherapeutic strategies. Therefore, extensive surgical resection, when feasible, will remain part of the equation to provide an environment in which active specific immunotherapy has the greatest chance of working. Toward that end, a number of vaccination protocols are under investigation. Vaccines studied to date have produced cellular and humoral antitumor responses, but unequivocal clinical efficacy has yet to be demonstrated. In addition, focus is shifting toward the prospect of therapies involving vaccines in combination with immune checkpoint inhibitors and other immunomodulatory agents so that effector cells remain active against their targets systemically and within the tumor microenvironment.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Vacinas Anticâncer/imunologia , Receptores Coestimuladores e Inibidores de Linfócitos T/imunologia , Glioblastoma/terapia , Imunoterapia/métodos , Animais , Glioblastoma/imunologia , Humanos , Imunomodulação , Terapia de Imunossupressão , Microambiente Tumoral
18.
Oncotarget ; 6(24): 20043-57, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26343374

RESUMO

Follicular T helper (Tfh) cells are recognized by the expression of CXCR5 and the transcriptional regulator Bcl-6. Tfh cells control B cell maturation and antibody production, and if deregulated, may lead to autoimmunity. Here, we study the role of the proto-oncogene survivin in the formation of Tfh cells. We show that blood Tfh cells of patients with the autoimmune condition rheumatoid arthritis, have intracellular expression of survivin. Survivin was co-localized with Bcl-6 in the nuclei of CXCR5+CD4 lymphocytes and was immunoprecipitated with the Bcl-6 responsive element of the target genes. Inhibition of survivin in arthritic mice led to the reduction of CXCR5+ Tfh cells and to low production of autoantibodies. Exposure to survivin activated STAT3 and induced enrichment of PD-1+Bcl-6+ subset within Tfh cells. Collectively, our study demonstrates that survivin belongs to the Tfh cell phenotype and ensures their optimal function by regulating transcriptional activity of Bcl-6.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Proteínas de Ligação a DNA/imunologia , Proteínas Inibidoras de Apoptose/imunologia , Animais , Autoimunidade/imunologia , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genes p53 , Humanos , Proteínas Inibidoras de Apoptose/química , Proteínas Inibidoras de Apoptose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Modelos Moleculares , Fator 1 de Ligação ao Domínio I Regulador Positivo , Estrutura Secundária de Proteína , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-bcl-6 , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Survivina
19.
Cancer Lett ; 213(1): 57-65, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15312684

RESUMO

2-Methoxyestradiol (2ME), a metabolite of estradiol (E), inhibits proliferation of various tumor cells. In this study we determined the effect of 2ME on human glioblastoma cell lines, in vitro. We compared these cells with cultured astrocytes obtained from traumatized adult rat striatum. Exposure to 2ME had a strong antiproliferative effect on human glioblastoma and caused an increase in the population of apoptotic cells, detected by flow cytometry, in some of the investigated cell lines. A significant number of cells were blocked in the G2/M phase of the cell cycle. Concurrently, the population of cells in the G1 phase decreased in all glioblastoma cell lines. Staining with Hoechst 33258 revealed abnormal nuclear morphology in the proliferating cells treated with 2ME. Treatment with 2ME induced upregulation of wild type p53 in one of the human glioblastoma cell lines as well as in proliferating adult rat astrocytes. We conclude that 2ME inhibits the growth of human glioblastoma cell lines and induces apoptosis, in vitro. This compound deserves further investigation as a treatment for gliomas.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Glioblastoma/patologia , 2-Metoxiestradiol , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/veterinária , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/veterinária , Humanos , Neoplasias Experimentais , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese , Regulação para Cima
20.
Cancer Control ; 11(3): 181-91, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15153842

RESUMO

BACKGROUND: Despite advances in surgery, radiation therapy, and chemotherapy, only modest improvement has been achieved in the survival of patients with malignant gliomas. METHODS: The authors review the immunologic aspects of gliomas, potential targets for therapy, and issues surrounding current immunotherapeutic strategies directed against malignant gliomas. RESULTS: The blood-brain barrier and the purported immunological privilege of the brain are not necessarily insurmountable obstacles to effective immunotherapy for brain tumors. Preclinical studies suggest a number of potential therapeutic avenues. Translational studies offer the prospect of providing substantial new information about immunological trafficking in the nervous system and suggesting the most fruitful approaches to immunotherapy for malignant gliomas. CONCLUSIONS: More effective adjuvant treatments for malignant gliomas are needed. The applicability of immunological approaches in the treatment of these tumors warrants continued study.


Assuntos
Glioma/imunologia , Glioma/terapia , Imunoterapia/métodos , Animais , Antígenos de Neoplasias/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Encéfalo/imunologia , Vacinas Anticâncer/uso terapêutico , Ensaios Clínicos como Assunto , Células Dendríticas/transplante , Sistemas de Liberação de Medicamentos/métodos , Genes erbB-1 , Glioma/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Tolerância Imunológica , Interleucina-2/uso terapêutico , Mutação , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA