Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
PLoS One ; 17(7): e0271066, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35816490

RESUMO

As ACE2 is the critical SARS-CoV-2 receptor, we hypothesized that aerosol administration of clinical grade soluble human recombinant ACE2 (APN01) will neutralize SARS-CoV-2 in the airways, limit spread of infection in the lung, and mitigate lung damage caused by deregulated signaling in the renin-angiotensin (RAS) and Kinin pathways. Here, after demonstrating in vitro neutralization of SARS-CoV-2 by APN01, and after obtaining preliminary evidence of its tolerability and preventive efficacy in a mouse model, we pursued development of an aerosol formulation. As a prerequisite to a clinical trial, we evaluated both virus binding activity and enzymatic activity for cleavage of Ang II following aerosolization. We report successful aerosolization for APN01, retaining viral binding as well as catalytic RAS activity. Dose range-finding and IND-enabling repeat-dose aerosol toxicology testing were conducted in dogs. Twice daily aerosol administration for two weeks at the maximum feasible concentration revealed no notable toxicities. Based on these results, a Phase I clinical trial in healthy volunteers has now been initiated (NCT05065645), with subsequent Phase II testing planned for individuals with SARS-CoV-2 infection.


Assuntos
Tratamento Farmacológico da COVID-19 , Aerossóis , Enzima de Conversão de Angiotensina 2 , Angiotensinas , Animais , Ensaios Clínicos Fase I como Assunto , Cães , Humanos , Camundongos , Nebulizadores e Vaporizadores , Peptidil Dipeptidase A/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina , SARS-CoV-2
2.
Bioorg Med Chem Lett ; 21(23): 7146-50, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22001089

RESUMO

Several acetyl-protected hydroxybenzyl diethyl phosphates (EHBPs) that are capable of forming quinone methide intermediates were synthesized and their cell growth inhibitory properties were evaluated in four different human cancer cell lines. Compounds 1, 1a, and 1b, corresponding to (4-acetyloxybenzyl diethylphosphate), (3-methyl-4-acetyloxybenzyl diethylphosphate), and (3-chloro-4-acetyloxybenzyl diethylphosphate), were significantly more potent than compounds 2 and 3, (2-acetyloxybenzyl diethylphosphate) and (3-acetyloxybenzyl diethylphosphate), respectively. Using HT-29 human colon cancer cells, compounds 1 and 3 increased apoptosis, inhibited proliferation, and caused a G(2)/M block in the cell cycle. Our data suggest that these compounds merit further investigation as potential anti-cancer agents.


Assuntos
Antineoplásicos , Apoptose/efeitos dos fármacos , Organofosfatos/síntese química , Organofosfatos/farmacologia , Acetilação , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Estrutura Molecular , Neoplasias/tratamento farmacológico , Organofosfatos/química
3.
Int J Toxicol ; 30(1): 19-20, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21098339

RESUMO

Recent chronic toxicity studies performed on green tea extracts in fasted dogs have revealed some unique dose-limiting lethal liver, gastrointestinal, and renal toxicities. Key findings included necrosis of hepatic cells, gastrointestinal epithelia and renal tubules, atrophy of reproductive organs, atrophy and necrosis of hematopoietic tissues, and associated hematological changes. The polyphenol cachetins (a mixture of primarily epigallocatechin gallate [≥55%]; plus up to 10% each of epigallocatechin, epicatechin, and epigallocatechin gallate) appeared to be the causative agents for the observed toxicities because they are the active ingredients of green tea extract studied. Conduct of the study in nonfasted dogs under the same testing conditions and dose levels showed unremarkable results. Assuming both studies were valid, at the identified no observed adverse effect levels (NOAEL) of each study, systemic exposures (based on area under the curve [AUC]) were actually lower in fasted than nonfasted dogs, suggesting that fasting may have rendered the target organ systems potentially more vulnerable to the effects of green tea extract. The toxicity mechanisms that produced lethality are not known, but the results are scientifically intriguing. Because tea drinking has become more popular in the United States and abroad, the mode of action and site of action of green tea extract-induced lethal toxicities during fasting and the role of other phytochemical components of Folia Camellia sinensis (including nonpolyphenol fractions, which are often consumed when whole-leaf products are presented) warrant further investigation.


Assuntos
Antioxidantes/toxicidade , Camellia sinensis/química , Catequina/toxicidade , Privação de Alimentos , Animais , Antioxidantes/farmacocinética , Área Sob a Curva , Catequina/farmacocinética , Cães , Relação Dose-Resposta a Droga , Enterócitos/efeitos dos fármacos , Enterócitos/patologia , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Longevidade/efeitos dos fármacos , Masculino , Necrose/induzido quimicamente , Nível de Efeito Adverso não Observado , Extratos Vegetais/farmacocinética , Extratos Vegetais/toxicidade
4.
bioRxiv ; 2021 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-34545364

RESUMO

To develop a universal strategy to block SARS-CoV-2 cellular entry and infection represents a central aim for effective COVID-19 therapy. The growing impact of emerging variants of concern increases the urgency for development of effective interventions. Since ACE2 is the critical SARS-CoV-2 receptor and all tested variants bind to ACE2, some even at much increased affinity (see accompanying paper), we hypothesized that aerosol administration of clinical grade soluble human recombinant ACE2 (APN01) will neutralize SARS-CoV-2 in the airways, limit spread of infection in the lung and mitigate lung damage caused by deregulated signaling in the renin-angiotensin (RAS) and Kinin pathways. Here we show that intranasal administration of APN01 in a mouse model of SARS-CoV-2 infection dramatically reduced weight loss and prevented animal death. As a prerequisite to a clinical trial, we evaluated both virus binding activity and enzymatic activity for cleavage of Ang II following aerosolization. We report successful aerosolization for APN01, retaining viral binding as well as catalytic RAS activity. Dose range-finding and IND-enabling repeat-dose aerosol toxicology testing were conducted in dogs. Twice daily aerosol administration for two weeks at the maximum feasible concentration revealed no notable toxicities. Based on these results, a Phase I clinical trial in healthy volunteers can now be initiated, with subsequent Phase II testing in individuals with SARS-CoV-2 infection. This strategy could be used to develop a viable and rapidly actionable therapy to prevent and treat COVID-19, against all current and future SARS-CoV-2 variants.

5.
Cancer Genomics Proteomics ; 6(1): 41-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19451089

RESUMO

Celecoxib, a selective inhibitor of cyclooxygenase-2 (Cox-2), was efficacious in clinical prevention trials of patients with familial adenomatous polyposis (FAP) and sporadic colorectal cancer. To identify as yet poorly defined molecular determinants of celecoxib efficacy, a multidimensional serum fractionation approach was used coupled with nanospray tandem mass spectrometry to perform label-free global proteomic profiling of serum samples from the FAP/celecoxib prevention trial. Subsequently, the application of an algorithm for large-scale biomarker discovery on comparative serum proteomic profiles of pre- and post-celecoxib treatment samples identified 83 potentially celecoxib-responsive proteins from various cellular compartments, biological processes and molecular functions. Celecoxib modulation of some of these proteins was confirmed in serum samples of FAP patients and colorectal cancer cell lines by Western blotting. Thus, using a shotgun procedure to rapidly identify important celecoxib-modulated proteins, this pilot study has uncovered novel systemic changes some of which are highly relevant for carcinogenesis and vascular biology. Validation of selected markers, especially those involved in key signaling networks and those considered molecular indicators of cardiovascular pathology, in larger celecoxib clinical trials is expected to provide insights into the molecular mechanisms of celecoxib and the efficacy/toxicity issues related to its use as a chemopreventive agent.


Assuntos
Polipose Adenomatosa do Colo/tratamento farmacológico , Polipose Adenomatosa do Colo/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Proteínas Sanguíneas/metabolismo , Proteômica , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Polipose Adenomatosa do Colo/patologia , Western Blotting , Celecoxib , Cromatografia Líquida , Biologia Computacional , Humanos , Fragmentos de Peptídeos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
6.
Cancer Epidemiol Biomarkers Prev ; 17(3): 674-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18349286

RESUMO

The cyclooxygenase (COX)-2 enzyme appears to be an important target for cancer chemoprevention. Given the recent emergence of potentially serious cardiovascular toxicity associated with selective COX-2 inhibitors, nonsteroidal antiinflammatory drugs (NSAIDs), which inhibit both COX-1 and COX-2, have received renewed attention as candidate chemoprevention agents. Sulindac has shown consistent chemopreventive potential in preclinical studies as well as in a limited number of clinical trials reported to date. For the current pharmacokinetic study, sulindac capsules were prepared to facilitate ample agent supplies for future intervention studies. Encapsulation of the parent compound (sulindac sulfoxide) can be readily accomplished, but the effects of alternate formulations on bioavailability have not been rigorously examined. In the present single-dose, two-period crossover trial, we conducted pharmacokinetic analyses of sulindac in capsule (test) versus tablet (reference) formulations. Overall, bioavailability appeared to be higher for the capsule compared with the tablet formulation based on test-to-reference pharmacokinetic variable ratios for the parent compound alone. However, additional analyses based on the sulfide and sulfone metabolites of sulindac with the same pharmacokinetic variables indicated similar chemopreventive exposures between the capsule and tablet formulations. These data support the use of sulindac capsules, which can be readily prepared with matching placebos, in future blinded chemoprevention trials.


Assuntos
Antineoplásicos/farmacocinética , Sulindaco/farmacocinética , Análise de Variância , Antineoplásicos/administração & dosagem , Área Sob a Curva , Disponibilidade Biológica , Cápsulas , Cromatografia Líquida de Alta Pressão , Estudos Cross-Over , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Sulindaco/administração & dosagem , Comprimidos
7.
Cancer Prev Res (Phila) ; 8(4): 296-302, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25762530

RESUMO

The COX inhibitors (NSAID/Coxibs) are a major focus for the chemoprevention of cancer. The COX-2-specific inhibitors have progressed to clinical trials and have shown preventive efficacy in colon and skin cancers. However, they have significant adverse cardiovascular effects. Certain NSAIDs (e.g., naproxen) have a good cardiac profile, but can cause gastric toxicity. The present study examined protocols to reduce this toxicity of naproxen. Female Fischer-344 rats were treated weekly with the urinary bladder-specific carcinogen hydroxybutyl(butyl)nitrosamine (OH-BBN) for 8 weeks. Rats were dosed daily with NPX (40 mg/kg body weight/day, gavage) or with the proton pump inhibitor omeprazole (4.0 mg/kg body weight/day) either singly or in combination beginning 2 weeks after the final OH-BBN. OH-BBN-treated rats, 96% developed urinary bladder cancers. While omeprazole alone was ineffective (97% cancers), naproxen alone or combined with omeprazole-prevented cancers, yielding 27 and 35% cancers, respectively. In a separate study, OH-BBN -: treated rats were administered naproxen: (A) daily, (B) 1 week daily naproxen/1week vehicle, (C) 3 weeks daily naproxen/3 week vehicle, or (D) daily vehicle beginning 2 weeks after last OH-BBN treatment. In the intermittent dosing study, protocol A, B, C, and D resulted in palpable cancers in 27%, 22%, 19%, and 96% of rats (P < 0.01). Short-term naproxen treatment increased apoptosis, but did not alter proliferation in the urinary bladder cancers. Two different protocols that should decrease the gastric toxicity of NSAIDs in humans did not alter chemopreventive efficacy. This should encourage the use of NSAIDs (e.g., naproxen) in clinical prevention trials.


Assuntos
Butilidroxibutilnitrosamina/toxicidade , Naproxeno/uso terapêutico , Estômago/efeitos dos fármacos , Neoplasias da Bexiga Urinária/prevenção & controle , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Western Blotting , Carcinógenos/toxicidade , Feminino , Imunofluorescência , Humanos , Camundongos , Ratos , Ratos Endogâmicos F344 , Estômago/patologia , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/patologia
8.
Cancer Prev Res (Phila) ; 7(2): 246-54, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24346344

RESUMO

Urinary bladder cancer prevention studies were performed with the nonsteroidal anti-inflammatory drugs (NSAID) naproxen (a standard NSAID with a good cardiovascular profile), sulindac, and their nitric oxide (NO) derivatives. In addition, the effects of the ornithine decarboxylase inhibitor, difluoromethylornithine (DFMO), alone or combined with a suboptimal dose of naproxen or sulindac was examined. Agents were evaluated at their human equivalent doses (HED), as well as at lower doses. In the hydroxybutyl(butyl)nitrosamine (OH-BBN) model of urinary bladder cancer, naproxen (400 or 75 ppm) and sulindac (400 ppm) reduced the incidence of large bladder cancers by 82%, 68%, and 44%, respectively, when the agents were initially given 3 months after the final dose of the carcinogen; microscopic cancers already existed. NO-naproxen was highly effective, whereas NO-sulindac was inactive. To further compare naproxen and NO-naproxen, we examined their effects on gene expression in rat livers following a 7-day exposure. Limited, but similar, gene expression changes in the liver were induced by both agents, implying that the primary effects of both are mediated by the parent NSAID. When agents were initiated 2 weeks after the last administration of OH-BBN, DFMO at 1,000 ppm had limited activity, a low dose of naproxen (75 ppm) and sulindac (150 ppm) were highly and marginally effective. Combining DFMO with suboptimal doses of naproxen had minimal effects, whereas the combination of DMFO and sulindac was more active than either agent alone. Thus, naproxen and NO-naproxen were highly effective, whereas sulindac was moderately effective in the OH-BBN model at their HEDs.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Eflornitina/administração & dosagem , Neoplasias Experimentais/prevenção & controle , Óxido Nítrico/administração & dosagem , Neoplasias da Bexiga Urinária/prevenção & controle , Animais , Quimioprevenção/métodos , Avaliação Pré-Clínica de Medicamentos , Feminino , Naproxeno/administração & dosagem , Naproxeno/análogos & derivados , Neoplasias Experimentais/induzido quimicamente , Ratos , Ratos Endogâmicos F344 , Sulindaco/administração & dosagem , Neoplasias da Bexiga Urinária/induzido quimicamente
9.
J Clin Pharmacol ; 53(4): 403-12, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23436338

RESUMO

Sulindac is a prescription-based non-steroidal anti-inflammatory drug (NSAID) that continues to be actively investigated as a candidate cancer chemoprevention agent. To further current understanding of sulindac bioavailability, metabolism, and disposition, we developed a population pharmacokinetic model for the parent compound and its active metabolites, sulindac sulfide, and exisulind. This analysis was based on data from 24 healthy subjects who participated in a bioequivalence study comparing two formulations of sulindac. The complex disposition of sulindac and its metabolites was described by a seven-compartment model featuring enterohepatic recirculation and is the first reported population pharmacokinetic model for sulindac. The derived model was used to explore effects of clinical variables on sulindac pharmacokinetics and revealed that body weight, creatinine clearance, and gender were significantly correlated with pharmacokinetic parameters. Moreover, the model quantifies the relative bioavailability of the sulindac formulations and illustrates the utility of population pharmacokinetics in bioequivalence assessment. This novel population pharmacokinetic model provides new insights regarding the factors that may affect the pharmacokinetics of sulindac and the exisulind and sulindac sulfide metabolites in generally healthy subjects, which have implications for future chemoprevention trial design for this widely available agent.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Antineoplásicos/farmacocinética , Modelos Biológicos , Sulindaco/farmacocinética , Adulto , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/sangue , Antineoplásicos/administração & dosagem , Antineoplásicos/sangue , Disponibilidade Biológica , Cápsulas , Estudos Cross-Over , Feminino , Humanos , Masculino , Neoplasias/prevenção & controle , Sulindaco/administração & dosagem , Sulindaco/análogos & derivados , Sulindaco/sangue , Comprimidos
10.
Biochem Pharmacol ; 83(6): 723-32, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22209867

RESUMO

Hormone-dependent estrogen receptor positive (ER+) breast cancers generally respond well to anti-estrogen therapy. Unfortunately, hormone-independent estrogen receptor negative (ER-) breast cancers are aggressive, respond poorly to current treatments and have a poor prognosis. New approaches and targets are needed for the prevention and treatment of ER- breast cancer. The NF-κB signaling pathway is strongly implicated in ER- tumor genesis, constituting a possible target for treatment. Hydrogen sulfide-releasing aspirin (HS-ASA), a novel and safer derivative of aspirin, has shown promise as an anti-cancer agent. We examined the growth inhibitory effect of HS-ASA via alterations in cell proliferation, cell cycle phase transitions, and apoptosis, using MDA-MB-231 cells as a model of triple negative breast cancer. Tumor xenografts in mice, representing human ER- breast cancer, were evaluated for reduction in tumor size, followed by immunohistochemical analysis for proliferation, apoptosis and expression of NF-κB. HS-ASA suppressed the growth of MDA-MB-231 cells by induction of G(0)/G(1) arrest and apoptosis, down-regulation of NF-κB, reduction of thioredoxin reductase activity, and increased levels reactive oxygen species. Tumor xenografts in mice, were significantly reduced in volume and mass by HS-ASA treatment. The decrease in tumor mass was associated with inhibition of cell proliferation, induction of apoptosis and decrease in NF-κB levels in vivo. HS-ASA has anti-cancer potential against ER- breast cancer and merits further study.


Assuntos
Antineoplásicos/farmacologia , Aspirina/análogos & derivados , Neoplasias da Mama/metabolismo , Proliferação de Células/efeitos dos fármacos , Sulfeto de Hidrogênio/metabolismo , NF-kappa B/antagonistas & inibidores , Receptores de Estrogênio/metabolismo , Tionas/farmacologia , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Aspirina/química , Aspirina/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Sulfeto de Hidrogênio/química , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Estrogênio/genética , Transdução de Sinais/efeitos dos fármacos , Tionas/química , Tiorredoxina Dissulfeto Redutase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Biochem Pharmacol ; 83(6): 715-22, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22222427

RESUMO

Hydrogen sulfide-releasing non-steroidal anti-inflammatory drugs (HS-NSAIDs) are an emerging novel class of compounds with significant anti-inflammatory properties. They consist of a traditional NSAID to which an H(2)S-releasing moiety is covalently attached. We examined the effects of four different HS-NSAIDs on the growth properties of eleven different human cancer cell lines of six different tissue origins. Human colon, breast, pancreatic, prostate, lung, and leukemia cancer cell lines were treated with HS-aspirin, -sulindac, -iburofen, -naproxen, and their traditional counterparts. HS-NSAIDs inhibited the growth of all cancer cell lines studied, with potencies of 28- to >3000-fold greater than that of their traditional counterparts. HS-aspirin (HS-ASA) was consistently the most potent. HS-NSAIDs inhibited cell proliferation, induced apoptosis, and caused G(0)/G(1) cell cycle block. Metabolism of HS-ASA by colon cells showed that the acetyl group of ASA was hydrolyzed rapidly, followed by hydrolysis of the ester bond linking the salicylate anion to the H(2)S releasing moiety, producing salicylic acid and ADT-OH from which H(2)S is released. In reconstitution studies, ASA and ADT-OH were individually less active than the intact HS-ASA towards cell growth inhibition. Additionally, the combination of these two components representing a fairly close approximation to the intact HS-ASA, was 95-fold less active than the intact HS-ASA for growth inhibition. Taken together, these results demonstrate that HS-NSAIDs have potential anti-growth activity against a wide variety of human cancer cells.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Antineoplásicos/farmacocinética , Aspirina/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Inibidores do Crescimento/farmacocinética , Sulfeto de Hidrogênio/metabolismo , Ibuprofeno/análogos & derivados , Naproxeno/análogos & derivados , Sulindaco/análogos & derivados , Tionas/farmacocinética , Anti-Inflamatórios não Esteroides/química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Aspirina/química , Aspirina/farmacocinética , Linhagem Celular Tumoral , Inibidores do Crescimento/química , Humanos , Sulfeto de Hidrogênio/química , Ibuprofeno/química , Ibuprofeno/farmacocinética , Naproxeno/química , Naproxeno/farmacocinética , Sulindaco/química , Sulindaco/farmacocinética , Tionas/química
12.
Biochem Pharmacol ; 83(6): 733-40, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22209714

RESUMO

The balance between phase-I carcinogen-activating and phase-II detoxifying xenobiotic metabolizing enzymes is critical to determining an individual's risk for cancer. We evaluated the effect of Hydrogen sulfide-releasing aspirin (HS-ASA) on xenobiotic metabolizing enzymes in HT-29 human colon and Hepa 1c1c7 mouse liver adenocarcinoma cells and in Wistar rats. HS-ASA inhibited the growth of HT-29 and Hepa 1c1c7 cells, with an IC(50) of 3.2 ± 0.3 µM and 4.2 ± 0.4 µM, respectively. The IC(50) for ASA in both cell lines was greater than 5000 µM at 24h. In these cell lines, HS-ASA caused a dose-dependent increase in activity and expression of the phase-II enzymes glutathione S-transferase (GST) and NAD(P)H:quinoneoxireductase (NQO1). It also caused an increase in UDP-glucuronosyltransferase (UGT) expression. The levels of CYP 1A1 a phase-I enzyme was increased by HS-ASA in both cell lines. Pretreatment of cells with NaF, an esterase inhibitor, abrogated the HS-ASA-mediated increases in NQO1 enzyme activity. HS-ASA increased the protein levels of the transcription factor Nrf2, which is a regulator of the phase-II enzymes. In vivo, HS-ASA at 100mg/kg/day had no effect on rat's weights; it induced a 3.4-fold and 1.4-fold increase in hepatic GST and NQO1 enzyme activities, respectively. GST and NQO1 protein levels were also increased. In contrast to that in cultured cells, CYP 1A1 protein levels were not altered in vivo. Therefore, HS-ASA induces phase-II enzymes, at least in part, through the action of H(2)S and by modulating Nrf2; these effects may be part of its mechanism of action against carcinogenesis.


Assuntos
Aspirina/análogos & derivados , Citocromo P-450 CYP1A1/metabolismo , Glutationa Transferase/metabolismo , Sulfeto de Hidrogênio/metabolismo , Desintoxicação Metabólica Fase II , Desintoxicação Metabólica Fase I , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias/enzimologia , Tionas/farmacologia , Animais , Aspirina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucuronosiltransferase/metabolismo , Células HT29 , Humanos , Concentração Inibidora 50 , Fígado/enzimologia , Masculino , Camundongos , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , Ratos , Ratos Wistar , Fluoreto de Sódio/farmacologia
13.
Cancer Prev Res (Phila) ; 4(3): 347-53, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21372034

RESUMO

SR13668, an orally active Akt pathway inhibitor, has demonstrated cancer chemopreventive potential in preclinical studies. To accelerate the clinical development of this promising agent, we designed and conducted the first-ever phase 0 chemoprevention trial to evaluate and compare the effects of food and formulation on SR13668 bioavailability. Healthy adult volunteers were randomly assigned to receive a single, 38-mg oral dose of SR13668 in one of five different formulations, with or without food. On the basis of existing animal data, SR13668 in a PEG400/Labrasol oral solution was defined as the reference formulation. Blood samples were obtained pre- and post-agent administration for pharmacokinetic analyses. Area under the plasma concentration-time curve (AUC(0-∞)) was defined as the primary endpoint. Data were analyzed and compared using established statistical methods for phase 0 trials with a limited sample size. Participants (n = 20) were rapidly accrued over a 5-month period. Complete pharmacokinetic data were available for 18 randomized participants. AUC(0-∞) values were highest in the fed state (range = 122-439 ng/mL × hours) and were statistically significantly different across formulations (P = 0.007), with Solutol HS15 providing the highest bioavailability. SR13668 time to peak plasma concentration (3 hours; range, 2-6 hours) and half-life were (11.2 ± 3.1 hours) were not formulation-dependent. Using a novel, highly efficient study design, we rapidly identified a lead formulation of SR13668 for further clinical testing. Our findings support application of the phase 0 trial paradigm to accelerate chemoprevention agent development.


Assuntos
Anticarcinógenos/farmacologia , Anticarcinógenos/uso terapêutico , Carbazóis/farmacologia , Quimioprevenção/métodos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Adulto , Área Sob a Curva , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Masculino , Oncologia/métodos , Pessoa de Meia-Idade , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Resultado do Tratamento
14.
Cancer Lett ; 298(2): 204-11, 2010 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-20674154

RESUMO

NO-NSAIDs are promising anticancer drugs, comprising an NSAID, an NO-releasing moiety, and a spacer linking them. Although the effect of NO-NSAIDs on a wide variety of signaling and other cellular mechanisms has been deciphered, a key question remains unanswered, that being the role of NO to the overall biological effect of these agents. It has been shown that NO can directly modify sulfhydryl residues of proteins through S-nitrosylation and induce apoptosis. We studied 3 NO-NSAIDs having a different NSAID, spacer, and NO-releasing moiety. In vitro: aspirin, NO-ASA, naproxen, and NO-naproxen inhibited HT-29 human colon cancer cell growth, the IC(50)s being >5000, 192±6, 2800±210 and 95±5µM at 24h, respectively. NO-Aspirin and NO-naproxen reduced NF-κB protein levels, and activated caspase-3 enzyme in a dose- and time-dependent manner. Based on the biotin switch assay, NO-ASA and NO-naproxen S-nitrosylated NF-κB p65 in a time-dependent manner. Pretreatment of the cells with carboxy-PTIO, abrogated the S-nitrosylation of NF-κB p65. In vivo: rats treated with NO-ASA, NONO-ASA, and NO-naproxen showed S-nitrosylation of NF-κB p65 in the stomach tissue, increases in plasma TNF-α, and reductions in mucosal PGE(2) levels. These data provide a mechanistic role for NO and a rational for the chemopreventive effects of NO-NSAIDs.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , NF-kappa B/metabolismo , S-Nitrosotióis/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/metabolismo , Aspirina/análogos & derivados , Aspirina/metabolismo , Aspirina/farmacologia , Western Blotting , Caspase 3/metabolismo , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Células HT29 , Humanos , Masculino , Naproxeno/análogos & derivados , Naproxeno/metabolismo , Naproxeno/farmacologia , Nitratos/metabolismo , Nitratos/farmacologia , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Ratos , Ratos Wistar , Estômago/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/sangue
15.
Biochem Pharmacol ; 79(12): 1759-71, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20188076

RESUMO

NO-donating aspirin (NO-ASA, para isomer) has been reported to exhibit strong growth inhibitory effect in Jurkat T-acute lymphoblastic leukemia (T-ALL) cells mediated in part by beta-catenin degradation and caspase activation, but the mechanism(s) still remains unclear. In this study, DNA oligoarrays with 263 genes were used to examine the gene expression profiles relating to stress and drug metabolism, and characterize the stress responses at IC(50) and subIC(50) concentrations of p-NO-ASA (20 and 10microM, respectively) in Jurkat T cells. A total of 22 genes related to heat shock response, apoptosis signaling, detoxifiers and Phase II enzymes, and regulators of cell growth were altered in expression by array analysis based on the expression fold change criteria of > or =1.5-fold or < or =0.65-fold. Real time quantitative RT-PCR confirmed that 20microM p-NO-ASA strongly upregulated the mRNA levels of two heat shock genes HSPA1A (41.5+/-7.01-fold) and HSPA6 (100.4+/-8.11-fold), and FOS (16.2+/-3.2-fold), moderately upregulated HSPH1 (1.71+/-0.43-fold), FMO4 (4.5+/-1.67-fold), CASP9 (1.77+/-0.03-fold), DDIT3 (5.6+/-0.51-fold), and downregulated NF-kappaB1 (0.54+/-0.01-fold) and CCND1 (0.69+/-0.06-fold). Protein levels of Hsp70, the product of HSPA1A, and fos were increased in p-NO-ASA-treated Jurkat T and HT-29 colon cancer cells in a dose-dependent manner. Silencing of Hsp70 enhanced the growth inhibitory effect of p-NO-ASA at low concentrations. The altered gene expression patterns by NO-ASA in Jurkat T cells suggest mechanisms for carcinogen metabolism, anti-proliferative activity and possible chemoprotective activity in T-ALL.


Assuntos
Antineoplásicos/farmacologia , Aspirina/análogos & derivados , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Leucemia de Células T/tratamento farmacológico , Antineoplásicos/química , Aspirina/química , Aspirina/farmacologia , Relação Dose-Resposta a Droga , Perfilação da Expressão Gênica , Inativação Gênica , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Células Jurkat , Estrutura Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Interferência de RNA
16.
Cancer Prev Res (Phila) ; 2(2): 161-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19196723

RESUMO

The statins are highly effective in lowering cholesterol by inhibiting 3-hydroxy-3-methylglutaryl CoA reductase. Recently, there has been conflicting epidemiologic data indicating that statins decrease the incidence of certain types of cancer, including breast cancer. Atorvastatin and lovastatin, statins with different lipophicilities, were administered in diet either as single agents or in combination with suboptimal doses of tamoxifen or the retinoid X receptor agonist bexarotene were evaluated for prevention of estrogen receptor-positive mammary cancers induced in the rat with methylnitrosourea. Atorvastatin (125 or 500 mg/kg diet) alone did not significantly alter cancer incidence or multiplicity. Suboptimal doses of tamoxifen (0.4 mg/kg diet) or bexarotene (80 mg/kg diet) reduced cancer multiplicity from 3.8 (control) to 2.9 and 0.9, respectively. Combining atorvastatin (500 mg/kg diet) with either of these effective agents minimally altered their efficacy. Although this dose of atorvastatin did not decrease serum triglyceride levels in control rats, it significantly decreased triglyceride levels that had been increased in bexarotene-treated rats. Experiments done with a second statin, lovastatin (100 and 400 mg/kg diet), yielded similar results: (a) limited activity when administered alone, (b) no obvious synergy with bexarotene, and (c) an ability to decrease bexarotene-induced increases in serum triglycerides. Thus, the statins had minimal activity in this model of mammary cancer in which approximately half of the cancers are mutated in the Ha Ras oncogene. Similarly, atorvastatin failed to alter the development of estrogen receptor-negative mammary carcinomas in a new animal model using bitransgenic mice (MMTV-Neu(+/-)/p53KO(+/-)), whereas bexarotene (250 mg/kg diet) was effective.


Assuntos
Anticolesterolemiantes/uso terapêutico , Modelos Animais de Doenças , Ácidos Heptanoicos/uso terapêutico , Lovastatina/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Pirróis/uso terapêutico , Alquilantes/toxicidade , Animais , Anticarcinógenos/farmacologia , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Atorvastatina , Bexaroteno , Proliferação de Células/efeitos dos fármacos , Dieta , Feminino , Genes erbB-2/fisiologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Metilnitrosoureia/toxicidade , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/metabolismo , Receptores X de Retinoides/metabolismo , Tamoxifeno/farmacologia , Tetra-Hidronaftalenos/farmacologia , Resultado do Tratamento , Proteína Supressora de Tumor p53/fisiologia
17.
Cancer Prev Res (Phila) ; 2(11): 951-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19892664

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAID) have been highly effective in preventing colon, urinary bladder, and skin cancer preclinically, and also in clinical trials of colon adenoma formation. However, certain NSAIDs cause gastrointestinal ulceration and may increase cardiovascular events. Naproxen seems to cause the lowest cardiovascular events of the common NSAIDs other than aspirin. Nitric oxide (NO)-naproxen was tested based on the finding that adding a NO group to NSAIDs may help alleviate GI toxicity. In the azoxymethane-induced rat colon aberrant crypt foci (ACF) model, naproxen administered at 200 and 400 ppm in the diet reduced mean ACFs in the colon by about 45% to 60%, respectively. NO-naproxen was likewise administered in the diet at roughly equimolar doses (300 and 600 ppm) and reduced total ACF by 20% to 40%, respectively. In the hydroxybutyl (butyl) nitrosamine rat urinary bladder cancer model, NO-naproxen was given at 183 or 550 ppm in the diet, and naproxen at 128 ppm. The NO-naproxen groups had 77% and 73% decreases, respectively, in the development of large urinary bladder tumors, whereas the 128 ppm naproxen group also showed a strong decrease (69%). If treatments were started 3 months after hydroxybutyl (butyl) nitrosamine, NO-naproxen (550 ppm) and naproxen (400 ppm) were also highly effective (86-94% decreases). In the methylnitrosourea-induced mammary cancer model in rats, NO-naproxen and naproxen showed nonsignificant inhibitions (12% and 24%) at 550 and 400 ppm, respectively. These data show that both naproxen and NO-naproxen are effective agents against urinary bladder and colon, but not mammary, carcinogenesis.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Neoplasias do Colo/prevenção & controle , Neoplasias Mamárias Experimentais/prevenção & controle , Naproxeno/análogos & derivados , Naproxeno/uso terapêutico , Doadores de Óxido Nítrico/uso terapêutico , Neoplasias da Bexiga Urinária/prevenção & controle , Alquilantes/toxicidade , Animais , Azoximetano/toxicidade , Butilidroxibutilnitrosamina/toxicidade , Carcinógenos/toxicidade , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Dieta , Feminino , Masculino , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/metabolismo , Metilnitrosoureia/toxicidade , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/prevenção & controle , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA