Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Clin Infect Dis ; 74(1): 95-104, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33693561

RESUMO

BACKGROUND: Inflammation is associated with end-organ disease and mortality for people with human immunodeficiency virus (PWH). Ruxolitinib, a Jak 1/2 inhibitor, reduces systemic inflammation for individuals without human immunodeficiency virus (HIV) and HIV reservoir markers ex vivo. The goal of this trial was to determine safety and efficacy of ruxolitinib for PWH on antiretroviral therapy (ART). METHODS: AIDS Clinical Trials Group (ACTG) A5336 was an open-label, multisite, randomized controlled trial (RCT). Participants were randomly assigned (2:1) using centralized software to ruxolitinib (10 mg twice daily) plus stable ART for 5 weeks vs ART alone, stratified by efavirenz use. Eligible participants were suppressed on ART for ≥2 years, without comorbidities, and had >350 CD4+ T cells/µL. Primary endpoints were premature discontinuation, safety events, and change in plasma interleukin 6 (IL-6). Secondary endpoints included other measures of inflammation/immune activation and HIV reservoir. RESULTS: Sixty participants were enrolled from 16 May 2016 to 10 January 2018. Primary safety events occurred in 2.5% (1 participant) for ruxolitinib and 0% for controls (P = .67). Three participants (7.5%) prematurely discontinued ruxolitinib. By week 5, differences in IL-6 (mean fold change [FC], 0.93 vs 1.10; P = .18) and soluble CD14 (mean FC, 0.96 vs 1.08; relative FC, 0.96 [90% confidence interval {CI}, .90-1.02]) levels for ruxolitinib vs controls was observed. Ruxolitinib reduced CD4+ T cells expressing HLA-DR/CD38 (mean difference, -0.34% [90% CI, -.66% to -.12%]) and Bcl-2 (mean difference, -3.30% [90% CI, -4.72% to -1.87%]). CONCLUSIONS: In this RCT of healthy, virologically suppressed PWH on ART, ruxolitinib was well-tolerated. Baseline IL-6 levels were normal and showed no significant reduction. Ruxolitinib significantly decreased markers of immune activation and cell survival. Future studies of Jak inhibitors should target PWH with residual inflammation despite suppressive ART. CLINICAL TRIALS REGISTRATION: NCT02475655.


Assuntos
Infecções por HIV , Pirimidinas , Adulto , HIV , Humanos , Nitrilas/uso terapêutico , Pirazóis , Pirimidinas/uso terapêutico
2.
Metabol Open ; 10: 100090, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33889834

RESUMO

BACKGROUND AND AIM: The incidence of hepatocellular carcinoma (HCC) decreases significantly in chronic hepatitis C (CHC) patients with sustained virologic response (SVR) after pegylated-interferon plus ribavirin (PR) or direct-acting antiviral (DAAs) therapy. We follow-up a single cohort of CHC patients to identify risk factors associated with HCC development post-SVR. METHOD: CHC patients with SVR in Beijing/Hong Kong were followed up at 12-24 weekly intervals with surveillance for HCC by ultrasonography and alpha-fetoprotein (AFP). Multivariate Cox proportional hazards regression analysis was used to explore factors associated with HCC occurrence. RESULTS: Between October 2015 and May 2017, SVR was observed in 519 and 817 CHC patients after DAAs and PR therapy respectively. After a median post -SVR follow-up of 48 months, HCC developed in 54 (4.4%) SVR subjects. By adjusted Cox analysis, older age (≥55 years) [HR 2.4, 95% CI (1.3-4.3)], non-alcoholic fatty liver diseases [HR 2.4, 95%CI (1.3-4.2), higher AFP level (≥20 ng/ml) [HR 3.4, 95%CI (2.0-5.8)], higher liver stiffness measurement (≥14.6 kPa) [HR 4.2, 95%CI (2.3-7.6)], diabetes mellitus [HR 4.2, 95%CI (2.4-7.4)] at pre-treatment were associated with HCC occurrence. HCC patients in the DAAs induced SVR group had a higher prevalence of NAFLD as compared with those in the PR induced SVR group, 62% (18/29) vs 28% (7/25), p = 0.026. A nomogram formulated with the above six independent variables had a Concordance-Index of 0.835 (95% CI 0.783-0.866). CONCLUSION: Underlying NAFLD is associated with increased incidence of HCC in chronic HCV patients post-SVR, particularly in those treated with DAA.

3.
J Neurovirol ; 26(6): 838-845, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32901392

RESUMO

Despite combined antiretroviral therapy (cART), HIV infection in the CNS persists with reported increases in activation of macrophages (MΦ), microglia, and surrounding astrocytes/neurons, conferring HIV-induced inflammation. Chronic inflammation results in HIV-associated neurocognitive disorders (HAND) with reported occurrence of up to half of individuals with HIV infection. The existing HAND mouse model used by laboratories including ours, and the effect of novel agents on its pathology present with labor-intensive and time-consuming limitations since brain sections and immunohistochemistry assays have to be performed and analyzed. A novel flow cytometry-based system to objectively quantify phenotypic effects of HIV using a SCID mouse HAND model was developed which demonstrated that the HIV-infected mice had significant increases in astrogliosis, loss of neuronal dendritic marker, activation of murine microglia, and human macrophage explants compared to uninfected control mice. HIV p24 could also be quantified in the brains of the infected mice. Correlation of these impairments with HIV-induced brain inflammation and previous behavioral abnormalities studies in mice suggests that this model can be used as a fast and relevant throughput methodology to quantify preclinical testing of novel treatments for HAND.


Assuntos
Encéfalo/metabolismo , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Gliose/genética , Infecções por HIV/genética , HIV-1/genética , Animais , Astrócitos/metabolismo , Astrócitos/virologia , Biomarcadores/metabolismo , Encéfalo/virologia , Disfunção Cognitiva/complicações , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/virologia , Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/complicações , Gliose/metabolismo , Gliose/virologia , Proteína do Núcleo p24 do HIV/genética , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/metabolismo , HIV-1/patogenicidade , Humanos , Inflamação , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Masculino , Camundongos , Camundongos SCID , Microglia/metabolismo , Microglia/virologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Fenótipo
5.
J Am Chem Soc ; 140(7): 2478-2484, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29406750

RESUMO

DNA origami is a promising molecular delivery system for a variety of therapeutic applications including cancer therapy, given its capability to fabricate homogeneous nanostructures whose physicochemical properties (size, shape, surface chemistry) can be precisely tailored. However, the correlation between DNA-origami design and internalization efficiency in different cancer cell lines remains elusive. We investigated the cellular uptake of four DNA-origami nanostructures (DONs) with programmed sizes and shapes in multiple human cancer cell lines. The cellular uptake efficiency of DONs was influenced by size, shape, and cell line. Scavenger receptors were responsible for the internalization of DONs into cancer cells. We observed distinct stages of the internalization process of a gold nanoparticle (AuNP)-tagged rod-shape DON, using high-resolution transmission electron microscopy. This study provides detailed understanding of cellular uptake and intracellular trafficking of DONs in cancer cells, and offers new insights for future optimization of DON-based drug delivery systems for cancer treatment.


Assuntos
DNA/farmacocinética , Ouro/farmacocinética , Nanopartículas Metálicas/química , Linhagem Celular Tumoral , DNA/química , Sistemas de Liberação de Medicamentos , Ouro/química , Humanos , Tamanho da Partícula
6.
Angew Chem Int Ed Engl ; 56(50): 16023-16027, 2017 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-29076273

RESUMO

Short interfering RNA (siRNA) is a promising molecular tool for cancer therapy, but its clinical success is limited by the lack of robust in vivo delivery systems. Rationally designed DNA nanoparticles (DNPs) have emerged as facile delivery vehicles because their physicochemical properties can be precisely controlled. Nonetheless, few studies have used DNPs to deliver siRNAs in vivo, and none has demonstrated therapeutic efficacy. Herein, we constructed a number of DNPs of rectangular and tubular shapes with varied dimensions using the modular DNA brick method for the systemic delivery of siRNA that targets anti-apoptotic protein Bcl2. The siRNA delivered by the DNPs inhibited cell growth both in vitro and in vivo, which suppressed tumor growth in a xenograft model that specifically correlated with Bcl2 depletion. This study suggests that DNPs are effective tools for the systemic delivery of therapeutic siRNA and have great potential for further clinical translation.


Assuntos
Antineoplásicos/administração & dosagem , DNA/química , Sistemas de Liberação de Medicamentos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Humanos , Camundongos , Neoplasias/genética , Neoplasias/patologia , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Tamanho da Partícula , Proteínas Proto-Oncogênicas c-bcl-2/deficiência , RNA Interferente Pequeno/síntese química , RNA Interferente Pequeno/química , Propriedades de Superfície , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Nat Cell Biol ; 17(11): 1484-96, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26479318

RESUMO

The oxidative pentose phosphate pathway (PPP) contributes to tumour growth, but the precise contribution of 6-phosphogluconate dehydrogenase (6PGD), the third enzyme in this pathway, to tumorigenesis remains unclear. We found that suppression of 6PGD decreased lipogenesis and RNA biosynthesis and elevated ROS levels in cancer cells, attenuating cell proliferation and tumour growth. 6PGD-mediated production of ribulose-5-phosphate (Ru-5-P) inhibits AMPK activation by disrupting the active LKB1 complex, thereby activating acetyl-CoA carboxylase 1 and lipogenesis. Ru-5-P and NADPH are thought to be precursors in RNA biosynthesis and lipogenesis, respectively; thus, our findings provide an additional link between the oxidative PPP and lipogenesis through Ru-5-P-dependent inhibition of LKB1-AMPK signalling. Moreover, we identified and developed 6PGD inhibitors, physcion and its derivative S3, that effectively inhibited 6PGD, cancer cell proliferation and tumour growth in nude mice xenografts without obvious toxicity, suggesting that 6PGD could be an anticancer target.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Lipogênese , Neoplasias/metabolismo , Via de Pentose Fosfato , Fosfogluconato Desidrogenase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Humanos , Neoplasias/patologia , Estresse Oxidativo , Ribulosefosfatos/metabolismo , Transdução de Sinais
8.
Cancer ; 120(24): 3940-51, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25103371

RESUMO

BACKGROUND: Platinum-based therapy combined with cetuximab is standard first-line therapy for recurrent or metastatic squamous cell carcinoma of the head and neck (RMSCCHN). Preclinical studies have suggested that mammalian target of rapamycin inhibitors may overcome resistance to epidermal growth factor receptor blockers and may augment cetuximab antitumor activity. We conducted a phase 1b trial of carboplatin, cetuximab, and everolimus for untreated RMSCCHN. METHODS: Patients received carboplatin (area under the curve = 2 mg/ml/min; 3 weeks on, 1 week off), cetuximab (with a loading dose of 400 mg/m(2) and then 250 mg/m(2) weekly), and dose-escalating everolimus (2.5, 5.0, 7.5, and 10 mg/day) with a 3+3 design. After 4 cycles, patients without progression continued cetuximab/everolimus until progression or intolerable toxicity. Patients (age ≥ 18 years) had previously untreated, unresectable RMSCCHN not amenable to radiotherapy and an Eastern Cooperative Oncology Group performance status of 0 to 2. RESULTS: The study enrolled 20 patients (male/female = 18/2) with RMSCCHN; the median age was 65 years (44-75 years). Thirteen patients received everolimus (male/female = 92%). Two of 6 patients receiving 2.5 mg/day experienced dose-limiting toxicity (DLT) with grade 3 hyponatremia and nausea. In 7 patients receiving de-escalated everolimus (2.5 mg every other day), grade 3 hyperglycemia produced DLT in 1 of 6 patients. The objective response rate (RR) was 61.5% (all partial responses). Progression-free survival (PFS) was 8.15 months. The pharmacokinetics of everolimus was described with a 2-compartment mixed-effects model. There was a significant correlation between tumor p-p44/42 staining and response (P = .044) and a marginally significant correlation between phosphorylated mammalian target of rapamycin and overall survival. CONCLUSIONS: The maximum tolerated dose of everolimus with cetuximab and carboplatin was 2.5 mg every other day. The regimen was associated with an encouraging RR and PFS, and this suggested possible clinical efficacy in a select group of patients with squamous cell carcinoma of the head and neck.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carboplatina/administração & dosagem , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Sirolimo/análogos & derivados , Adulto , Idoso , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carboplatina/efeitos adversos , Carboplatina/farmacocinética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Cetuximab , Intervalo Livre de Doença , Esquema de Medicação , Everolimo , Feminino , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Metástase Neoplásica , Recidiva , Sirolimo/administração & dosagem , Sirolimo/efeitos adversos , Sirolimo/farmacocinética , Carcinoma de Células Escamosas de Cabeça e Pescoço
9.
Cancer Prev Res (Phila) ; 7(3): 283-91, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24085777

RESUMO

Epidermal growth factor receptor (EGFR) and COX-2 inhibitors synergistically inhibit head and neck squamous cell carcinoma tumorigenesis in preclinical studies. We conducted a phase I and pharmacokinetic study with the erlotinib and celecoxib combination in patients with advanced premalignant lesions. Thirty-six subjects with oral leukoplakia, mild, moderate, or severe dysplasia, or carcinoma in situ were screened for study participation; 12 consented and received therapy for a median of 5.38 months. Erlotinib was escalated following a standard 3+3 design at 50, 75, and 100 mg orally daily and celecoxib was fixed at 400 mg twice daily for 6 months. Biopsy of lesions and cytobrush of normal mucosa were performed at baseline, 3, 6, and 12 months. Erlotinib pharmacokinetics were analyzed in 10 subjects. The maximum tolerated dose of erlotinib with celecoxib 400 mg BID was 50 mg per day with skin rash being the main observed toxicity. Overall histologic response rate was 63% (complete response, 43%; partial response, 14%; stable disease, 29%; and disease progression, 14%). With median follow-up of 36 months, mean time to progression to higher-grade dysplasia or carcinoma was 25.4 months. Downregulation of EGFR and p-ERK in follow-up biopsies correlated with response to treatment. Larger average erlotinib V/F (approximately 308 L) and CL/F (8.3 L/h) compared with previous studies may be related to relatively large average bodyweights. Average erlotinib t1/2 was 25.6 hours. Encouraging responses to the celecoxib and erlotinib combination correlated with EGFR pathway inhibition. Although erlotinib-related rash was the main limitation to dose escalation, the intervention was well tolerated.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/prevenção & controle , Quimioprevenção/métodos , Neoplasias de Cabeça e Pescoço/prevenção & controle , Pirazóis/administração & dosagem , Quinazolinas/administração & dosagem , Sulfonamidas/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carcinoma de Células Escamosas/metabolismo , Celecoxib , Progressão da Doença , Cloridrato de Erlotinib , Feminino , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Pirazóis/farmacocinética , Quinazolinas/farmacocinética , Sulfonamidas/farmacocinética
10.
Curr Opin HIV AIDS ; 8(6): 556-64, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24100876

RESUMO

PURPOSE OF REVIEW: This review focuses on the chemical and pharmacological rationale behind the development of nucleoside antiviral prodrugs (NAPs). RECENT FINDINGS: Highly efficacious NAPs have been developed that extend and improve the quality of lives of individuals infected with HIV and hepatitis B virus (HBV), herpes viruses, and adenovirus infection in immunocompromised individuals. A very high rate of hepatitis C virus (HCV) cure is now possible using NAPs combined with other direct acting antiviral agents (DAAs). SUMMARY: Prodrug strategies can address the issues of poor oral bioavailability and delivery of active metabolites to the targeted cells. Additionally, NAPs demonstrate potential for improving deficiencies in oral absorption, metabolism, tissue distribution, cellular accumulation, phosphorylation, and overall potency, in addition to diminishing potential for in-vivo selection of resistant viruses. NAPs continue to be the backbone for the treatment of HIV and HBV, herpesviruses, and adenovirus infections because their active forms are potent, have long intracellular half-lives and are relatively safe with high barrier to resistance.


Assuntos
Antivirais/administração & dosagem , Antivirais/farmacologia , Nucleosídeos/administração & dosagem , Nucleosídeos/farmacologia , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacologia , Viroses/tratamento farmacológico , Antivirais/farmacocinética , Humanos , Nucleosídeos/farmacocinética , Pró-Fármacos/farmacocinética
11.
Antimicrob Agents Chemother ; 57(3): 1262-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23263005

RESUMO

Understanding the cellular pharmacology of antiretroviral agents in macrophages and subsequent correlation with antiviral potency provides a sentinel foundation for definition of the dynamics between antiretroviral agents and viral reservoirs across multiple cell types, with the goal of eradication of HIV-1 from these cells. Various clinically relevant nucleoside antiviral agents, and the integrase inhibitor raltegravir, were selected for this study. The intracellular concentrations of the active metabolites of the nucleoside analogs were found to be 5- to 140-fold lower in macrophages than in lymphocytes, and their antiviral potency was significantly lower in macrophages constitutively activated with macrophage colony-stimulating factor (M-CSF) during acute infection than in resting macrophages (EC(50), 0.4 to 9.42 µM versus 0.03 to 0.4 µM, respectively). Although tenofovir-treated cells displayed significantly lower intracellular drug levels than cells treated with its prodrug, tenofovir disoproxil fumarate, the levels of tenofovir-diphosphate for tenofovir-treated cells were similar in lymphocytes and macrophages. Raltegravir also displayed significantly lower intracellular concentrations in macrophages than in lymphocytes, independent of the activation state, but had similar potencies in resting and activated macrophages. These data underscore the importance of delivering adequate levels of drug to macrophages to reduce and eradicate HIV-1 infection.


Assuntos
Adenina/análogos & derivados , Adenina/farmacologia , Fármacos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Organofosfonatos/farmacologia , Pirrolidinonas/farmacologia , Transporte Biológico , Inibidores de Integrase de HIV/farmacologia , HIV-1/enzimologia , HIV-1/crescimento & desenvolvimento , Humanos , Linfócitos/virologia , Ativação de Macrófagos/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/virologia , Especificidade de Órgãos , Cultura Primária de Células , Raltegravir Potássico , Inibidores da Transcriptase Reversa/farmacologia , Tenofovir
12.
Antivir Ther ; 17(7): 1233-42, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23018140

RESUMO

BACKGROUND: Monocytes transmigrating to the brain play a central role in HIV neuropathology. We hypothesized that the continued existence of neurocognitive impairment (NCI) despite potent antiretroviral (ARV) therapy is mediated by the inability of such therapy to control this monocyte/macrophage reservoir. METHODS: Cross-sectional and longitudinal analyses were conducted within a prospectively enrolled cohort. We devised a monocyte efficacy (ME) score based on the anticipated effectiveness of ARV medications against monocytes/macrophages using published macrophage in vitro drug efficacy data. We examined, within an HIV neurocognitive database, its association with composite neuropsychological test scores (NPZ8) and clinical cognitive diagnoses among subjects on stable ARV medications unchanged for >6 months prior to assessment. RESULTS: Among 139 subjects on ARV therapy, higher ME score correlated with better NPZ8 performance (r=0.23, P<0.01), whereas a score devised to quantify expected penetration effectiveness of ARVs into the brain (CPE score) did not (r=0.12, P=0.15). In an adjusted model (adjusted r(2)=0.12), ME score (ß=0.003, P=0.02), CD4(+) T-cell nadir (ß=0.001, P<0.01) and gender (ß=-0.456, P=0.02) were associated with NPZ8, whereas CPE score was not (ß=0.003, P=0.94). A higher ME score was associated with better clinical cognitive status (P<0.01). With a range of 12.5-433.0 units, a 100-unit increase in ME score resulted in a 10.6-fold decrease in the odds of a dementia diagnosis compared with normal cognition (P=0.01). CONCLUSIONS: ARV efficacy against monocytes/macrophages correlates with cognitive function in HIV-infected individuals on ARV therapy within this cohort. If validated, efficacy against monocytes/macrophages may provide a new target to improve HIV NCI.


Assuntos
Transtornos Cognitivos/patologia , HIV-1/patogenicidade , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Complexo AIDS Demência/patologia , Complexo AIDS Demência/virologia , Adulto , Antirretrovirais/metabolismo , Antirretrovirais/farmacologia , Contagem de Linfócito CD4 , Cognição , Transtornos Cognitivos/virologia , Estudos Transversais , Feminino , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Infecções por HIV/virologia , HIV-1/metabolismo , Humanos , Estudos Longitudinais , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Testes Neuropsicológicos , Valor Preditivo dos Testes , Estudos Prospectivos , Estatísticas não Paramétricas
13.
Cancer Chemother Pharmacol ; 69(3): 577-90, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21913035

RESUMO

PURPOSE: DT-IgG is a fully humanized dual-target therapeutic antibody being developed to simultaneously target epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF), important signaling molecules for tumor growth. The antitumor pharmacodynamics (PD) of DT-IgG was studied in nude mice bearing human tumor xenografts with different EGFR and VEGF expressions and K-ras oncogene status and compared with bevacizumab, cetuximab and bevacizumab + cetuximab. METHODS: Mice bearing human oral squamous cell carcinoma (Tu212), lung adenocarcinoma (A549), or colon cancer (GEO) subcutaneous xenografts were administered with the antibodies intraperitoneally (i.p.), and tumor volumes were measured versus time. Nonlinear mixed effects modeling (NONMEM) was used to study drug potencies (IC(50)) and variations in tumor growth. RESULTS: The PD models adequately described tumor responses for the antibody dose regimens. In vivo IC(50) values varied with EGFR and K-ras status. DT-IgG had a similar serum t (1/2) as cetuximab (~1.7 vs. 1.5 day), was more rapid than bevacizumab (~6 day), and had the largest apparent distribution volume (DT-IgG > cetuximab > bevacizumab). The efficacy of DT-IgG was comparable to bevacizumab despite lower serum concentrations, but was less than bevacizumab + cetuximab. CONCLUSIONS: A lower IC(50) of DT-IgG partially compensated for lower serum concentrations than bevacizumab and cetuximab, but may require higher doses for comparable efficacy as the combination. The model adequately predicted variations of tumor response at the DT-IgG doses tested and could be used for targeting specific tumor efficacies for future testing.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Imunoglobulina G/farmacologia , Neoplasias Experimentais/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/sangue , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/sangue , Antineoplásicos/uso terapêutico , Bevacizumab , Linhagem Celular Tumoral , Cetuximab , Receptores ErbB/genética , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/uso terapêutico , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Neoplasias Experimentais/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Head Neck ; 31(10): 1263-73, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19399750

RESUMO

BACKGROUND: This study was to explore whether the efficacy of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor ZD1839 (Z, Iressa, gefitinib) plus chemotherapeutic agents docetaxel (D) and cisplatin (P) may benefit from sequencing of the combination. METHODS: Three head and neck cancer cell lines were used to study the effect of various combinations of and relative sequencing of D, P, and Z in cell growth inhibition. A population pharmacokinetic stimulation study was conducted on Z in silico and used together with the growth inhibition data to derive principles for future in vivo use of this drug combination. RESULTS: The inhibitory effects of Z on combinations of D and P were sequence dependent. Treatment simultaneously with DPZ or with DP followed by Z (DP-->Z) showed synergistic effects in all 3 cell lines. However, sequencing with Z followed by DP (Z-->DP), gave an antagonistic effect, suggesting that D and P should be administered when the effect of Z is low. The induction of apoptosis was also sequence dependent. The in silico pharmacokinetic study suggested the feasibility of deriving a 5-day-on/2-day-off regimen for Z, in which D and P administration commences when levels of Z are low, allowing levels of Z to accumulate sufficiently during the remainder of the cycle. CONCLUSION: These data suggests that it is feasible to design clinical trials with these settings to maximize the efficacy of this combined drug regimen.


Assuntos
Antineoplásicos/farmacocinética , Cisplatino/farmacocinética , Receptores ErbB/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacocinética , Quinazolinas/farmacocinética , Taxoides/farmacocinética , Anexina A5/farmacologia , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Docetaxel , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Gefitinibe , Humanos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Taxoides/administração & dosagem , Células Tumorais Cultivadas
15.
Antivir Chem Chemother ; 18(6): 329-41, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18320937

RESUMO

Current highly active antiretroviral therapy (HAART) requires the use of combinations of three drugs to minimize the early emergence of drug-resistant HIV strains. Therefore, long-term monotherapy data with new agents are unavailable. However, the development of computer models for Monte-Carlo-type simulations of antiviral monotherapy, which incorporate HIV infection dynamic distributions from previously studied populations, together with pharmacokinetics and pharmacodynamic parameters of the new agent, could serve as an important tool. The nucleoside lamivudine (3TC) was used as a representative drug to standardize an improved pharmacodynamic and infection dynamic monotherapy model. 3TC plasma concentration versus time profiles was used to drive the cellular accumulation of 3TC-triphosphate (TP) in primary human lymphocytes in the model, over a 16 week period. The fraction of HIV reverse transcription inhibited was calculated using the median inhibitory concentration and intracellular 3TC-TP levels. Virus loads and activated CD4+ T-cell counts were generated for 2,200 theoretical individuals and compared with the outcomes of an actual 3TC monotherapy trial at the same dose. Pharmacokinetic variance alone did not account for the interindividual HIV-load variability. However, selection of appropriate distributions of the various pharmacokinetic and infection dynamics parameters produced a similar range of virus load reductions to actual observations. Therefore, once parameter and variance distributions are standardized, this modelling approach could be helpful in planning clinical trials and predicting the antiviral contribution of each agent in a HAART modality.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Citidina Trifosfato/análogos & derivados , Didesoxinucleotídeos/farmacologia , Didesoxinucleotídeos/uso terapêutico , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV/efeitos dos fármacos , Lamivudina/análogos & derivados , Modelos Biológicos , Fármacos Anti-HIV/sangue , Fármacos Anti-HIV/farmacocinética , Fármacos Anti-HIV/farmacologia , Contagem de Linfócito CD4 , Ensaios Clínicos como Assunto , Citidina Trifosfato/sangue , Citidina Trifosfato/farmacocinética , Citidina Trifosfato/farmacologia , Citidina Trifosfato/uso terapêutico , Didesoxinucleotídeos/sangue , Didesoxinucleotídeos/farmacocinética , HIV/enzimologia , HIV/genética , HIV/fisiologia , Infecções por HIV/imunologia , Humanos , Concentração Inibidora 50 , Cinética , Lamivudina/sangue , Lamivudina/farmacocinética , Lamivudina/farmacologia , Lamivudina/uso terapêutico , Método de Monte Carlo , Inibidores da Transcriptase Reversa/sangue , Inibidores da Transcriptase Reversa/farmacocinética , Inibidores da Transcriptase Reversa/farmacologia , Inibidores da Transcriptase Reversa/uso terapêutico , Resultado do Tratamento , Carga Viral
16.
Antivir Chem Chemother ; 18(6): 343-6, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18320938

RESUMO

Amdoxovir, currently in Phase II clinical trials, is rapidly converted to 9-(beta-D-1,3-dioxolan-4-yl)guanine (DXG) by adenosine deaminase in vitro and in humans. The cellular pharmacology of DXG in primary human lymphocytes, including dose-response relationships, intracellular half-life of DXG triphosphate (DXG-TP), and combination studies were determined. DXG produced high levels of DXG-TP with a long half-life (16 h) in activated human peripheral blood mononuclear cells. Since zidovudine (ZDV) and DXG select for different resistance mutations, co-formulation of the these two drugs is an attractive proposition. A combination study between DXG and ZDV showed no reduction of DXG-TP or ZDV-TP. Taken together, these results suggest that an appropriately designed DXG prodrug could be given once a day and that co-formulation with ZDV might be a possibility.


Assuntos
Dioxolanos/farmacologia , Guanina/análogos & derivados , Linfócitos/efeitos dos fármacos , Zidovudina/farmacologia , Células Cultivadas , Dioxolanos/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Guanina/metabolismo , Guanina/farmacologia , HIV/efeitos dos fármacos , Meia-Vida , Humanos , Linfócitos/virologia , Mutação , Fosforilação/efeitos dos fármacos , Pró-Fármacos/farmacologia , Zidovudina/metabolismo
17.
J Neurooncol ; 74(3): 275-80, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16132519

RESUMO

Boron-neutron capture therapy (BNCT) is based on the preferential targeting of tumor cells with (10)B and subsequent irradiation with epithermal neutrons to produce a highly localized field of lethal alpha particles, while sparing neighboring non-targeted cells. BNCT treatment of 9L brain tumors in a rat model using beta-D-5-o-carboranyl-2'-deoxyuridine (D-CDU) resulted in greater efficacy than predicted based on the assumption of a uniform tumor distribution of (10)B. Thus, the geometric heterogeneity of dividing cells in brain tumors warranted studies on the cell cycle dependency of D-CDU accumulation, metabolism and entrapment in a relevant brain tumor cell system. U-271 human glioma cells were synchronized in G(1) or S-phases of the cell cycle. The cellular accumulation and phosphorylation of D-CDU was measured in the G(1) and S-phase cells using high-performance liquid chromatography (HPLC). Cells synchronized in the S-phase accumulated significantly higher amounts of D-CDU and produced larger amounts of negatively charged D-CDU monophosphate (D-CDU-MP) and nido-CDU metabolites than resting cells. Since brain tumors contain a larger proportion of cycling cells than neighboring tissue, these results support the hypothesis that in addition to breakdown of the blood-brain-barrier (BBB) in tumors, the preferential phosphorylation of D-CDU in cycling cells may further enrich the distribution of (10)B in dividing cells. Therefore, dosimetry calculations that include the spatial distribution of cycling cells may be warranted for D-CDU.


Assuntos
Terapia por Captura de Nêutron de Boro , Neoplasias Encefálicas/metabolismo , Ciclo Celular/fisiologia , Desoxiuridina/metabolismo , Glioma/metabolismo , Radiossensibilizantes/metabolismo , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Proliferação de Células , Cromatografia Líquida de Alta Pressão , Desoxiuridina/uso terapêutico , Citometria de Fluxo , Glioma/terapia , Humanos , Radiossensibilizantes/uso terapêutico
18.
Antimicrob Agents Chemother ; 49(7): 2589-97, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15980324

RESUMO

Beta-D-2',3'-dideoxy-3'-oxa-5-fluorocytidine (D-FDOC) is an effective inhibitor of human immunodeficiency virus 1 (HIV-1) and HIV-2, simian immunodeficiency virus, and hepatitis B virus (HBV) in vitro. The purpose of this study was to evaluate the intracellular metabolism of d-FDOC in human hepatoma (HepG2), human T-cell lymphoma (CEM), and primary human peripheral blood mononuclear (PBM) cells by using tritiated compound. By 24 h, the levels of D-FDOC-triphosphate (D-FDOC-TP) were 2.8 +/- 0.4, 6.7 +/- 2.3, and 2.0 +/- 0.1 pmol/10(6) cells in HepG2, CEM, and primary human PBM cells, respectively. Intracellular D-FDOC-TP concentrations remained greater than the 50% inhibitory concentration for HIV-1 reverse transcriptase for up to 24 h after removal of the drug from cell cultures. In addition to d-FDOC-monophosphate (D-FDOC-MP), -diphosphate (D-FDOC-DP), and -TP, D-FDOC-DP-ethanolamine and d-FDOC-DP-choline were detected in all cell extracts as major intracellular metabolites. D-FDOC was not a substrate for Escherichia coli thymidine phosphorylase. No toxicity was observed in mice given D-FDOC intraperitoneally for 6 days up to a dose of 100 mg/kg per day. Pharmacokinetic studies in rhesus monkeys indicated that D-FDOC has a t(1/2) of 2.1 h in plasma and an oral bioavailability of 38%. The nucleoside was excreted unchanged primary in the urine, and no metabolites were detected in plasma or urine. These results suggest that further safety and pharmacological studies are warranted to assess the potential of this nucleoside for the treatment of HIV- and HBV-infected individuals.


Assuntos
Antivirais , Citidina , Citidina/análogos & derivados , Animais , Antivirais/administração & dosagem , Antivirais/metabolismo , Antivirais/farmacocinética , Antivirais/toxicidade , Linhagem Celular Tumoral , Citidina/administração & dosagem , Citidina/metabolismo , Citidina/farmacocinética , Citidina/toxicidade , Escherichia coli/enzimologia , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Macaca mulatta , Masculino , Camundongos , Timidina Fosforilase/metabolismo
19.
Nucleosides Nucleotides Nucleic Acids ; 23(1-2): 291-306, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15043155

RESUMO

The carboranyl nucleotides beta-D-5-o-carboranyl-2'-deoxyuridine (D-CDU), 1-(beta-L-arabinosyl)-5-o-carboranyluracil (D-ribo-CU) and the nucleotide base 5-o-carboranyluracil (CU), were developed as sensitizers for boron neutron capture therapy (BNCT). A structure activity study was initiated to determine the agent most suitable for targeting prostate tumors. Cellular accumulation studies were performed using LNCaP human prostate tumor cells, and the respective tumor disposition profiles were investigated in male nude mice bearing LNCaP and 9479 human prostate tumor xenografts in their flanks. D-CDU achieved high cellular concentrations in LNCaP cells and up to 2.5% of the total cellular compound was recovered in the 5'-monophosphorylated form. In vivo concentrations of D-CDU were similar in LNCaP and 9479 tumor xenografts. Studies in 9479 xenografted bearing mice indicated that increasing the number of hydroxyl groups in the sugar moeity of the carboranyl nucleosides corresponded with an increased rate and extent of renal elimination, shorter serum half-lives and an increased tissue specificity. Tumor/normal prostate ratios were greatest with the nucleoside base CU. These studies indicate that similar nucleoside analogues and bases may have different tissue affinities and retention properties, which should be considered when selecting agents for sensitizing specific tumors for eventual BNCT treatment. CU was found to be the most suitable compound for further development to treat prostate cancer.


Assuntos
Próstata/metabolismo , Neoplasias da Próstata/radioterapia , Uracila/análogos & derivados , Animais , Compostos de Boro/farmacocinética , Terapia por Captura de Nêutron de Boro , Humanos , Masculino , Camundongos , Camundongos Nus , Especificidade de Órgãos , Neoplasias da Próstata/metabolismo , Fatores de Tempo , Uracila/química , Uracila/farmacocinética
20.
Antimicrob Agents Chemother ; 47(6): 1922-8, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12760868

RESUMO

1-(2,3-Dideoxy-2-fluoro-beta-L-glyceropent-2-enofuranosyl)cytosine (L-2'-Fd4C) is an L-nucleoside analogue with both anti-human immunodeficiency virus (HIV) and anti-hepatitis B virus (HBV) activity with median effective concentrations of 0.12 microM in peripheral blood mononuclear cells and 0.002 microM in HepG2-2.2.15 cells, respectively. The purpose of this study was to examine the antihepadnavirus potency and pharmacokinetics of L-2'-Fd4C in vivo. HBV-transgenic mice treated intraperitoneally with L-2'-Fd4C showed a reduction of HBV levels in their blood comparable to that produced by lamivudine. The pharmacokinetics of L-2'-Fd4C in rhesus monkeys was evaluated after intravenous and oral administration. The concentrations in plasma declined in a biexponential manner after intravenous administration, with a long terminal-phase half-life of 5.02 h. The steady-state volumes of distribution and systemic clearance were 1.09 liter x kg(-1) and 0.25 liter x h(-1) x kg(-1), respectively, with a renal clearance of 0.16 liter x h(-1) x kg(-1). The oral bioavailability was approximately 44%. About 53% of the compound administered intravenously and 19% of that administered orally were recovered unchanged in the urine within the 24-h urine collection period, and no other metabolite was detected. The compound penetrated the central nervous system at concentrations that exceeded the median effective antiviral concentration against HIV in cell cultures. Based upon these observations, further testing to develop this agent for treatment of HIV and HBV infections is warranted.


Assuntos
Antivirais/farmacocinética , Citosina/análogos & derivados , Citosina/farmacocinética , Vírus da Hepatite B/efeitos dos fármacos , Hepatite B Crônica/metabolismo , Animais , Antivirais/antagonistas & inibidores , Antivirais/sangue , Antivirais/farmacologia , Antivirais/urina , Citosina/sangue , Citosina/líquido cefalorraquidiano , Citosina/farmacologia , Citosina/urina , Replicação do DNA/efeitos dos fármacos , DNA Viral/química , DNA Viral/genética , Feminino , Vírus da Hepatite B/genética , Vírus da Hepatite B/metabolismo , Hepatite B Crônica/sangue , Hepatite B Crônica/líquido cefalorraquidiano , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/urina , Macaca mulatta , Masculino , Camundongos , Camundongos Transgênicos , Células Tumorais Cultivadas , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA