Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Transl Res ; 217: 11-22, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31954097

RESUMO

Metabolic remodeling contributes to the development and progression of some cancers and exposes them to vulnerabilities, including specific nutrient dependencies that can be targeted therapeutically. Arginine is a semiessential amino acid, and several cancers are unable to endogenously synthesize sufficient levels of arginine for survival and proliferation, most commonly due to reduced expression of argininosuccinate synthase (ASS1). Such cancers are dependent on arginine and they can be targeted via enzyme-mediated depletion of systemic arginine. We report the preclinical safety, antitumor efficacy, and immune-potentiating effects of pegzilarginase, a highly potent human arginine-degrading enzyme. Toxicology studies showed that pegzilarginase-mediated arginine depletion is well tolerated at therapeutic levels that elicit an antitumor growth effect. To determine which tumor types are best suited for clinical development, we profiled clinical tumor samples for ASS1 expression, which correlated with pegzilarginase sensitivity in vivo in patient-derived xenograft (PDx) models. Among the histologies tested, malignant melanoma, small cell lung cancer and Merkel cell carcinoma had the highest prevalence of low ASS1 expression, the highest proportion of PDx models responding to pegzilarginase, and the strongest correlation between low or no ASS1 expression and sensitivity to pegzilarginase. In an immune-competent syngeneic mouse model, pegzilarginase slowed tumor growth and promoted the recruitment of CD8+ tumor infiltrating lymphocytes. This is consistent with the known autophagy-inducing effects of arginine depletion, and the link between autophagy and major histocompatibility complex antigen presentation to T cells. Our work supports the ongoing clinical investigations of pegzilarginase in solid tumors and clinical combination of pegzilarginase with immune checkpoint inhibitors.


Assuntos
Antineoplásicos/farmacologia , Arginase/farmacologia , Animais , Arginase/análise , Arginase/toxicidade , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/enzimologia , Proteínas Recombinantes/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Nat Med ; 23(1): 120-127, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27869804

RESUMO

Cancer cells experience higher oxidative stress from reactive oxygen species (ROS) than do non-malignant cells because of genetic alterations and abnormal growth; as a result, maintenance of the antioxidant glutathione (GSH) is essential for their survival and proliferation. Under conditions of elevated ROS, endogenous L-cysteine (L-Cys) production is insufficient for GSH synthesis. This necessitates uptake of L-Cys that is predominantly in its disulfide form, L-cystine (CSSC), via the xCT(-) transporter. We show that administration of an engineered and pharmacologically optimized human cyst(e)inase enzyme mediates sustained depletion of the extracellular L-Cys and CSSC pool in mice and non-human primates. Treatment with this enzyme selectively causes cell cycle arrest and death in cancer cells due to depletion of intracellular GSH and ensuing elevated ROS; yet this treatment results in no apparent toxicities in mice even after months of continuous treatment. Cyst(e)inase suppressed the growth of prostate carcinoma allografts, reduced tumor growth in both prostate and breast cancer xenografts and doubled the median survival time of TCL1-Tg:p53-/- mice, which develop disease resembling human chronic lymphocytic leukemia. It was observed that enzyme-mediated depletion of the serum L-Cys and CSSC pool suppresses the growth of multiple tumors, yet is very well tolerated for prolonged periods, suggesting that cyst(e)inase represents a safe and effective therapeutic modality for inactivating antioxidant cellular responses in a wide range of malignancies.


Assuntos
Neoplasias da Mama/metabolismo , Cistationina gama-Liase/farmacologia , Cisteína/efeitos dos fármacos , Cistina/efeitos dos fármacos , Leucemia Linfocítica Crônica de Células B/metabolismo , Polietilenoglicóis/farmacologia , Neoplasias da Próstata/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Cisteína/metabolismo , Cistina/metabolismo , Feminino , Glutationa/metabolismo , Humanos , Leucemia Linfocítica Crônica de Células B/genética , Macaca fascicularis , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transplante de Neoplasias , Estresse Oxidativo , Proteína Supressora de Tumor p53/genética
3.
Hum Mol Genet ; 24(22): 6417-27, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26358771

RESUMO

Arginase deficiency is caused by deficiency of arginase 1 (ARG1), a urea cycle enzyme that converts arginine to ornithine. Clinical features of arginase deficiency include elevated plasma arginine levels, spastic diplegia, intellectual disability, seizures and growth deficiency. Unlike other urea cycle disorders, recurrent hyperammonemia is typically less severe in this disorder. Normalization of plasma arginine levels is the consensus treatment goal, because elevations of arginine and its metabolites are suspected to contribute to the neurologic features. Using data from patients enrolled in a natural history study conducted by the Urea Cycle Disorders Consortium, we found that 97% of plasma arginine levels in subjects with arginase deficiency were above the normal range despite conventional treatment. Recently, arginine-degrading enzymes have been used to deplete arginine as a therapeutic strategy in cancer. We tested whether one of these enzymes, a pegylated human recombinant arginase 1 (AEB1102), reduces plasma arginine in murine models of arginase deficiency. In neonatal and adult mice with arginase deficiency, AEB1102 reduced the plasma arginine after single and repeated doses. However, survival did not improve likely, because this pegylated enzyme does not enter hepatocytes and does not improve hyperammonemia that accounts for lethality. Although murine models required dosing every 48 h, studies in cynomolgus monkeys indicate that less frequent dosing may be possible in patients. Given that elevated plasma arginine rather than hyperammonemia is the major treatment challenge, we propose that AEB1102 may have therapeutic potential as an arginine-reducing agent in patients with arginase deficiency.


Assuntos
Arginase/uso terapêutico , Arginina/sangue , Hiperargininemia/tratamento farmacológico , Animais , Arginase/sangue , Arginase/genética , Arginina/metabolismo , Encéfalo/metabolismo , Criança , Pré-Escolar , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Humanos , Hiperamonemia/sangue , Hiperamonemia/metabolismo , Hiperargininemia/sangue , Hiperargininemia/genética , Hiperargininemia/metabolismo , Estudos Longitudinais , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/uso terapêutico , Convulsões/sangue , Convulsões/metabolismo
5.
PLoS One ; 7(11): e50630, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23189208

RESUMO

OBJECTIVES: Glucagon-like peptide 2 (GLP2) is an intestinal growth factor that has been shown to stimulate intestinal growth and reduce disease severity in preclinical models of short bowel syndrome and inflammatory bowel disease. Teduglutide, a recombinant human GLP2 variant (GLP2-2G), has increased half-life and stability as compared to the native GLP2 peptide, but still requires twice daily dosing in preclinical models and daily dosing in the clinic. The goal of this study was to produce and characterize the preclinical pharmacokinetic and therapeutic properties of GLP2-2G-XTEN, a novel, long-acting form of GLP2-2G. METHODOLOGY AND RESULTS: A GLP2-2G-XTEN fusion protein with extended exposure profile was produced by genetic fusion of GLP2-2G peptide to XTEN, a long, unstructured, non-repetitive, hydrophilic sequence of amino acids. The serum half-life of GLP2-2G-XTEN in mice, rats and monkeys was 34, 38 and 120 hours, respectively. Intestinotrophic effects were demonstrated in normal rats, where GLP2-2G-XTEN administration resulted in a significant increase in both small intestine weight and length. Efficacy of the GLP2-2G-XTEN protein was compared to that of GLP2-2G peptide in a rat Crohn's disease model, indomethacin-induced inflammation. Prophylactic administration of GLP2-2G-XTEN significantly increased the length, reduced the number of trans-ulcerations and adhesions, and reduced the TNFα content of the small intestine. GLP2-2G-XTEN demonstrated greater in vivo potency as compared to GLP2-2G peptide, and improvement in histopathology supported the GLP2-2G-XTEN treatment effects. CONCLUSIONS AND SIGNIFICANCE: GLP2-2G-XTEN is intestinotrophic and demonstrates efficacy in a rat Crohn's disease model requiring a lower molar dose and less frequent dosing relative to GLP2-2G peptide. Allometric scaling based on pharmacokinetics from mouse, rat and monkey projects a human half-life of 240 hours. These improvements in preclinical pharmacokinetics and dosing indicate that GLP2-2G-XTEN may offer a superior therapeutic benefit for treatment of gastrointestinal diseases including Crohn's disease.


Assuntos
Doença de Crohn/tratamento farmacológico , Peptídeo 2 Semelhante ao Glucagon/uso terapêutico , Animais , Doença de Crohn/sangue , Doença de Crohn/induzido quimicamente , Modelos Animais de Doenças , Feminino , Peptídeo 2 Semelhante ao Glucagon/química , Peptídeo 2 Semelhante ao Glucagon/farmacocinética , Humanos , Indometacina , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Macaca fascicularis , Masculino , Camundongos , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico
6.
J Pharm Sci ; 101(8): 2744-54, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22678811

RESUMO

A novel recombinant human growth hormone (rhGH) fusion protein (VRS-317) was designed to minimize receptor-mediated clearance through a reduction in receptor binding without mutations to rhGH by genetically fusing with XTEN amino acid sequences to the N-terminus and the C-terminus of the native hGH sequence. Although in vitro potency of VRS-317 was reduced approximately 12-fold compared with rhGH, in vivo potency was increased because of the greatly prolonged exposure to the target tissues and organs. VRS-317 was threefold more potent than daily rhGH in hypophysectomized rats and fivefold more potent than daily rhGH in juvenile monkeys. In juvenile monkeys, a monthly dose of 1.4 mg/kg VRS-317 (equivalent to 0.26 mg/kg rhGH) caused a sustained pharmacodynamic response for 1 month equivalent to 0.05 mg/kg/day rhGH (1.4 mg/kg rhGH total over 28 days). In monkeys, VRS-317, having a terminal elimination half-life of approximately 110 h, was rapidly and near-completely absorbed, and was well tolerated with no observed adverse effects after every alternate week subcutaneous dosing for 14 weeks. VRS-317 also did not cause lipoatrophy in pig and monkey studies. VRS-317 is currently being studied in GH-deficient patients to confirm the observations in these animal studies.


Assuntos
Hormônio do Crescimento Humano/farmacologia , Hormônio do Crescimento Humano/farmacocinética , Animais , Linhagem Celular , Clonagem Molecular , Feminino , Expressão Gênica , Meia-Vida , Haplorrinos , Hormônio do Crescimento Humano/efeitos adversos , Hormônio do Crescimento Humano/genética , Humanos , Masculino , Ratos , Receptores da Somatotropina/metabolismo , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Suínos
7.
Clin Cancer Res ; 13(7): 2216-25, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17404106

RESUMO

PURPOSE: The antitumor activities and pharmacokinetics of the hypoxia-activated cytotoxin AQ4N and its metabolites were assessed in several preclinical models of pancreatic cancers. EXPERIMENTAL DESIGN: The cytotoxic effects of AQ4N prodrug and its bioreduced form, AQ4, were tested against multiple human tumor cell lines using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Nude mice bearing s.c. or orthotopically implanted human BxPC-3 or Panc-1 tumor cells were treated with AQ4N. Tumor growth inhibition, time to progression/end point, and liver metastasis were evaluated in treatment versus control groups. Plasma and tumor levels of AQ4N and its metabolites were quantitated by liquid chromatography-tandem mass spectrometry. RESULTS: In contrast to AQ4N, the bioreduced AQ4 metabolite displayed potent cytotoxicity in many human tumor lines, including those derived from human pancreatic adenocarcinomas. Single-agent administration of AQ4N significantly delayed tumor growth, progression, and survival in a manner comparable with gemcitabine in multiple pancreatic tumor models in vivo. Survival increases were accompanied by a reduction in incidence and spread of liver metastasis. Quantitation of AQ4N and its metabolites in tumor-bearing mice showed that the prodrug is rapidly cleared from the circulation by 24 h and neither of the bioreduced metabolites was detected in plasma. In contrast, AQ4N readily penetrated BxPC-3 tumors and the cytotoxic AQ4 metabolite rapidly accumulated in tumor tissues at high levels in a dose-dependent fashion. CONCLUSION: AQ4N undergoes rapid and selective conversion into the potent antineoplastic metabolite AQ4 in tumors in vivo and provides proof of principle for the use of hypoxia-activated prodrugs in the treatment against pancreatic cancers.


Assuntos
Antraquinonas/farmacologia , Antineoplásicos/farmacologia , Hipóxia Celular/fisiologia , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Antraquinonas/metabolismo , Antineoplásicos/metabolismo , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Camundongos , Camundongos Nus , Metástase Neoplásica/tratamento farmacológico , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia
8.
Pharm Res ; 21(10): 1880-5, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15553236

RESUMO

PURPOSE: In order to improve the in vitro and in vivo efficacy of an integrin antagonist (IA) of the extracellular domain of the alphavbeta3 integrin, a receptor upregulated on tumor neovasculature, the IA was attached to the surface of a dextran-coated liposome (DCL). IA-DCLs were characterized in vitro, and the pharmacokinetic and antitumor properties were assessed in vivo. METHODS: The in vitro binding properties were measured with purified integrin, endothelial cells, and melanoma cells. The pharmacokinetic parameters were measured in healthy mice with 14C-labeled IA-DCLs and anti-tumor efficacy was assessed with the M21 human melanoma xenograft mouse model. RESULTS: In vitro, IC50 values for IA-DCLs and IA are similar, and IA-DCLs inhibit cell proliferation relative to controls. IA-DCLs are stable in serum, and the pharmacokinetic half-life in mice is 23 h. In the M21/mouse model, statistically significant inhibition of tumor growth was observed for mice treated with IA-DCLs, whereas controls including saline, DCLs lacking IA, and cyclo(RGDfV) were ineffective. Increased apoptosis and a reduction in vessel counts relative to controls were present in tumors from animals treated with IA-DCLs. CONCLUSIONS: These results demonstrate that IA-DCLs are potent anti-angiogenic therapeutic agents with superior in vivo activity and pharmacology compared to unmodified IA.


Assuntos
Antineoplásicos/administração & dosagem , Materiais Biocompatíveis , Integrina alfaVbeta3/antagonistas & inibidores , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dextranos , Portadores de Fármacos , Composição de Medicamentos , Sistemas de Liberação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Meia-Vida , Humanos , Marcação In Situ das Extremidades Cortadas , Lipossomos , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Distribuição Tecidual
9.
Biotechniques ; 36(3): 520-4, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15038168

RESUMO

White blood cells and their secreted products are key elements of immune systems biology that are important indicators of patient health and disease. We have developed the SurroScan microvolume laser scanning cytometer to immunoprofile hundreds of variables, including cell populations, cell surface antigens, and intracellular molecules in antibody-based assays on small samples (about 1 mL) of whole blood, processed blood, or other fluids without cell purification or washing steps. The system enables high-throughput, robust and automated data capture and analysis. We demonstrate the utility of this immunoprofiling technology platform by surveying patient samples before and after glucocorticosteroid administration and show both the expected and novel response characteristics. This system complements recent advances in genomic and proteomic approaches to disease prediction and monitoring.


Assuntos
Análise de Falha de Equipamento , Citometria de Fluxo/instrumentação , Imunoensaio/instrumentação , Mapeamento de Interação de Proteínas/instrumentação , Robótica/instrumentação , Desenho de Equipamento , Citometria de Fluxo/métodos , Humanos , Imunoensaio/métodos , Mapeamento de Interação de Proteínas/métodos , Reprodutibilidade dos Testes , Robótica/métodos , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...