Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
3.
Nat Rev Drug Discov ; 18(3): 219-234, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30679806

RESUMO

Over the past decade, preclinical and clinical research have confirmed the essential role of interferons for effective host immunological responses to malignant cells. Type I interferons (IFNα and IFNß) directly regulate transcription of >100 downstream genes, which results in a myriad of direct (on cancer cells) and indirect (through immune effector cells and vasculature) effects on the tumour. New insights into endogenous and exogenous activation of type I interferons in the tumour and its microenvironment have given impetus to drug discovery and patient evaluation of interferon-directed strategies. When combined with prior observations or with other effective modalities for cancer treatment, modulation of the interferon system could contribute to further reductions in cancer morbidity and mortality. This Review discusses new interferon-directed therapeutic opportunities, ranging from cyclic dinucleotides to genome methylation inhibitors, angiogenesis inhibitors, chemoradiation, complexes with neoantigen-targeted monoclonal antibodies, combinations with other emerging therapeutic interventions and associations of interferon-stimulated gene expression with patient prognosis - all of which are strategies that have or will soon enter translational clinical evaluation.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Fatores Imunológicos/uso terapêutico , Interferon-alfa/uso terapêutico , Interferon beta/uso terapêutico , Neoplasias/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Animais , Humanos , Neoplasias/imunologia
4.
JAMA Oncol ; 3(7): 944-952, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28196207

RESUMO

IMPORTANCE: After identification of activating mutations of the KIT gene in gastrointestinal stromal tumor (GIST)-the most common sarcomaof the gastrointestinal tract-a phase 2 study demonstrated efficacy of imatinib mesylate in patients with metastatic GIST harboring a KIT exon 11 mutation. Initial results of long-term follow-up have found a survival benefit in this subgroup of patients. OBECTIVE: To assess the long-term survival of patients with GIST who were treated in SWOG study S0033 and to present new molecular data regarding treatment outcomes. DESIGN, SETTING, AND PARTICIPANTS: In this follow-up of randomized clinical trial participants (from December 15, 2000, to September 1, 2001), patients were required to have advanced GIST that was not surgically curable. Postprotocol data collection occurred from August 29, 2011, to July 15, 2015. Using modern sequencing technologies, 20 cases originally classified as having wild-type tumors underwent reanalysis. This intergroup study was coordinated by SWOG, a cooperative group member within the National Clinical Trials Network, with participation by member/affiliate institutions. This follow-up was not planned as part of the initial study. INTERVENTIONS: Patients were randomized to 1 of 2 dose levels of imatinib mesylate, including 400 mg once daily (400 mg/d) vs 400 mg twice daily (800 mg/d), and were treated until disease progression or unacceptable toxic effects of the drug occurred. MAIN OUTCOMES AND MEASURES: The primary end point was overall survival. Updated survival data were correlated with clinical and molecular factors, and patterns of postprotocol therapies were enumerated and described in long-term survivors. RESULTS: Of 695 eligible patients (376 men [54.1%]; 319 women [45.9%]; mean [SD] age, 60.1 [14.0] years), 189 survived 8 years or longer, including 95 in the 400-mg/d dose arm and 94 in the 800-mg/d arm. The 10-year estimate of overall survival was 23% (95% CI, 20%-26%). Among 142 long-term survivors, imatinib was the sole therapy administered in 69 (48.6%), with additional systemic agents administered to 54 patients (38.0%). Resequencing studies of 20 cases originally classified as KIT/PDGFRA wild-type GIST revealed that 17 (85.0%) harbored a pathogenic mutation, most commonly a mutation of a subunit of the succinate dehydrogenase complex. CONCLUSIONS AND RELEVANCE: A subset of patients with metastatic GIST experiences durable, long-term overall survival with imatinib treatment. Although this study provides guidance for management of GIST harboring the most common KIT and PDGFRA mutations, optimal management of other genotypic subtypes remains unclear. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00009906.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Gastrointestinais/tratamento farmacológico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Mesilato de Imatinib/uso terapêutico , Proteínas Proto-Oncogênicas c-kit/genética , Idoso , Ensaios Clínicos Fase III como Assunto , Intervalo Livre de Doença , Feminino , Seguimentos , Neoplasias Gastrointestinais/genética , Tumores do Estroma Gastrointestinal/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Modelos de Riscos Proporcionais , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Succinato Desidrogenase/genética , Taxa de Sobrevida , Resultado do Tratamento
6.
Semin Oncol ; 41(2): 156-73, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24787290

RESUMO

Constitutive expression of interferons (IFNs) and activation of their signaling pathways have pivotal roles in host responses to malignant cells in the tumor microenvironment. IFNs are induced by the innate immune system and in tumors through stimulation of Toll-like receptors (TLRs) and through other signaling pathways in response to specific cytokines. Although in the oncologic context IFNs have been thought of more as exogenous pharmaceuticals, the autocrine and paracrine actions of endogenous IFNs probably have even more critical effects on neoplastic disease outcomes. Through high-affinity cell surface receptors, IFNs modulate transcriptional signaling, leading to regulation of more than 2,000 genes with varying patterns of temporal expression. Induction of the gene products by both unphosphorylated and phosphorylated STAT1 after ligand binding results in alterations in tumor cell survival, inhibition of angiogenesis, and augmentation of actions of T, natural killer (NK), and dendritic cells. The interferon-stimulated gene (ISG) signature can be a favorable biomarker of immune response but, in a seemingly paradoxical finding, a specific subset of the full ISG signature indicates an unfavorable response to DNA-damaging interventions such as radiation. IFNs in the tumor microenvironment thus can alter the emergence, progression, and regression of malignancies.


Assuntos
Interferons/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Microambiente Tumoral , Animais , Antineoplásicos/química , Apoptose , Citocinas/metabolismo , DNA/metabolismo , Dano ao DNA , Células Dendríticas/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes p53 , Humanos , Imunidade Inata , Camundongos , Camundongos Transgênicos , Neovascularização Patológica , Fosforilação , Retroviridae/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
7.
Cancer Biol Ther ; 15(8): 975-81, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24842548

RESUMO

Our prior phase I study of the combination of vascular endothelial growth factor (VEGF) antibody, bevacizumab, and VEGF receptor (VEGFR) inhibitor, sunitinib, in advanced solid tumors identified an encouraging response evaluation. An expansion phase of this study was thus undertaken to obtain further safety data, response assessment and characterization of pharmacodynamic biomarkers in melanoma, renal, and adrenal carcinoma patients. Patients with metastatic solid tumors received sunitinib (37.5 mg/d, 4 wk on/2 wk off) and bevacizumab (5 mg/kg intravenously every 2 wk). Responses were assessed every 2 cycles. Serum levels of angiogenic molecules were measured using ELISA assays. Twenty-two patients were enrolled, including 11 melanoma, 5 renal cell carcinoma (RCC), 5 adrenal cancer, and 1 angiosarcoma. Grade 3 or higher adverse events were observed in 15 patients, including hypertension (41%), thrombocytopenia (23%), and fatigue (14%). Three RCC patients, and 1 melanoma patient developed thrombotic microangiopathy (TMA). Partial response (PR) occurred in 21% patients, including melanoma (2), adrenal (1), and renal (1) carcinomas. Overall, 6 patients demonstrated some reduction in their tumor burden. Serum VEGF and several other proangiogenic proteins declined over the first 4 wk of treatment whereas the putative VEGF-resistant protein, prokineticin-2, increased over 10-fold. Occurrence of TMA related to dual VEGF/VEGFR inhibition can result from systemic or nephron specific injury even in non-renal malignancies. While the combination of sunitinib and bevacizumab was clinically efficacious in renal cell carcinoma and melanoma, the observance of microangiopathy, even in non-RCC patients, is a significant toxicity that precludes further clinical development.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Anticorpos Monoclonais Humanizados/administração & dosagem , Bevacizumab , Biomarcadores Farmacológicos/metabolismo , Feminino , Hormônios Gastrointestinais/metabolismo , Humanos , Indóis/administração & dosagem , Masculino , Pessoa de Meia-Idade , Neoplasias/metabolismo , Neuropeptídeos/metabolismo , Pirróis/administração & dosagem , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Sunitinibe , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
J Biol Chem ; 289(12): 8442-9, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24492617

RESUMO

Notch1 is an evolutionarily conserved signaling molecule required for stem cell maintenance that is inappropriately reactivated in several cancers. We have previously shown that melanomas reactivate Notch1 and require its function for growth and survival. However, no Notch1-activating mutations have been observed in melanoma, suggesting the involvement of other activating mechanisms. Notch1 activation requires two cleavage steps: first by a protease and then by γ-secretase, which releases the active intracellular domain (Notch1(NIC)). Interestingly, although ADAM10 and -17 are generally accepted as the proteases responsible of Notch1 cleavage, here we show that MT1-MMP, a membrane-tethered matrix metalloproteinase involved in the pathogenesis of a number of tumors, is a novel protease required for the cleavage of Notch1 in melanoma cells. We find that active Notch1 and MT1-MMP expression correlate significantly in over 70% of melanoma tumors and 80% of melanoma cell lines, whereas such correlation does not exist between Notch1(NIC) and ADAM10 or -17. Modulation of MT1-MMP expression in melanoma cells affects Notch1 cleavage, whereas MT1-MMP expression in ADAM10/17 double knock-out fibroblasts restores the processing of Notch1, indicating that MT1-MMP is sufficient to promote Notch1 activation independently of the canonical proteases. Importantly, we find that MT1-MMP interacts with Notch1 at the cell membrane, supporting a potential direct cleavage mechanism of MT1-MMP on Notch1, and that MT1-MMP-dependent activation of Notch1 sustains melanoma cell growth. Together, the data highlight a novel mechanism of activation of Notch1 in melanoma cells and identify Notch1 as a new MT1-MMP substrate that plays important biological roles in melanoma.


Assuntos
Metaloproteinase 14 da Matriz/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Receptor Notch1/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM17 , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 14 da Matriz/genética , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/genética , Proteínas de Membrana/metabolismo , Camundongos
9.
Clin Cancer Res ; 19(14): 3996-4007, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23589178

RESUMO

PURPOSE: VB-111 is an antiangiogenic agent consisting of a nonreplicating adenovirus vector (Ad-5) with a modified murine pre-proendothelin promoter leading to apoptosis of tumor vasculature by expressing a Fas-chimera transgene in angiogenic endothelial cells. In a phase I dose-escalation study, pharmacokinetics, pharmacodynamics, safety, and efficacy of a single dose of VB-111 in patients with advanced solid tumors were evaluated. EXPERIMENTAL DESIGN: VB-111 was administered as a single i.v. infusion at escalating doses from 1 × 10(10) (cohort 1) to 1 × 10(13) (cohort 7) viral particles (VP) in successive cohorts. Assessments included pharmacokinetic and pharmacodynamic profiles, tumor response, and overall survival. RESULTS: Thirty-three patients were enrolled. VB-111 was safe and well-tolerated; self-limited fever and chills were seen at doses above 3 × 10(11) VPs. Transgene expression was not detected in blood but was detected in an aspirate from a subcutaneous metastasis after treatment. One patient with papillary thyroid carcinoma had a partial response. CONCLUSIONS: VB-111 was safe and well tolerated in patients with advanced metastatic cancer at a single administration of up to 1 × 10(13) VPs. Evidence of transgene expression in tumor tissue and tumor response was observed.


Assuntos
Adenocarcinoma/terapia , Adenoviridae/genética , Neoplasias Colorretais/terapia , Neovascularização Patológica/terapia , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Adenocarcinoma/secundário , Adenoviridae/imunologia , Adulto , Idoso , Proteínas Angiogênicas/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Citocinas/metabolismo , Intervalo Livre de Doença , Feminino , Terapia Genética , Humanos , Masculino , Pessoa de Meia-Idade , Distribuição Tecidual , Resultado do Tratamento
10.
Pigment Cell Melanoma Res ; 26(3): 408-14, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23480537

RESUMO

We recently identified neuregulin-1 (NRG1) as a novel target of Notch1 required in Notch-dependent melanoma growth. ERBB3 and ERBB4, tyrosine kinase receptors specifically activated by NRG1, have been shown to be either elevated in melanoma cell lines and tumors or to be mutated in 20% of melanomas, respectively. While these data support key roles of NRG1 and its receptors in the pathogenesis of melanoma, whether ERBB3 and ERBB4 display redundant or exclusive functions is not known. Here, we show that ERBB3 and ERBB4 inhibition results in distinct outcomes. ERBB3 inhibition ablates the cellular responses to NRG1, results in AKT inactivation and leads to cell growth arrest and apoptotic cell death. In contrast, ERBB4 knockdown mildly affects cell growth, has no effects on cell survival and, importantly, does not alter the responses to NRG1. Finally, we identified ERBB2 as a key coreceptor in NRG1-dependent ERBB3 signaling. ERBB2 forms a complex with ERBB3, and its inhibition recapitulates the phenotypes observed upon ERBB3 ablation. We propose that an NRG1-ERBB3-ERBB2 signaling unit operates in melanoma cells where it promotes growth and survival.


Assuntos
Melanoma/metabolismo , Melanoma/patologia , Neuregulina-1/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Receptores ErbB/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Fosforilação , Multimerização Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-4
11.
Matrix Biol ; 32(2): 123-32, 2013 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-23202046

RESUMO

BACKGROUND: Reversibility of aberrant methylation via pharmacological means is an attractive target for therapies through epigenetic reprogramming. To establish that pharmacologic reversal of methylation could result in functional inhibition of angiogenesis, we undertook in vitro and in vivo studies of thrombospondin-1 (TSP1), a known inhibitor of angiogenesis. TSP1 is methylated in several malignancies, and can inhibit angiogenesis in melanoma xenografts. We analyzed effects of 5-Aza-deoxycytidine (5-Aza-dC) on melanoma cells in vitro to confirm reversal of promoter hypermethylation and restoration of TSP1 expression. We then investigated the effects of TSP1 expression on new blood vessel formation and tumor growth in vivo. Finally, to determine potential for clinical translation, the methylation status of TSP1 promoter regions of nevi and melanoma tissues was investigated. RESULTS: 5-Aza-dC reduced DNA (cytosine-5)-methyltransferase 1 (DNMT1) protein, reversed promoter hypermethylation, and restored TSP1 expression in five melanoma cell lines, while having no effect on TSP1 protein levels in normal human melanocytes. In in vivo neovascularization studies, mice were implanted with melanoma cells (A375) either untreated or treated with 5Aza-dC. Vessels at tumor sites were counted by an observer blinded to treatments and the number of tumor vessels was significantly decreased at pretreated tumor sites. This difference occurred before a significant difference in tumor volumes was seen, yet in further studies the average tumor volume in mice treated in vivo with 5-Aza-dC was decreased by 55% compared to untreated controls. Knockdown of TSP1 expression with shRNA enhanced tumor-induced angiogenesis by 68%. Analyses of promoter methylation status of TSP1 in tumors derived from untreated and treated mice identified 67% of tumors from untreated and 17% of tumors from treated mice with partial methylation consistent with the methylation specific PCR analysis of A375 cells. Examination of methylation patterns in the promoter of TSP1 and comparison of aberrantly methylated TSP1 in melanoma with non-malignant nevi identified a significantly higher frequency of promoter methylation in tumor samples from melanoma patients. CONCLUSIONS: Pharmacological reversal of methylation silenced TSP1 had functional biological consequences in enhancing angiogenesis inhibition and inducing antitumor effects to decrease murine melanoma growth. Angiogenesis inhibition is an additional mechanism by which epigenetic modulators can have antitumor effects.


Assuntos
Azacitidina/análogos & derivados , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Epigênese Genética/genética , Melanoma/genética , Neovascularização Patológica/genética , Animais , Azacitidina/administração & dosagem , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/antagonistas & inibidores , Decitabina , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma/metabolismo , Melanoma/terapia , Metilação/efeitos dos fármacos , Camundongos , Neovascularização Patológica/tratamento farmacológico , Regiões Promotoras Genéticas/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Curr Urol ; 7(2): 65-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24917761

RESUMO

BACKGROUND: Circulating tumor cells (CTC) predict overall survival in patients with metastatic prostate cancer. The objective of this study is to measure CTC before radical prostatectomy in intermediate- and high-risk prostate cancer patients. MATERIALS AND METHODS: The study accrued 12 patients and 10 provided adequate peripheral blood sample. Blood was drawn preoperatively and assayed for CTC using the CellSearch system. Patients were categorized as CTC positive (≥ 1 CTC) or CTC negative (no CTC). RESULTS: Median age was 64.5 years (range 49-77 years), median prostate specific antigen was 7.4 ng/ml (range 5.7-25.7 ng/ml). Seven patients had intermediate-risk and 3 patients had high-risk prostate cancer. One patient was found to be CTC positive. CONCLUSIONS: Our pilot study shows that CTC are rare in patients with clinically localized disease despite intermediate- to high-risk features. CTC may not be the optimal marker to predict prognosis or detect residual disease after radical prostatectomy.

13.
J Transl Med ; 10: 241, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23217102

RESUMO

BACKGROUND: Blood biomarkers are needed to monitor anti-angiogenic treatments for cancer. The association of blood levels of microRNAs (miRs) implicated in angiogenesis with circulating endothelial cells (CEC) and with angiogenic proteins was examined in patients administered drugs with anti-angiogenic activity. METHODS: Blood was collected from patients with uveal melanoma enrolled on an adjuvant therapy trial in which they were treated sequentially with dacarbazine and interferon-alfa-2b. Plasma levels of nine angioregulatory miRs, miR-16, 20a, 106a, 125b, 126, 146a, 155, 199a, and 221, were determined by quantitative real time polymerase chain reaction; CEC, by semi-automated immunomagnetic; and plasma angiogenic proteins, by enzyme linked immunosorbent assays. RESULTS: Levels of miR-199a were positively correlated and miR-106a negatively correlated with CEC pre-therapy. Decreases in miR-126 and miR-199a and increases in miR-16 and miR-106a were observed after interferon-alfa-2b, but not after dacarbazine. CEC also increased after treatment with interferon but not after treatment with dacarbazine. Levels of miRs did not correlate with levels of vascular endothelial growth factor, basic fibroblast growth factor, and interleukin-8. Angiogenic proteins also did not change significantly with treatment. CONCLUSIONS: Blood levels of specific angioregulatory miRs are associated with CEC, and changes in specific angioregulatory miRs parallel increases in CEC after treatment with interferon-alfa-2b. Blood levels of specific angioregulatory miRs are not associated with levels of angiogenic proteins. miRs warrant further evaluation as blood biomarkers of angiogenesis.


Assuntos
Proteínas Angiogênicas/sangue , Movimento Celular , Dacarbazina/uso terapêutico , Células Endoteliais/patologia , Interferon-alfa/uso terapêutico , MicroRNAs/sangue , Movimento Celular/efeitos dos fármacos , Dacarbazina/administração & dosagem , Dacarbazina/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Interferon-alfa/farmacologia , Melanoma/sangue , Melanoma/irrigação sanguínea , Melanoma/tratamento farmacológico , Melanoma/patologia , Neovascularização Patológica/sangue , Neovascularização Patológica/tratamento farmacológico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Neoplasias Uveais/sangue , Neoplasias Uveais/irrigação sanguínea , Neoplasias Uveais/tratamento farmacológico , Neoplasias Uveais/patologia
14.
Nat Rev Drug Discov ; 11(12): 889-90, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23197020

RESUMO

As cancer therapy progresses through the development of molecularly targeted drugs, the focus of Phase I trials needs to shift towards investigating end points that could guide more effective trial design, dosing and choice of tumour setting for Phase II studies.


Assuntos
Antineoplásicos/efeitos adversos , Ensaios Clínicos Fase I como Assunto , Neoplasias/tratamento farmacológico , Humanos , Projetos de Pesquisa
15.
Oncotarget ; 2(12): 1155-64, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22201704

RESUMO

Since sodium stibogluconate (SSG) inhibited phosphatases including SHP-1 and augmented anti-tumor actions of IFN-α2b in vitro and in mice, two Phase I trials of SSG/IFN-α2b combination were undertaken to evaluate safety and target inhibition. Escalating doses of SSG (200-1200 mg/m2) and fixed doses of IFN-α2b (3x106 units/m2) with or without chemotherapy (dacarbazine, vinblastine, cisplatin) were evaluated for side effects and impact on SHP-1 phospho-substrates and IFNα-stimulated-genes (ISGs) in peripheral blood in 40 patients with metastatic melanoma, soft tissue sarcomas, gastrointestinal stromal tumors, and breast or colorectal carcinomas who did not have other established treatment options. Common adverse events were bone marrow suppression, fatigue, gastrointestinal upset, and asymptomatic lipase elevation (n=13); the latter was dose related and mostly after 10d of SSG/IFN-α2b in combination. Levels of SHP-1 substrates (pSTAT1, pSTAT3, pLck and pSlp76) were increased (up to 3x) in peripheral blood cells following SSG with no potentiation by combination with IFN-α2b. Representative ISGs in peripheral blood were induced after IFN-α2b at 4 and 24 hrs with selective modulations by combination. The median time on trials was 2.3 months (10-281d) with no objective regression of disease. Alive at 1y were 17/40 (43%) patients and after 2y were 8/40 (20%) following treatment initiation. These data demonstrate that SSG impacted signal molecules consistent with PTP inhibition and was tolerated in combination with IFN-α2b. Phase II investigations of SSG could safely utilize doses of up to 1200 mg/m2 of SSG for up to 10d alone or in combination with IFN-α2b with or without chemotherapy.


Assuntos
Gluconato de Antimônio e Sódio , Quimioterapia Combinada/efeitos adversos , Quimioterapia Combinada/métodos , Interferon-alfa/uso terapêutico , Adulto , Idoso , Gluconato de Antimônio e Sódio/administração & dosagem , Gluconato de Antimônio e Sódio/efeitos adversos , Gluconato de Antimônio e Sódio/farmacocinética , Gluconato de Antimônio e Sódio/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Cisplatino/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Dacarbazina/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Humanos , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Masculino , Melanoma/tratamento farmacológico , Pessoa de Meia-Idade , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Monoéster Fosfórico Hidrolases/sangue , Proteína Tirosina Fosfatase não Receptora Tipo 6/antagonistas & inibidores , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Sarcoma/tratamento farmacológico , Vimblastina/farmacologia
16.
Expert Opin Biol Ther ; 11(12): 1669-76, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21961496

RESUMO

INTRODUCTION: Antibody, small molecule and protein inhibitors of angiogenesis are used in the management of several cancers. These do not specifically target tumor vascularity, and resistance can be problematic. VB-111 is a vascular-targeting agent consisting of a non-replicating adenovirus vector with a pre-proendothelin-1 promoter that encodes an apoptotic receptor. AREAS COVERED: The rationale and design of VB-111, its mechanism of action, and preclinical studies examining antitumor activity, toxicology and pharmacodynamics are reviewed. Phase I and Phase II clinical trials are also reviewed. EXPERT OPINION: VB-111 is a vascular-targeting gene therapeutic that is both tissue- and condition-specific, with effects limited to endothelial cells undergoing angiogenesis. Systemic administration produces selective destruction of tumor vascularity. Synergistic antitumor activity can be observed when combined with chemotherapy. VB-111 has been found to be safe and well tolerated in a Phase I clinical trial in patients with advanced solid tumors. Phase II clinical trials are in progress. VB-111 is novel agent for cancer that may have application as monotherapy and in combination with other therapies.


Assuntos
Terapia Genética , Neoplasias/terapia , Neovascularização Patológica/prevenção & controle , Adenoviridae/genética , Animais , Apoptose , Endotelina-1/genética , Terapia Genética/efeitos adversos , Vetores Genéticos , Humanos , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Regiões Promotoras Genéticas , Receptores Tipo I de Fatores de Necrose Tumoral/biossíntese , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Resultado do Tratamento , Receptor fas/biossíntese , Receptor fas/genética
17.
Neuro Oncol ; 13(6): 591-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21636707

RESUMO

To assess the accumulation of myeloid-derived suppressor cells (MDSCs) in the peripheral blood of patients with glioma and to define their heterogeneity and their immunosuppressive function. Peripheral blood mononuclear cells (PBMCs) from healthy control subjects and from patients with newly diagnosed glioma were stimulated with anti-CD3/anti-CD28 and T cells assessed for intracellular expression of interferon (IFN)-γ. Antibody staining of PBMCs from glioma patients and healthy donors (CD33, HLADR, CD15, and CD14) followed by 4-color flow cytometry analysis-defined MDSC levels in the peripheral blood. To assess the role of MDSCs in suppressing T cell IFNγ production, PBMCs were depleted of MDSCs using anti-CD33 and anti-CD15 antibody-coated beads prior to T cell stimulation. Enzyme-linked immunosorbent assays were used to assess plasma arginase activity and the level of granulocyte colony-stimulating factor (G-CSF). Patients with glioblastoma have increased MDSC counts (CD33+HLADR-) in their blood that are composed of neutrophilic (CD15(+); >60%), lineage-negative (CD15(-)CD14(-); 31%), and monocytic (CD14(+); 6%) subsets. After stimulation, T cells from patients with glioblastoma had suppressed IFN-γ production when compared with healthy, age-matched donor T cells. Removal of MDSCs from the PBMCs with anti-CD33/CD15-coated beads significantly restored T cell function. Significant increases in arginase activity and G-CSF levels were observed in plasma specimens obtained from patients with glioblastoma. The accumulation of MDSCs in peripheral blood in patients with glioma likely promotes T cell immune suppression that is observed in this patient population. Increased plasma levels of arginase and G-CSF may relate to MDSC suppressor function and MDSC expansion, respectively, in patients with glioma.


Assuntos
Glioblastoma/diagnóstico , Glioblastoma/imunologia , Leucócitos Mononucleares/imunologia , Células Mieloides/imunologia , Células Mieloides/patologia , Linfócitos T/imunologia , Arginase/metabolismo , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/imunologia , Estudos de Casos e Controles , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Glioblastoma/sangue , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Interferon gama/metabolismo , Ativação Linfocitária , Pessoa de Meia-Idade
19.
J Interferon Cytokine Res ; 31(1): 1-4, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21226605

RESUMO

Studies of the action of interferon-stimulated genes (ISGs) and their protein products have resulted in fundamental discoveries relevant to translational control, regulation of RNA stability and editing, and protein transport and turnover. Actions of ISGs will remain critical to improved clinical application of agonists and antagonists of the toll-like receptor and the interferon signaling cascades--now 25 years after the U.S. Food and Drug Administration and worldwide regulatory approval of the pharmaceutical product produced by recombinant DNA technology. Because the antiviral and cellular actions of these several hundred genes (what?) and their protein products are now being functionally (how?) further elucidated but have been comprehensively summarized to only limited extents, we have selected some of the most potently induced ISGs for review in this special issue of the Journal of Interferon & Cytokine Research.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Interferons/uso terapêutico , Animais , Aniversários e Eventos Especiais , Aprovação de Drogas , Humanos , Interferons/genética , Interferons/farmacologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Estados Unidos , United States Food and Drug Administration
20.
J Interferon Cytokine Res ; 31(5): 433-40, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21235385

RESUMO

Interferon (IFN)-ß in preclinical studies, compared to IFN-α2, bound with higher affinity to its receptor, induced to higher levels of IFN-stimulated gene products, induced more apoptosis in melanoma cells, and had antitumor effects against melanoma. A maximally tolerated dose of 12 × 10(6) international units/m(2) after 2 weeks subcutaneously daily with dose escalation to 18 × 10(6) international units/m(2) was thus used in a phase II trial of IFN-ß1a in cutaneous metastatic melanoma (n = 17) and uveal melanoma (n = 4). It resulted in expected but reversible drug-related severe (grade 3) adverse events in 13/21 patients; anorexia and fatigue were mostly of mild or moderate severity and infrequently needed dose reduction. Although a single patient had a sustained regression, overall IFN-ß1a did not have clinical benefit (response rate <10%; median progression-free survival 1.8 months). Effective and potent induction in peripheral blood cells and into serum of products of IFN-stimulated genes such as the pro-apoptotic cytokine, TRAIL, and the immunomodulatory and anti-angiogenic chemokines, CXCL10 and CCL8, confirmed gene regulatory actions. To probe further anti-angiogenic mechanisms, both VEGF-A and CXCL-5 were assessed; compared to before treatment, both proteins decreased. Continued improvements in understanding of antitumor mechanisms will enhance usefulness of IFNs for nodal or distant metastases from melanoma.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Interferon beta/farmacologia , Interferon beta/uso terapêutico , Melanoma/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Perfilação da Expressão Gênica , Humanos , Interferon beta/efeitos adversos , Masculino , Melanoma/genética , Melanoma/mortalidade , Melanoma/secundário , Pessoa de Meia-Idade , Projetos de Pesquisa , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...