Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Mol Pharm ; 21(7): 3310-3320, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38856116

RESUMO

Siponimod is a promising agent for the inhibition of ocular neovascularization in diabetic retinopathy and age-related macular degeneration. Siponimod's development for ophthalmological application is hindered by the limited information available on the drug's solubility, stability, ocular pharmacokinetics (PK), and toxicity in vivo. In this study, we investigated the aqueous stability of siponimod under stress conditions (up to 60 °C) and its degradation behavior in solution. Additionally, siponimod's ocular PK and toxicity were investigated using intravitreal injection of two different doses (either 1300 or 6500 ng) in an albino rabbit model. Siponimod concentration was quantified in the extracted vitreous, and the PK parameters were calculated. The drug half-life after administration of the low and high doses was 2.8 and 3.9 h, respectively. The data obtained in vivo was used to test the ability of published in silico models to predict siponimod's PK accurately. Two models that correlated siponimod's molecular descriptors with its elimination from the vitreous closely predicted the half-life. Furthermore, 24 h and 7 days after intravitreal injections, the retinas showed no signs of toxicity. This study provides important information necessary for the formulation and development of siponimod for ophthalmologic applications. The short half-life of siponimod necessitates the development of a sustained drug delivery system to maintain therapeutic concentrations over an extended period, while the lack of short-term ocular toxicity observed in the retinas of siponimod-treated rabbits supports possible clinical use.


Assuntos
Azetidinas , Injeções Intravítreas , Animais , Coelhos , Azetidinas/farmacocinética , Azetidinas/administração & dosagem , Meia-Vida , Corpo Vítreo/efeitos dos fármacos , Corpo Vítreo/metabolismo , Masculino , Retina/efeitos dos fármacos , Retina/metabolismo , Olho/efeitos dos fármacos , Olho/metabolismo , Retinopatia Diabética/tratamento farmacológico , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/toxicidade , Solubilidade , Degeneração Macular/tratamento farmacológico , Compostos de Benzil
2.
Cancer Chemother Pharmacol ; 90(1): 97-104, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35598186

RESUMO

INTRODUCTION: The combination of vemurafenib, a proto-oncogene B-Raf inhibitor (BRAFi) and cobimetinib, an inhibitor of mitogen-activated protein kinase kinase (MEKi) has shown to improve survival in patients with BRAF V600-mutated melanoma. BRAF mutations are also frequently detected driver mutations in other tumor types, including thyroid carcinoma. Since thyroid carcinoma is not a labeled indication for BRAF/MEKi, a cohort for patients with BRAF V600-mutated thyroid carcinoma was opened within the Drug Rediscovery Protocol (DRUP), a national ongoing pan-cancer multi-drug trial, in which patients receive off-label treatment with approved drugs based on their molecular tumor profile. RESULTS: Here, we present two patients with BRAF-mutated thyroid carcinoma, who were successfully treated with vemurafenib/cobimetinib administered via a feeding tube. Plasma concentrations of vemurafenib and cobimetinib were determined. A partial response was observed in both patients, but they experienced significant toxicity. CONCLUSION: Our cases show that vemurafenib/cobimetinib treatment is effective in BRAF V600-mutated thyroid carcinoma, also when administered via a feeding tube. Although serious side effects occurred in both patients, we hypothesize that this was not attributable to the administration route. Therefore, administration of vemurafenib/cobimetinib by feeding tube is feasible and effective. TRIAL REGISTRATION: Clinical trial identification: NCT02925234.


Assuntos
Antineoplásicos , Azetidinas , Piperidinas , Neoplasias Cutâneas , Neoplasias da Glândula Tireoide , Vemurafenib , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Azetidinas/efeitos adversos , Azetidinas/farmacocinética , Humanos , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/genética , Vemurafenib/efeitos adversos , Vemurafenib/farmacocinética
3.
Clin Pharmacol Drug Dev ; 10(2): 144-152, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32696585

RESUMO

Cobimetinib is a kinase inhibitor indicated for use in combination with vemurafenib for treatment of unresectable/metastatic melanoma with specific BRAF mutations. Cobimetinib is extensively metabolized in liver; thus, patients with hepatic impairment (HI) might have increased cobimetinib exposure. In this study, we investigated the impact of HI on the pharmacokinetics (PK) and safety of cobimetinib. Subjects with normal hepatic function and mild to severe HI were enrolled. All subjects received a single oral dose of 10 mg cobimetinib, and serial blood samples were collected at specified times. Cobimetinib PK in subjects with mild and moderate HI was similar to that in those with normal liver function. However, subjects with severe HI, on average, showed ∼30% lower total AUC0-∞ and ∼2-fold higher unbound AUC0-∞ compared with those with normal hepatic function. These exposure differences can be explained by lower albumin levels observed in subjects with severe HI, the strong correlation between albumin level and the unbound fraction and the general PK variability of cobimetinib. In addition, previous studies with cobimetinib showed a lack of an exposure-response relationship for efficacy and safety. Therefore, collectively, our results suggest that the starting dose for patients with hepatic impairment can be the same as that for those with normal hepatic function.


Assuntos
Azetidinas/farmacocinética , Hepatopatias/fisiopatologia , Piperidinas/farmacocinética , Inibidores de Proteínas Quinases/farmacocinética , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Área Sob a Curva , Azetidinas/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Piperidinas/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Índice de Gravidade de Doença
4.
Invest New Drugs ; 39(1): 163-174, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32737717

RESUMO

BACKGROUND: This Phase Ib study explored combination dosing of the allosteric MEK1/2 inhibitor cobimetinib and the ATP-competitive pan-AKT inhibitor ipatasertib. METHODS: Patients with advanced solid tumors were enrolled to two dose escalation arms, each using a 3 + 3 design in 28-day cycles. In Arm A, patients received concurrent cobimetinib and ipatasertib on days 1-21. In Arm B, cobimetinib was administered intermittently with ipatasertib for 21 days. Primary objectives evaluated dose-limiting toxicities (DLTs), maximum tolerated doses (MTD), and the recommended Phase II dose (RP2D). Secondary objectives included analysis of pharmacokinetic parameters, MAPK and PI3K pathway alterations, changes in tissue biomarkers, and preliminary anti-tumor efficacy. Expansion cohorts included patients with PTEN-deficient triple-negative breast cancer and endometrial cancer. RESULTS: Among 66 patients who received ≥1 dose of study drug, all experienced an adverse event (AE). Although no DLTs were reported, 6 patients experienced Cycle 1 DLT-equivalent AEs. The most common treatment-related AEs were diarrhea, nausea, vomiting, dermatitis acneiform, and fatigue. Thirty-five (53%) patients experienced drug-related AEs of ≥ grade 3 severity. Cobimetinb/ipatasertib MTDs were 60/200 mg on Arm A and 150/300 mg on Arm B; the latter was chosen as the RP2D. No pharmacokinetic interactions were identified. Biomarker analyses indicated pathway blockade and increases in IFNγ and PD-L1 gene expression following the combination. Three patients with endometrial or ovarian cancer achieved partial response, all with PTEN-low disease and two with tumor also harboring KRAS mutation. CONCLUSION: There was limited tolerability and efficacy for this MEK and AKT inhibitor combination. Nonetheless, pharmacodynamic analyses indicated target engagement and suggest rationale for further exploration of cobimetinib or ipatasertib in combination with other anticancer agents. ClinicalTrials.gov identifier: NCT01562275.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Azetidinas/farmacologia , Azetidinas/uso terapêutico , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Azetidinas/efeitos adversos , Azetidinas/farmacocinética , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Piperazinas/efeitos adversos , Piperazinas/farmacocinética , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Pirimidinas/efeitos adversos , Pirimidinas/farmacocinética
5.
Int J Nanomedicine ; 15: 5561-5571, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32801704

RESUMO

PURPOSE: Platinum/paclitaxel-based chemotherapy is the strategy for ovarian cancer, but chemoresistance, inherent or acquired, occurs and hinders therapy. Therefore, further understanding of the mechanisms of drug resistance and adoption of novel therapeutic strategies are urgently needed. METHODS: In this study, we report that sphingosine-1-phosphate receptor-1 (S1PR1)-mediated chemoresistance for ovarian cancer. Then we developed nanoparticles with a hydrophilic PEG2000 chain and a hydrophobic DSPE and biodegradable CaP (calcium ions and phosphate ions) shell with pH sensitivity as a delivery system (CaP-NPs) to carry BAF312, a selective antagonist of S1PR1 (BAF312@CaP-NPs), to overcome the cisplatin (DDP) resistance of the ovarian cancer cell line SKOV3DR. RESULTS: We found that S1PR1 affected acquired chemoresistance in ovarian cancer by increasing the phosphorylated-signal transduction and activators of transcription 3 (P-STAT3) level. The mean size and zeta potential of BAF312@CaP-NPs were 116 ± 4.341 nm and -9.67 ± 0.935 mV, respectively. The incorporation efficiency for BAF312 in the CaP-NPs was 76.1%. The small size of the nanoparticles elevated their enrichment in the tumor, and the degradable CaP shell with smart pH sensitivity of the BAF312@CaP-NPs ensured the release of BAF312 in the acidic tumor niche. BAF312@CaP-NPs caused substantial cytotoxicity in DDP-resistant ovarian cancer cells by downregulating S1PR1 and P-STAT3 levels. CONCLUSION: We found that BAF312@CaP-NPs act as an effective and selective delivery system for overcoming S1PR1-mediated chemoresistance in ovarian carcinoma by inhibiting S1PR1 and P-STAT3.


Assuntos
Azetidinas/administração & dosagem , Compostos de Benzil/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Nanopartículas/química , Neoplasias Ovarianas/tratamento farmacológico , Fator de Transcrição STAT3/antagonistas & inibidores , Receptores de Esfingosina-1-Fosfato/genética , Azetidinas/farmacocinética , Compostos de Benzil/farmacocinética , Fosfatos de Cálcio/química , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação/efeitos dos fármacos , Polietilenoglicóis/química , Fator de Transcrição STAT3/metabolismo , Receptores de Esfingosina-1-Fosfato/antagonistas & inibidores , Receptores de Esfingosina-1-Fosfato/metabolismo
6.
Eur J Pharmacol ; 885: 173505, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32861662

RESUMO

Pharmacological modulation of the Janus kinase (JAK) family has achieved clinically meaningful therapeutic outcomes for the treatment of inflammatory and hematopoietic diseases. Several JAK1 selective compounds are being investigated clinically to determine their anti-inflammatory potential. We used recombinant enzymes and primary human lymphocytes to assess the JAK1 specificity of itacitinib (INCB039110) and study inhibition of signal transducers and activators of transcription (STAT) signaling. Rodent models of arthritis and inflammatory bowel disease were subsequently explored to elucidate the efficacy of orally administered itacitinib on inflammatory pathogenesis. Itacitinib is a potent and selective JAK1 inhibitor when profiled against the other JAK family members. Upon oral administration in rodents, itacitinib achieved dose-dependent pharmacokinetic exposures that highly correlated with STAT3 pharmacodynamic pathway inhibition. Itacitinib ameliorated symptoms and pathology of established experimentally-induced arthritis in a dose-dependent manner. Furthermore, itacitinib effectively delayed disease onset, reduced symptom severity, and accelerated recovery in three distinct mouse models of inflammatory bowel disease. Low dose itacitinib administered via cannula directly into the colon was highly efficacious in TNBS-induced colitis but with minimal systemic drug exposure, suggesting localized JAK1 inhibition is sufficient for disease amelioration. Itacitinib treatment in an acute graft-versus-host disease (GvHD) model rapidly reduced inflammatory markers within lymphocytes and target tissue, resulting in a marked improvement in disease symptoms. This is the first manuscript describing itacitinib as a potent and selective JAK1 inhibitor with anti-inflammatory activity across multiple preclinical disease models. These data support the scientific rationale for ongoing clinical trials studying itacitinib in select GvHD patient populations.


Assuntos
Azetidinas/farmacologia , Inflamação/tratamento farmacológico , Ácidos Isonicotínicos/farmacologia , Janus Quinase 1/antagonistas & inibidores , Animais , Artrite Experimental/tratamento farmacológico , Azetidinas/farmacocinética , Azetidinas/uso terapêutico , Quimiocina CCL2/efeitos dos fármacos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Relação Dose-Resposta a Droga , Doença Enxerto-Hospedeiro/tratamento farmacológico , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Ácidos Isonicotínicos/farmacocinética , Ácidos Isonicotínicos/uso terapêutico , Linfócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Cultura Primária de Células , Ratos , Ratos Endogâmicos Lew , Fatores de Transcrição STAT/efeitos dos fármacos , Fator de Transcrição STAT3/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos
7.
J Med Chem ; 63(15): 8584-8607, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32667798

RESUMO

The C-C chemokine receptor 4 (CCR4) is broadly expressed on regulatory T cells (Treg) as well as other circulating and tissue-resident T cells. Treg can be recruited to the tumor microenvironment (TME) through the C-C chemokines CCL17 and CCL22. Treg accumulation in the TME has been shown to dampen the antitumor immune response and is thought to be an important driver in tumor immune evasion. Preclinical and clinical data suggest that reducing the Treg population in the TME can potentiate the antitumor immune response of checkpoint inhibitors. We have developed small-molecule antagonists of CCR4, featuring a novel piperidinyl-azetidine motif, that inhibit the recruitment of Treg into the TME and elicit antitumor responses as a single agent or in combination with an immune checkpoint blockade. The discovery of these potent, selective, and orally bioavailable CCR4 antagonists, and their activity in in vitro and in vivo models, is described herein.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Azetidinas/química , Azetidinas/farmacologia , Receptores CCR4/antagonistas & inibidores , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Azetidinas/farmacocinética , Azetidinas/uso terapêutico , Linhagem Celular Tumoral , Cães , Humanos , Macaca fascicularis , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Piperidinas/química , Piperidinas/farmacocinética , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Receptores CCR4/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
8.
Drug Saf ; 43(8): 711-725, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32367507

RESUMO

The introduction of novel, small-molecule Janus kinase inhibitors namely tofacitinib, baricitinib and upadacitinib has provided an alternative treatment option for patients with rheumatoid arthritis outside of traditional drugs and expensive biologics. This review aimed to critically assess the drug-drug interaction potential of tofacitinib, baricitinib and upadacitinib and provide a balanced perspective for choosing the most appropriate Janus kinase inhibitor based on the needs of patients with rheumatoid arthritis including co-medications and renal/hepatic impairment status. Based on the critical assessment, all three approved Janus kinase inhibitors generally provide a favourable opportunity for co-prescription with a plethora of drugs. While cytochrome P450 3A4-related inhibition or induction altered the exposures (area under the curve) of tofacitinib and upadacitinib, it did not impact the exposure of baricitinib. Transporter drug-drug interaction studies revealed that the disposition of baricitinib was altered with certain transporter inhibitors as compared with either tofacitinib or upadacitinib. Adjustment of tofacitinib or baricitinib dosages but not that of upadacitinib is required with the progression of renal impairment from a mild to a severe condition. While the dosage of tofacitinib needs to be adjusted for patients with moderate hepatic impairment status, it is not the case for either baricitinib or upadacitinib. Assessment of the drug-drug interaction potential suggests that tofacitinib, baricitinib and upadacitinib generally show a favourable disposition with no perpetrator activity; however, as victim drugs, they show subtle pharmacokinetic differences that may be considered during polypharmacy. Moreover, careful choice of the three drugs could be made in patients with rheumatoid arthritis with varying degrees of renal/hepatic impairments.


Assuntos
Antirreumáticos/efeitos adversos , Azetidinas/efeitos adversos , Interações Medicamentosas , Compostos Heterocíclicos com 3 Anéis/efeitos adversos , Janus Quinases/antagonistas & inibidores , Farmacocinética , Piperidinas/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Purinas/efeitos adversos , Pirazóis/efeitos adversos , Pirimidinas/efeitos adversos , Sulfonamidas/efeitos adversos , Animais , Antirreumáticos/farmacocinética , Antirreumáticos/uso terapêutico , Azetidinas/farmacocinética , Azetidinas/toxicidade , Compostos Heterocíclicos com 3 Anéis/farmacocinética , Compostos Heterocíclicos com 3 Anéis/uso terapêutico , Humanos , Piperidinas/farmacocinética , Piperidinas/toxicidade , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Purinas/farmacocinética , Purinas/toxicidade , Pirazóis/farmacocinética , Pirazóis/toxicidade , Pirimidinas/farmacocinética , Pirimidinas/toxicidade , Sulfonamidas/farmacocinética , Sulfonamidas/toxicidade
9.
Biol Pharm Bull ; 43(2): 325-333, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32009119

RESUMO

Colony stimulating factor 1 (CSF1) receptor (CSF1R) is a receptor protein-tyrosine kinase specifically expressed in monocyte-lineage cells, such as monocytes and macrophages. In this study, we characterized the pharmacological properties of an azetidine compound, JTE-952 ((2S)-3-{[2-({3-[4-(4-cyclopropylbenzyloxy)-3-methoxyphenyl]azetidine-1-yl}carbonyl)pyridin-4-yl]methoxy}propane-1,2-diol), which is a novel CSF1R tyrosine kinase inhibitor. JTE-952 potently inhibited human CSF1R kinase activity, with a half maximal inhibitory concentration of 11.1 nmol/L, and inhibited the phosphorylation of CSF1R in human macrophages and the CSF1-induced proliferation of human macrophages. It also inhibited human tropomyosin-related kinase A activity, but only at concentrations 200-fold higher than that required to inhibit the activity of CSF1R in inducing the proliferation of human macrophages. JTE-952 displayed no marked inhibitory activity against other kinases. JTE-952 potently inhibited lipopolysaccharide-induced proinflammatory cytokine production by human macrophages and in whole blood. JTE-952 (≥3 mg/kg given orally) also significantly attenuated the CSF1-induced priming of lipopolysaccharide-induced tumor necrosis factor-alpha production in mice and arthritis severity in a mouse model of collagen-induced arthritis. Taken together, these results indicate that JTE-952 is an orally available compound with potent and specific inhibitory activity against CSF1R, both in vitro and in vivo. JTE-952 is a potentially clinically useful agent for various human inflammatory diseases, including rheumatoid arthritis.


Assuntos
Azetidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Animais , Artrite Experimental/tratamento farmacológico , Azetidinas/farmacocinética , Células Cultivadas , Citocinas/sangue , Citocinas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Ratos Endogâmicos Lew , Receptor trkA/metabolismo
10.
Invest New Drugs ; 38(2): 419-432, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31020608

RESUMO

Purpose We investigated the combination of the MEK inhibitor, cobimetinib, and the pan-PI3K inhibitor, pictilisib, in an open-label, phase Ib study. Experimental Design Patients with advanced solid tumors were enrolled in 3 dose escalation schedules: (1) both agents once-daily for 21-days-on 7-days-off ("21/7"); (2) intermittent cobimetinib and 21/7 pictilisib ("intermittent"); or (3) both agents once-daily for 7-days-on 7-days-off ("7/7"). Starting doses for the 21/7, intermittent, and 7/7 schedules were 20/80, 100/130, and 40/130 mg of cobimetinib/pictilisib, respectively. Nine indication-specific expansion cohorts interrogated the recommended phase II dose and schedule. Results Of 178 enrollees (dose escalation: n = 98), 177 patients were dosed. The maximum tolerated doses for cobimetinib/pictilisib (mg) were 40/100, 125/180, and not reached, for the 21/7, intermittent, and 7/7 schedules, respectively. Six dose-limiting toxicities included grade 3 (G3) elevated lipase, G4 elevated creatine phosphokinase, and G3 events including fatigue concurrent with a serious adverse event (SAE) of diarrhea, decreased appetite, and SAEs of hypersensitivity and dehydration. Common drug-related adverse events included nausea, fatigue, vomiting, decreased appetite, dysgeusia, rash, and stomatitis. Pharmacokinetic parameters of the drugs used in combination were unaltered compared to monotherapy exposures. Confirmed partial responses were observed in patients with BRAF-mutant melanoma (n = 1) and KRAS-mutant endometrioid adenocarcinoma (n = 1). Eighteen patients remained on study ≥6 months. Biomarker data established successful blockade of MAP kinase (MAPK) and PI3K pathways. The metabolic response rate documented by FDG-PET was similar to that observed with cobimetinib monotherapy. Conclusions Cobimetinib and pictilisib combination therapy in patients with solid tumors had limited tolerability and efficacy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Azetidinas/administração & dosagem , Indazóis/administração & dosagem , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase/administração & dosagem , Piperidinas/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Sulfonamidas/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Azetidinas/efeitos adversos , Azetidinas/farmacocinética , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe I de Fosfatidilinositol 3-Quinases/genética , Feminino , GTP Fosfo-Hidrolases/genética , Humanos , Indazóis/efeitos adversos , Indazóis/farmacocinética , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Neoplasias/genética , Neoplasias/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/efeitos adversos , Inibidores de Fosfoinositídeo-3 Quinase/farmacocinética , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Proto-Oncogênicas p21(ras)/genética , Sulfonamidas/efeitos adversos , Sulfonamidas/farmacocinética , Resultado do Tratamento , Adulto Jovem
11.
Bioorg Med Chem Lett ; 29(16): 2090-2093, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31311734

RESUMO

Phenolic groups are responsible for the high clearance and low oral bioavailability of the estrogen receptor alpha (ERα) clinical candidate GDC-0927. An exhaustive search for a backup molecule with improved pharmacokinetic (PK) properties identified several metabolically stable analogs, although in general at the expense of the desired potency and degradation efficiency. C-8 hydroxychromene 30 is the first example of a phenol-containing chromene that not only maintained excellent potency but also exhibited 10-fold higher oral exposure in rats. The improved in vivo clearance in rat was hypothesized to be the result of C-8 hydroxy group being sterically protected from glucuronide conjugation. The excellent potency underscores the possibility of replacing the presumed indispensable phenolic group at C-6 or C-7 of the chromene core. Co-crystal structures were obtained to highlight the change in key interactions and rationalize the retained potency.


Assuntos
Azetidinas/farmacologia , Receptor alfa de Estrogênio/metabolismo , Flavonoides/farmacologia , Administração Oral , Animais , Azetidinas/administração & dosagem , Azetidinas/metabolismo , Azetidinas/farmacocinética , Cristalografia por Raios X , Descoberta de Drogas , Estabilidade de Medicamentos , Flavonoides/administração & dosagem , Flavonoides/metabolismo , Flavonoides/farmacocinética , Humanos , Células MCF-7 , Microssomos Hepáticos/metabolismo , Ratos , Estereoisomerismo , Relação Estrutura-Atividade
12.
Artigo em Inglês | MEDLINE | ID: mdl-29544202

RESUMO

A sensitive and selective method of high performance liquid chromatography (HPLC) coupled to tandem mass spectrometry (MS/MS) has been developed for the simultaneous quantification of six anticancer protein kinase inhibitors (PKIs), dabrafenib, trametinib, vemurafenib, cobimetinib, pazopanib, regorafenib, and two active metabolites (regorafenib-M2 and regorafenib-M5) in human plasma. Plasma protein precipitation with methanol enables the sample extraction of 100 µL aliquot of plasma. Analytes are detected by electrospray triple-stage quadrupole mass spectrometry and quantified using the calibration curves with stable isotope-labeled internal standards. The method was validated based on FDA recommendations, including assessment of extraction yield (74-104%), matrix effects, analytical recovery (94-104%) with low variability (<15%). The method is sensitive (lower limits of quantification within 1 to 200 ng/mL), accurate (intra- and inter-assay bias: -0.3% to +12.7%, and -3.2% to +6.3%, respectively) and precise (intra- and inter-assay CVs within 0.7-7.3% and 2.5-8.0%, respectively) over the clinically relevant concentration range (upper limits of quantification 500 to 100,000 ng/mL). This method is applied in our laboratory for both clinical research programs and routine therapeutic drug monitoring service of PKIs.


Assuntos
Antineoplásicos/sangue , Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas em Tandem/métodos , Administração Oral , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacocinética , Azetidinas/administração & dosagem , Azetidinas/sangue , Azetidinas/química , Azetidinas/farmacocinética , Criança , Humanos , Imidazóis/administração & dosagem , Imidazóis/sangue , Imidazóis/química , Imidazóis/farmacocinética , Indazóis , Indóis/administração & dosagem , Indóis/sangue , Indóis/química , Indóis/farmacocinética , Limite de Detecção , Modelos Lineares , Oximas/administração & dosagem , Oximas/sangue , Oximas/química , Oximas/farmacocinética , Compostos de Fenilureia/administração & dosagem , Compostos de Fenilureia/sangue , Compostos de Fenilureia/química , Compostos de Fenilureia/farmacocinética , Piperidinas/administração & dosagem , Piperidinas/sangue , Piperidinas/química , Piperidinas/farmacocinética , Piridinas/administração & dosagem , Piridinas/sangue , Piridinas/química , Piridinas/farmacocinética , Piridonas/administração & dosagem , Piridonas/sangue , Piridonas/química , Piridonas/farmacocinética , Pirimidinas/administração & dosagem , Pirimidinas/sangue , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinonas/administração & dosagem , Pirimidinonas/sangue , Pirimidinonas/química , Pirimidinonas/farmacocinética , Reprodutibilidade dos Testes
13.
J Pharmacol Exp Ther ; 365(2): 301-313, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29491038

RESUMO

Methionine aminopeptidase 2 (MetAP2) inhibition is a promising approach to treating diabetes, obesity, and associated metabolic disorders. Beloranib, a MetAP2 inhibitor previously investigated for treatment of Prader-Willi syndrome, was associated with venous thrombotic adverse events likely resulting from drug effects on vascular endothelial cells (ECs). Here, we report the pharmacological characterization of ZGN-1061, a novel MetAP2 inhibitor being investigated for treatment of diabetes and obesity. Four weeks of subcutaneous administration of ZGN-1061 to diet-induced obese (DIO) insulin-resistant mice produced a 25% reduction in body weight, primarily due to reduced fat mass, that was comparable to beloranib. ZGN-1061 also produced improvements in metabolic parameters, including plasma glucose and insulin, and, in HepG2 cells, initiated gene changes similar to beloranib that support observed in vivo pharmacodynamics. In vitro studies in ECs demonstrated that ZGN-1061 effects on EC proliferation and coagulation proteins were greatly attenuated, or absent, relative to beloranib, due to lower intracellular drug concentrations, shorter half-life of inhibitor-bound MetAP2 complex, and reduced cellular enzyme inhibition. In dogs, ZGN-1061 was more rapidly absorbed and cleared, with a shorter half-life than beloranib. Unlike beloranib, ZGN-1061 did not increase coagulation markers in dogs, and ZGN-1061 had a greatly improved safety profile in rats relative to beloranib. In conclusion, ZGN-1061 and beloranib demonstrated similar efficacy in a mouse model of obesity, while ZGN-1061 had a markedly improved safety profile in multiple in vitro and in vivo models. The lower duration of exposure characteristic of ZGN-1061 is expected to provide a meaningfully enhanced clinical safety profile.


Assuntos
Aminopeptidases/antagonistas & inibidores , Azetidinas/efeitos adversos , Azetidinas/farmacologia , Metaloendopeptidases/antagonistas & inibidores , Morfolinas/efeitos adversos , Morfolinas/farmacologia , Obesidade/tratamento farmacológico , Segurança , Animais , Azetidinas/farmacocinética , Azetidinas/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Cinamatos/farmacocinética , Cinamatos/farmacologia , Cicloexanos/farmacocinética , Cicloexanos/farmacologia , Cães , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Compostos de Epóxi/farmacocinética , Compostos de Epóxi/farmacologia , Feminino , Células Hep G2 , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfolinas/farmacocinética , Morfolinas/uso terapêutico , Obesidade/enzimologia , Ratos , Sesquiterpenos/farmacocinética , Sesquiterpenos/farmacologia , Distribuição Tecidual
14.
Clin Pharmacol Ther ; 103(2): 188-192, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29193027

RESUMO

The utility of animal models in understanding drug disposition and extrapolating to human in a direct manner is limited, confounded by differences in enzyme and transporter substrate specificity and expression between species. Conversely, certain clinical pharmacology or mechanistic studies cannot be conducted for ethical and practical reasons. Thus, humanized animal models could be useful to bridge the gap between preclinical and clinical pharmacology and gain relevant insight into the determinants of drug disposition and drug-drug interaction (DDI).


Assuntos
Desenvolvimento de Medicamentos/métodos , Descoberta de Drogas/métodos , Medicina Baseada em Evidências/métodos , Modelos Animais , Farmacocinética , Pesquisa Translacional Biomédica/métodos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Azetidinas/administração & dosagem , Azetidinas/farmacocinética , Mineração de Dados , Bases de Dados Factuais , Desenvolvimento de Medicamentos/tendências , Descoberta de Drogas/tendências , Interações Medicamentosas , Medicina Baseada em Evidências/tendências , Previsões , Humanos , Aprendizagem , Camundongos Transgênicos , Modelos Teóricos , Segurança do Paciente , Piperidinas/administração & dosagem , Piperidinas/farmacocinética , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacocinética , Medição de Risco , Especificidade da Espécie , Pesquisa Translacional Biomédica/tendências
15.
Ugeskr Laeger ; 179(44)2017 Oct 30.
Artigo em Dinamarquês | MEDLINE | ID: mdl-29084618

RESUMO

Rheumatoid arthritis (RA) is a chronic inflammatory joint disease. The introduction of a new class of disease-modifying anti-rheumatic drugs, which work by inhibiting the Janus kinase-signal transducer and activator of transcription (JAK/STAT) pathway, has led to new possibilities for achieving remission of RA. Tofacitinib and baricitinib are both JAK/STAT inhibitors, which have shown efficacy in line with anti-tumour necrosis factor treatment. The side effects seem manageable, and up to now only increased risk of herpes zoster has raised consideration. JAK/STAT inhibitors create new possibilities for reaching low disease activity or remission for patients with RA.


Assuntos
Antirreumáticos , Artrite Reumatoide/tratamento farmacológico , Inibidores de Janus Quinases , Antirreumáticos/efeitos adversos , Antirreumáticos/farmacocinética , Antirreumáticos/uso terapêutico , Artrite Reumatoide/fisiopatologia , Azetidinas/efeitos adversos , Azetidinas/farmacocinética , Azetidinas/uso terapêutico , Herpes Zoster/induzido quimicamente , Humanos , Inibidores de Janus Quinases/efeitos adversos , Inibidores de Janus Quinases/farmacocinética , Inibidores de Janus Quinases/uso terapêutico , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Piperidinas/uso terapêutico , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Purinas , Pirazóis , Pirimidinas/efeitos adversos , Pirimidinas/farmacocinética , Pirimidinas/uso terapêutico , Pirróis/efeitos adversos , Pirróis/farmacocinética , Pirróis/uso terapêutico , Sulfonamidas/efeitos adversos , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapêutico
16.
Clin Transl Sci ; 10(6): 509-519, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28749581

RESUMO

Baricitinib, an oral selective Janus kinase 1 and 2 inhibitor, undergoes active renal tubular secretion. Baricitinib was not predicted to inhibit hepatic and renal uptake and efflux drug transporters, based on the ratio of the unbound maximum eliminating-organ inlet concentration and the in vitro half-maximal inhibitory concentrations (IC50 ). In vitro, baricitinib was a substrate for organic anion transporter (OAT)3, multidrug and toxin extrusion protein (MATE)2-K, P-glycoprotein (P-gp), and breast cancer resistance protein (BCRP). Probenecid, a strong OAT3 inhibitor, increased the area under the concentration-time curve from time zero to infinity (AUC[0-∞] ) of baricitinib by twofold and decreased renal clearance to 69% of control in healthy subjects. Physiologically based pharmacokinetic (PBPK) modeling reproduced the renal clearance of baricitinib and the inhibitory effect of probenecid using the in vitro IC50 value of 4.4 µM. Using ibuprofen and diclofenac in vitro IC50 values of 4.4 and 3.8 µM toward OAT3, 1.2 and 1.0 AUC(0-∞) ratios of baricitinib were predicted. These predictions suggest clinically relevant drug-drug interactions (DDIs) with ibuprofen and diclofenac are unlikely.


Assuntos
Azetidinas/farmacologia , Proteínas de Membrana Transportadoras/metabolismo , Sulfonamidas/farmacologia , Adulto , Área Sob a Curva , Azetidinas/sangue , Azetidinas/farmacocinética , Interações Medicamentosas , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Purinas , Pirazóis , Sulfonamidas/sangue , Sulfonamidas/farmacocinética , Fatores de Tempo , Adulto Jovem
17.
Ann Pharmacother ; 51(2): 146-153, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27701080

RESUMO

OBJECTIVE: To review and summarize data on cobimetinib, which was approved by the US Food and Drug Administration (FDA) in November 2015 for use in combination with vemurafenib for unresectable or metastatic melanoma with a BRAFV600E or V600K mutation. DATA SOURCES: A literature search using PubMed was conducted using the terms cobimetinib, MEK inhibitor, and melanoma from January 2000 to June 2016. STUDY SELECTION AND DATA EXTRACTION: The literature search was confined to human studies published in English. Trials of cobimetinib for melanoma were prioritized. DATA SYNTHESIS: Cobimetinib is a reversible inhibitor of MEK1 and MEK2. Its FDA approval was based on a phase III, randomized trial of vemurafenib monotherapy (n = 248) or vemurafenib and cobimetinib (n = 247) in unresectable stage IIIC or IV melanoma with a BRAFV600 mutation. Cobimetinib was administered as 60 mg orally daily for 21 days/7 days off, whereas vemurafenib was administered as 960 mg twice daily. Vemurafenib and cobimetinib were associated with an objective response rate of 68%, and median progression-free survival of 9.9 months. The overall survival was not reached at the time of first interim analysis. Clinically relevant grade ≥3 adverse events were diarrhea (6%), rash (6%), photosensitivity (2%), elevated liver function tests (LFTs) (8%-12%), increased creatine kinase (11%), and retinal detachment (3%). CONCLUSION: Cobimetinib combined with vemurafenib is an alternative BRAF/MEK inhibitor therapy for unresectable or metastatic melanoma with BRAFV600 mutation. The role of cobimetinib in melanoma and other solid tumors is likely to expand as the results from ongoing studies become available.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Azetidinas/uso terapêutico , Indóis/uso terapêutico , Melanoma/tratamento farmacológico , Piperidinas/uso terapêutico , Sulfonamidas/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Azetidinas/administração & dosagem , Azetidinas/farmacocinética , Ensaios Clínicos como Assunto , Intervalo Livre de Doença , Humanos , Indóis/administração & dosagem , Indóis/farmacocinética , Melanoma/genética , Melanoma/patologia , Mutação , Piperidinas/administração & dosagem , Piperidinas/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacocinética , Estados Unidos , United States Food and Drug Administration , Vemurafenib
18.
Invest New Drugs ; 34(5): 604-13, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27424159

RESUMO

Objective Cobimetinib, a MEK1/2 inhibitor, was administered to patients with advanced solid tumors to assess safety, pharmacokinetics, pharmacodynamics, and anti-tumor activity. Methods For dose-escalation, a 3 + 3 design was used. Oral cobimetinib was administered once daily on a 21-day on/7-day off (21/7) or a 14-day on/14-day off (14/14) schedule. Serial plasma samples were collected for pharmacokinetic (PK) analysis on Day 1 and at steady state. In expansion stages, patients with RAS or RAF mutant tumors were treated at the maximum tolerated dose (MTD) of the 21/7 or 14/14 schedule. Results Ninety-seven patients received cobimetinib. In the 21/7 dose escalation, 36 patients enrolled in 8 cohorts (0.05 mg/kg-80 mg). Dose-limiting toxicities (DLTs) were Grade 4 hepatic encephalopathy, Grade 3 diarrhea, and Grade 3 rash. In the 14/14 dose escalation, 20 patients enrolled in 4 cohorts (60-125 mg). DLTs were Grade 3 rash and Grade 3 blurred vision associated with presence of reversible subretinal fluid. The MTD was 60 mg on 21/7 schedule and 100 mg on 14/14 schedule. Cobimetinib PK showed dose-proportional increases in exposure. The most frequent adverse events attributed to cobimetinib were diarrhea, rash, fatigue, edema, nausea, and vomiting. In patients treated at the 60-mg (21/7) or 100-mg (14/14) dose, one unconfirmed complete response and 6 confirmed partial responses were observed. All responses occurred in melanoma patients; 6 harbored the BRAF(V600E) mutation. Conclusions Cobimetinib is generally well tolerated and durable responses were observed in BRAF(V600E) mutant melanoma patients. Evaluation of cobimetinib in combination with other therapies is ongoing.


Assuntos
Antineoplásicos , Azetidinas , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Piperidinas , Inibidores de Proteínas Quinases , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Azetidinas/efeitos adversos , Azetidinas/farmacocinética , Azetidinas/farmacologia , Azetidinas/uso terapêutico , Classe I de Fosfatidilinositol 3-Quinases/genética , Feminino , Genes ras/genética , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Resultado do Tratamento
19.
Nucl Med Biol ; 43(6): 372-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27260778

RESUMO

INTRODUCTION: Nicotinic acetylcholine receptors (nAChRs) are of great interest because they are implicated in higher brain functions. Nuclear medical imaging is one of the useful techniques for noninvasive evaluation of physiological and pathological function in living subjects. Recent progress in nuclear medical imaging modalities enables the clear visualization of the organs of small rodents. Thus, translational research using nuclear medical imaging in transgenic mice has become possible and helps to elucidate human disease pathology. However, imaging of α4ß2 nAChRs in the mouse brain has not yet been performed. The purpose of this study was to assess the feasibility of single-photon emission computed tomography (SPECT) with 5-[(123)I]iodo-3-[2(S)-azetidinylmethoxy]pyridine ([(123)I]5IA) for evaluating α4ß2 nAChR availability in the mouse brain. METHODS: A 60-min dynamic SPECT imaging session of α4ß2 nAChRs in the mouse brain was performed. The regional distribution of radioactivity in the SPECT images was compared to the density of α4ß2 nAChRs measured in an identical mouse. Alteration of nAChR density in the brains of Tg2576 mice was also evaluated. RESULTS: The mouse brain was clearly visualized by [(123)I]5IA-SPECT and probe accumulation was significantly inhibited by pretreatment with (-)-nicotine. The regional distribution of radioactivity in SPECT images showed a significant positive correlation with α4ß2 nAChR density measured in an identical mouse brain. Moreover, [(123)I]5IA-SPECT was able to detect the up-regulation of α4ß2 nAChRs in the brains of Tg2576 transgenic mice. CONCLUSIONS: [(123)I]5IA-SPECT imaging would be a promising tool for evaluating α4ß2 nAChR availability in the mouse brain and may be useful in translational research focused on nAChR-related diseases.


Assuntos
Azetidinas , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Piridinas , Receptores Nicotínicos/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Animais , Autorradiografia , Azetidinas/farmacocinética , Encéfalo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nicotina/farmacologia , Piridinas/farmacocinética , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA