Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Expert Opin Pharmacother ; 25(9): 1111-1120, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38896547

RESUMO

INTRODUCTION: The breakthrough in erythropoietin-stimulating agents in the 1990s improved the prognosis and treatment of complications in chronic kidney disease patients and renal anemia. Discovery of the novel molecular mechanisms for hypoxia-inducible factor (HIF) transcription factor under hypoxic conditions has led to the development of oral drugs, HIF-Prolyl Hydroxylase inhibitors (HIF-PHIs), that constantly activate erythropoietin by inhibiting prolyl hydroxylase. HIF-PHIs have gained rapid approval in Asian countries, including Japan, with six distinct types entering clinical application. AREAS COVERED: This article provides a comprehensive review of the latest literature, with a particular focus on the effectiveness and safety of vadadustat. EXPERT OPINION: A phase 3, randomized, open-label, clinical trial (PRO2TECT) demonstrated that vadadustat had the prespecified non-inferiority for hematologic efficacy as compared with darbepoetin alfa in non-dialysis-dependent patients not previously treated with ESA. However, vadadustat did not show non-inferiority in major adverse cardiovascular events in the non-US/non-Europe patients. It may partly because of imbalances of the baseline eGFR level in those countries. In dialysis-dependent patients, a phase 3 clinical trial (INNO2VATE) showed vadadustat was non-inferior to darbepoetin alfa in cardiovascular safety and maintenance of hemoglobin levels. Adverse events including cancer, retinopathy, thrombosis, and vascular calcification should be evaluated in future clinical studies.


Assuntos
Anemia , Hematínicos , Insuficiência Renal Crônica , Humanos , Anemia/tratamento farmacológico , Anemia/etiologia , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/tratamento farmacológico , Hematínicos/uso terapêutico , Hematínicos/efeitos adversos , Glicina/análogos & derivados , Glicina/uso terapêutico , Glicina/efeitos adversos , Inibidores de Prolil-Hidrolase/uso terapêutico , Inibidores de Prolil-Hidrolase/efeitos adversos , Inibidores de Prolil-Hidrolase/farmacologia , Inibidores de Prolil-Hidrolase/administração & dosagem , Ensaios Clínicos Controlados Aleatórios como Assunto , Eritropoetina/uso terapêutico , Eritropoetina/efeitos adversos , Ácidos Picolínicos/uso terapêutico , Ácidos Picolínicos/efeitos adversos , Ácidos Picolínicos/farmacologia
2.
Proc Natl Acad Sci U S A ; 119(14): e2111804119, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35353625

RESUMO

The receptor for colony stimulating factor 1 (CSF-1R) is important for the survival and function of myeloid cells that mediate pathology during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). CSF-1 and IL-34, the ligands of CSF-1R, have similar bioactivities but distinct tissue and context-dependent expression patterns, suggesting that they have different roles. This could be the case in EAE, given that CSF-1 expression is up-regulated in the CNS, while IL-34 remains constitutively expressed. We found that targeting CSF-1 with neutralizing antibody halted ongoing EAE, with efficacy superior to CSF-1R inhibitor BLZ945, whereas IL-34 neutralization had no effect, suggesting that pathogenic myeloid cells were maintained by CSF-1. Both anti­CSF-1 and BLZ945 treatment greatly reduced the number of monocyte-derived cells and microglia in the CNS. However, anti­CSF-1 selectively depleted inflammatory microglia and monocytes in inflamed CNS areas, whereas BLZ945 depleted virtually all myeloid cells, including quiescent microglia, throughout the CNS. Anti­CSF-1 treatment reduced the size of demyelinated lesions and microglial activation in the gray matter. Lastly, we found that bone marrow­derived immune cells were the major mediators of CSF-1R­dependent pathology, while microglia played a lesser role. Our findings suggest that targeting CSF-1 could be effective in ameliorating MS pathology, while preserving the homeostatic functions of myeloid cells, thereby minimizing risks associated with ablation of CSF-1R­dependent cells.


Assuntos
Encefalomielite Autoimune Experimental , Fator Estimulador de Colônias de Macrófagos , Esclerose Múltipla , Animais , Benzotiazóis/farmacologia , Benzotiazóis/uso terapêutico , Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Ácidos Picolínicos/farmacologia , Ácidos Picolínicos/uso terapêutico , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores
3.
Ther Apher Dial ; 26(1): 45-54, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34115437

RESUMO

Vadadustat is an oral hypoxia-inducible factor prolyl hydroxylase inhibitor approved in Japan for the treatment of anemia in patients with chronic kidney disease (CKD). This phase 3, open-label, single-arm study evaluated the efficacy and safety of vadadustat in 24 Japanese patients with CKD-associated anemia on hemodialysis who were not receiving erythropoiesis-stimulating agents (ESAs). Patients received vadadustat for 24 weeks; the starting dose was 300 mg/day and doses were adjusted to achieve the target hemoglobin (Hb) range of 10.0-12.0 g/dL. The least squares mean of average Hb at Weeks 20 and 24 (95% confidence interval) was 10.75 g/dL (10.35, 11.14). The most common adverse event was shunt stenosis (25.0%). Adverse drug reactions (diarrhea and vomiting) occurred in two patients (8.3%) and the severity was mild. Vadadustat increased and maintained Hb levels within the target range and was generally well-tolerated in Japanese patients with anemia on hemodialysis not receiving ESAs.


Assuntos
Anemia/tratamento farmacológico , Anemia/etiologia , Glicina/análogos & derivados , Ácidos Picolínicos/uso terapêutico , Diálise Renal/efeitos adversos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia , Feminino , Glicina/uso terapêutico , Hematínicos , Humanos , Japão , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
4.
J Med Chem ; 64(16): 11904-11933, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34382802

RESUMO

Due to increased lactate production during glucose metabolism, tumor cells heavily rely on efficient lactate transport to avoid intracellular lactate accumulation and acidification. Monocarboxylate transporter 4 (MCT4/SLC16A3) is a lactate transporter that plays a central role in tumor pH modulation. The discovery and optimization of a novel class of MCT4 inhibitors (hit 9a), identified by a cellular screening in MDA-MB-231, is described. Direct target interaction of the optimized compound 18n with the cytosolic domain of MCT4 was shown after solubilization of the GFP-tagged transporter by fluorescence cross-correlation spectroscopy and microscopic studies. In vitro treatment with 18n resulted in lactate efflux inhibition and reduction of cellular viability in MCT4 high expressing cells. Moreover, pharmacokinetic properties of 18n allowed assessment of lactate modulation and antitumor activity in a mouse tumor model. Thus, 18n represents a valuable tool for investigating selective MCT4 inhibition and its effect on tumor biology.


Assuntos
Antineoplásicos/uso terapêutico , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Proteínas Musculares/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Ácidos Picolínicos/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Células HEK293 , Humanos , Ácido Láctico/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos SCID , Estrutura Molecular , Ácidos Picolínicos/síntese química , Ácidos Picolínicos/farmacologia , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
N Engl J Med ; 384(17): 1601-1612, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33913638

RESUMO

BACKGROUND: Vadadustat is an oral hypoxia-inducible factor prolyl hydroxylase inhibitor, a class of compounds that stimulate endogenous erythropoietin production. METHODS: We conducted two randomized, open-label, noninferiority phase 3 trials to evaluate the safety and efficacy of vadadustat, as compared with darbepoetin alfa, in patients with anemia and incident or prevalent dialysis-dependent chronic kidney disease (DD-CKD). The primary safety end point, assessed in a time-to-event analysis, was the first occurrence of a major adverse cardiovascular event (MACE, a composite of death from any cause, a nonfatal myocardial infarction, or a nonfatal stroke), pooled across the trials (noninferiority margin, 1.25). A key secondary safety end point was the first occurrence of a MACE plus hospitalization for either heart failure or a thromboembolic event. The primary and key secondary efficacy end points were the mean change in hemoglobin from baseline to weeks 24 to 36 and from baseline to weeks 40 to 52, respectively, in each trial (noninferiority margin, -0.75 g per deciliter). RESULTS: A total of 3923 patients were randomly assigned in a 1:1 ratio to receive vadadustat or darbepoetin alfa: 369 in the incident DD-CKD trial and 3554 in the prevalent DD-CKD trial. In the pooled analysis, a first MACE occurred in 355 patients (18.2%) in the vadadustat group and in 377 patients (19.3%) in the darbepoetin alfa group (hazard ratio, 0.96; 95% confidence interval [CI], 0.83 to 1.11). The mean differences between the groups in the change in hemoglobin concentration were -0.31 g per deciliter (95% CI, -0.53 to -0.10) at weeks 24 to 36 and -0.07 g per deciliter (95% CI, -0.34 to 0.19) at weeks 40 to 52 in the incident DD-CKD trial and -0.17 g per deciliter (95% CI, -0.23 to -0.10) and -0.18 g per deciliter (95% CI, -0.25 to -0.12), respectively, in the prevalent DD-CKD trial. The incidence of serious adverse events in the vadadustat group was 49.7% in the incident DD-CKD trial and 55.0% in the prevalent DD-CKD trial, and the incidences in the darbepoetin alfa group were 56.5% and 58.3%, respectively. CONCLUSIONS: Among patients with anemia and CKD who were undergoing dialysis, vadadustat was noninferior to darbepoetin alfa with respect to cardiovascular safety and correction and maintenance of hemoglobin concentrations. (Funded by Akebia Therapeutics and Otsuka Pharmaceutical; INNO2VATE ClinicalTrials.gov numbers, NCT02865850 and NCT02892149.).


Assuntos
Anemia/tratamento farmacológico , Darbepoetina alfa/uso terapêutico , Glicina/análogos & derivados , Hematínicos/uso terapêutico , Ácidos Picolínicos/uso terapêutico , Inibidores de Prolil-Hidrolase/uso terapêutico , Diálise Renal/efeitos adversos , Insuficiência Renal Crônica/complicações , Idoso , Anemia/sangue , Anemia/etiologia , Doenças Cardiovasculares/induzido quimicamente , Darbepoetina alfa/efeitos adversos , Feminino , Glicina/efeitos adversos , Glicina/uso terapêutico , Hematínicos/efeitos adversos , Hemoglobinas/análise , Humanos , Masculino , Pessoa de Meia-Idade , Ácidos Picolínicos/efeitos adversos , Inibidores de Prolil-Hidrolase/efeitos adversos , Insuficiência Renal Crônica/mortalidade , Insuficiência Renal Crônica/terapia
6.
N Engl J Med ; 384(17): 1589-1600, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33913637

RESUMO

BACKGROUND: Vadadustat is an oral hypoxia-inducible factor (HIF) prolyl hydroxylase inhibitor, a class of drugs that stabilize HIF and stimulate erythropoietin and red-cell production. METHODS: In two phase 3, randomized, open-label, active-controlled, noninferiority trials, we compared vadadustat with the erythropoiesis-stimulating agent (ESA) darbepoetin alfa in patients with non-dialysis-dependent chronic kidney disease (NDD-CKD) not previously treated with an ESA who had a hemoglobin concentration of less than 10 g per deciliter and in patients with ESA-treated NDD-CKD and a hemoglobin concentration of 8 to 11 g per deciliter (in the United States) or 9 to 12 g per deciliter (in other countries). The primary safety end point, assessed in a time-to-event analysis, was the first major adverse cardiovascular event (MACE; a composite of death from any cause, nonfatal myocardial infarction, or nonfatal stroke), pooled across the two trials. Secondary safety end points included expanded MACE (MACE plus hospitalization for either heart failure or a thromboembolic event). The primary and key secondary efficacy end points in each trial were the mean change in hemoglobin concentration from baseline during two evaluation periods: weeks 24 through 36 and weeks 40 through 52. RESULTS: A total of 1751 patients with ESA-untreated NDD-CKD and 1725 with ESA-treated NDD-CKD underwent randomization in the two trials. In the pooled analysis, in which 1739 patients received vadadustat and 1732 received darbepoetin alfa, the hazard ratio for MACE was 1.17 (95% confidence interval [CI], 1.01 to 1.36), which did not meet the prespecified noninferiority margin of 1.25. The mean between-group differences in the change in the hemoglobin concentration at weeks 24 through 36 were 0.05 g per deciliter (95% CI, -0.04 to 0.15) in the trial involving ESA-untreated patients and -0.01 g per deciliter (95% CI, -0.09 to 0.07) in the trial involving ESA-treated patients, which met the prespecified noninferiority margin of -0.75 g per deciliter. CONCLUSIONS: Vadadustat, as compared with darbepoetin alfa, met the prespecified noninferiority criterion for hematologic efficacy but not the prespecified noninferiority criterion for cardiovascular safety in patients with NDD-CKD. (Funded by Akebia Therapeutics and Otsuka Pharmaceutical; PRO2TECT ClinicalTrials.gov numbers, NCT02648347 and NCT02680574.).


Assuntos
Anemia/tratamento farmacológico , Darbepoetina alfa/uso terapêutico , Glicina/análogos & derivados , Hematínicos/uso terapêutico , Ácidos Picolínicos/uso terapêutico , Inibidores de Prolil-Hidrolase/uso terapêutico , Insuficiência Renal Crônica/complicações , Administração Oral , Idoso , Anemia/sangue , Anemia/etiologia , Doenças Cardiovasculares/induzido quimicamente , Darbepoetina alfa/efeitos adversos , Feminino , Glicina/efeitos adversos , Glicina/uso terapêutico , Hematínicos/efeitos adversos , Hemoglobinas/análise , Hospitalização/estatística & dados numéricos , Humanos , Masculino , Pessoa de Meia-Idade , Ácidos Picolínicos/efeitos adversos , Inibidores de Prolil-Hidrolase/efeitos adversos , Insuficiência Renal Crônica/mortalidade
7.
Saudi J Kidney Dis Transpl ; 32(6): 1671-1678, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35946280

RESUMO

Metabolic syndrome (MS) is caused by environmental factors as well as genetic. Human studies of efficacy of chromium for glucose and lipid metabolism and insulin function is not still definitive. Furthermore, the effect of chromium supplementation on the expression of inflammatory genes in patients with MS has not been studied. We will assess effects of chromium picolinate supplementation on gene expression of tumor necrosis factor-α (TNF-α) and DNA damage in MS patients. In this triple-blind randomized placebo-controlled clinical trial, 48 MS patients will be randomly assigned into two groups to receive daily 400 µg chromium picolinate supplement or placebo for 12 weeks. The outcome measures include of change in fasting blood sugar, glycosylated hemoglobin A1C, inflammatory biomarkers, lipid profile, blood pressure, gene expression of TNF-α, and 8-hydroxy-deoxyguanosine concentration as DNA damage biomarker, will be quantified at baseline and end of intervention. This protocol was approved by Institutional Research Ethics Committee School of Public Health Shahid Sadoughi University of Medical Sciences (Approval ID: IR.SSU.SPH.REC.1399.141).


Assuntos
Suplementos Nutricionais , Síndrome Metabólica , Ácidos Picolínicos , Biomarcadores , Glicemia , Cromo/uso terapêutico , Dano ao DNA , Expressão Gênica , Humanos , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/genética , Ácidos Picolínicos/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Fator de Necrose Tumoral alfa
8.
Nephrol Dial Transplant ; 36(11): 2039-2048, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33188693

RESUMO

BACKGROUND: Erythropoiesis-stimulating agents (ESAs) are currently the mainstay of treatment for anaemia of chronic kidney disease (CKD). Vadadustat is an investigational oral hypoxia-inducible factor prolyl-hydroxylase inhibitor that stimulates endogenous erythropoietin formation. The INNO2VATE programme comprises two studies designed to evaluate the safety and efficacy of vadadustat versus the ESA darbepoetin alfa in ameliorating anaemia in patients with dialysis-dependent CKD (DD-CKD). Here we describe the trial design along with patient demographics and baseline characteristics. METHODS: Two Phase 3, open-label, sponsor-blind, active-controlled trials enrolled adults with anaemia of CKD who recently initiated dialysis and had limited ESA exposure (incident DD-CKD trial) or were receiving maintenance dialysis with ESA treatment (prevalent DD-CKD trial). Study periods include correction/conversion (Weeks 0-23), maintenance (Weeks 24-52), long-term treatment (Weeks 53 to end of treatment) and safety follow-up. The primary safety endpoint is the time to the first major adverse cardiovascular event and the primary efficacy endpoint is the change in haemoglobin (baseline to Weeks 24-36). RESULTS: A total of 369 and 3554 patients were randomized in the incident DD-CKD and prevalent DD-CKD trials, respectively. Demographics and baseline characteristics were similar among patients in both trials and comparable to those typically observed in DD-CKD. CONCLUSIONS: The two INNO2VATE trials will provide important information on the safety and efficacy of a novel approach for anaemia management in a diverse DD-CKD population. Demographics and baseline characteristics of enrolled patients suggest that study results will be representative for a large proportion of the DD-CKD population.


Assuntos
Anemia , Glicina/uso terapêutico , Hematínicos , Ácidos Picolínicos/uso terapêutico , Insuficiência Renal Crônica , Adulto , Anemia/tratamento farmacológico , Anemia/etiologia , Eritropoetina , Glicina/análogos & derivados , Hematínicos/uso terapêutico , Hemoglobinas/análise , Humanos , Diálise Renal , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia
9.
Ann Neurol ; 86(4): 561-571, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31359452

RESUMO

OBJECTIVE: Clinical trials targeting ß-amyloid peptides (Aß) for Alzheimer disease (AD) failed for arguable reasons that include selecting the wrong stages of AD pathophysiology or Aß being the wrong target. Targeting Aß to prevent cerebral amyloid angiopathy (CAA) has not been rigorously followed, although the causal role of Aß for CAA and related hemorrhages is undisputed. CAA occurs with normal aging and to various degrees in AD, where its impact and treatment is confounded by the presence of parenchymal Aß deposition. METHODS: APPDutch mice develop CAA in the absence of parenchymal amyloid, mimicking hereditary cerebral hemorrhage with amyloidosis Dutch type (HCHWA-D). Mice were treated with a ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitor. We used 3-dimensional ultramicroscopy and immunoassays for visualizing CAA and assessing Aß in cerebrospinal fluid (CSF) and brain. RESULTS: CAA onset in mice was at 22 to 24 months, first in frontal leptomeningeal and superficial cortical vessels followed by vessels penetrating the cortical layers. CSF Aß increased with aging followed by a decrease of both Aß40 and Aß42 upon CAA onset, supporting the idea that combined reduction of CSF Aß40 and Aß42 is a specific biomarker for vascular amyloid. BACE1 inhibitor treatment starting at CAA onset and continuing for 4 months revealed a 90% Aß reduction in CSF and largely prevented CAA progression and associated pathologies. INTERPRETATION: This is the first study showing that Aß reduction at early disease time points largely prevents CAA in the absence of parenchymal amyloid. Our observation provides a preclinical basis for Aß-reducing treatments in patients at risk of CAA and in presymptomatic HCHWA-D. ANN NEUROL 2019;86:561-571.


Assuntos
Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Angiopatia Amiloide Cerebral/tratamento farmacológico , Progressão da Doença , Ácidos Picolínicos/uso terapêutico , Tiazinas/uso terapêutico , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Biomarcadores/líquido cefalorraquidiano , Biomarcadores/metabolismo , Encéfalo/irrigação sanguínea , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Ácidos Picolínicos/farmacologia , Tiazinas/farmacologia
10.
Immunobiology ; 224(3): 355-361, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30926154

RESUMO

Docetaxel (DTXL), a new member of the taxoid family, has been used for cancer treatment. However, increasing cases of DTXL resistance have been reported. Tumor-associated macrophages (TAMs) have been implicated in tumor invasion and chemo-resistance. Eliminating TAMs by inhibiting colony stimulating factor-1 receptor (CSF-1R) has emerged as a promising strategy for cancer treatment. BLZ945 is a CSF-1R inhibitor and has anti-tumor function. In present study, anti-tumor effects of combination treatment of BLZ945 and DTXL were investigated. We established a mouse ovarian cancer model and investigated the effect of BLZ945, DTXL single treatment or combination treatment on TAMs infiltration, tumor growth, CD8+ T cell infiltration and cancer metastasis. DTXL treatment increased the infiltration while BLZ945 induced cell apoptosis in macrophages. DTXL/BLZ945 combination treatment significantly inhibited tumor growth, reduced the abundance of TAMs, increased CD8+ T cell infiltration and prevented lung metastasis. Depletion of Tumor-Associated Macrophages (TAMs) by BLZ945 enhanced the anti-tumor effect of DTXL in a murine epithelial ovarian cancer.


Assuntos
Antineoplásicos/uso terapêutico , Benzotiazóis/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Carcinoma Epitelial do Ovário/tratamento farmacológico , Docetaxel/uso terapêutico , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/metabolismo , Ácidos Picolínicos/uso terapêutico , Animais , Apoptose , Benzotiazóis/farmacologia , Carcinoma Epitelial do Ovário/imunologia , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Quimioterapia Combinada , Feminino , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Ácidos Picolínicos/farmacologia , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Microambiente Tumoral
11.
Glia ; 67(2): 291-308, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30456797

RESUMO

Multiple sclerosis (MS) is one of the most common causes of progressive disability affecting young people with very few therapeutic options available for its progressive forms. Its pathophysiology involves demyelination and neurodegeneration apparently driven by microglial activation, which is physiologically dependent on colony-stimulating factor-1 receptor (CSF-1R) signaling. In the present work, we used microglial modulation through oral administration of brain-penetrant CSF-1R inhibitor BLZ945 in acute and chronic cuprizone (CPZ)-induced demyelination to evaluate preventive and therapeutic effects on de/remyelination and neurodegeneration. Our results show that BLZ945 induced a significant reduction in the number of microglia. Preventive BLZ945 treatment attenuated demyelination in the acute CPZ model, mainly in cortex and external capsule. In contrast, BLZ945 treatment in the acute CPZ model failed to protect myelin or foster remyelination in myelin-rich areas, which may respond to a loss in microglial phagocytic capacity and the consequent impairment in oligodendroglial differentiation. Preventive and therapeutic BLZ945 treatment promoted remyelination and neuroprotection in the chronic model. These results could be potentially transferred to the treatment of progressive forms of MS.


Assuntos
Doenças Desmielinizantes/metabolismo , Microglia/metabolismo , Receptores de Fator Estimulador de Colônias/antagonistas & inibidores , Receptores de Fator Estimulador de Colônias/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Benzotiazóis/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/ultraestrutura , Bromodesoxiuridina/metabolismo , Cuprizona/toxicidade , Citocinas/metabolismo , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/tratamento farmacológico , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Microglia/ultraestrutura , Microscopia Eletrônica de Transmissão , Proteína Básica da Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ácidos Picolínicos/uso terapêutico , Receptores de Fator Estimulador de Colônias/genética , Fatores de Tempo
12.
Biol Trace Elem Res ; 184(1): 119-126, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28975521

RESUMO

In vivo and in vitro studies suggested that chromium enhances insulin sensitivity by promoting insulin receptor signaling. However, its effect on insulin clearance has not been yet identified. Nigella sativa, a widely used spice, possesses an antidiabetic activity. We, therefore, hypothesized that chromium picolinate may alter insulin clearance by modulating insulin-degrading enzyme (IDE) in insulin-resistant rats. We evaluated also the effect of Nigella sativa oil on insulin signaling and degradation with respect to chromium picolinate. To assess these hypotheses, insulin resistance was induced in 30 male Wistar albino rats through daily oral administration of high-fructose water (HFW, 20% w/v) for 45 days. These rats were then divided into three groups (n = 10/group). They were given either no treatment (control group) or Nigella sativa oil (500 mg/kg bw/day) or chromium picoloinate (200 µg/kg bw/day) orally along with HFW (20% w/v) for 45 days. Nigella sativa oil or chromium picolinate concurrent administration with HFW significantly decreased body weight, serum lipids, glucagon, insulin resistance, and hepatic IDE level but increased its mRNA expression and insulin receptor phosphorlyation as well as high-density lipoprotein cholesterol (HDL-C) level as compared to control group values, suggesting their potential as modulators for insulin signaling and clearance. However, Nigella sativa oil exerted better improvement in feeding efficacy ratio as well as the levels of glucagon, insulin, insulin resistance, hepatic IDE level and insulin receptor phosphorylation than chromium picolinate, suggesting its greater insulin sensitizing capacity. Our data, for the first time, prove that Nigella sativa oil and chromium picolinate monotherapy can reduce fructose-induced insulin resistance by reduction of hepatic IDE protein and activation of insulin receptor signaling.


Assuntos
Frutose/toxicidade , Hiperinsulinismo/induzido quimicamente , Hiperinsulinismo/tratamento farmacológico , Insulina/metabolismo , Insulisina/metabolismo , Nigella sativa/química , Ácidos Picolínicos/uso terapêutico , Óleos de Plantas/uso terapêutico , Animais , Hiperinsulinismo/metabolismo , Masculino , Ratos , Ratos Wistar
13.
Hemodial Int ; 21 Suppl 1: S110-S124, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28449418

RESUMO

A classic response to systemic hypoxia is the increase in red blood cell production. This response is controlled by the prolyl hydroxylase domain/hypoxia-inducible factor (HIF) pathway, which regulates a broad spectrum of cellular functions. The discovery of this pathway as a key regulator of erythropoiesis has led to the development of small molecules that stimulate the production of endogenous erythropoietin and enhance iron metabolism. This review provides a concise overview of the cellular and molecular mechanisms that govern HIF-induced erythropoietic responses and provides an update on clinical experience with compounds that target HIF-prolyl hydroxylases for anemia therapy.


Assuntos
Eritropoese/efeitos dos fármacos , Prolina Dioxigenases do Fator Induzível por Hipóxia/fisiologia , Ferro/metabolismo , Inibidores de Prolil-Hidrolase/uso terapêutico , Anemia/tratamento farmacológico , Barbitúricos/efeitos adversos , Barbitúricos/uso terapêutico , Ensaios Clínicos como Assunto , Eritropoetina/biossíntese , Glicina/efeitos adversos , Glicina/análogos & derivados , Glicina/uso terapêutico , Humanos , Isoquinolinas/efeitos adversos , Isoquinolinas/uso terapêutico , Ácidos Picolínicos/efeitos adversos , Ácidos Picolínicos/uso terapêutico , Diálise Renal/efeitos adversos
14.
Am J Nephrol ; 45(5): 380-388, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28343225

RESUMO

BACKGROUND: Therapeutic options for the treatment of anemia secondary to chronic kidney disease (CKD) remain limited. Vadadustat (AKB-6548) is an oral hypoxia-inducible factor prolyl-hydroxylase domain (HIF-PHD) inhibitor that is being investigated for the treatment of anemia secondary to CKD. METHODS: A phase 2a, multicenter, randomized, double-blind, placebo-controlled, dose-ranging trial (NCT01381094) was undertaken in adults with anemia secondary to CKD stage 3 or 4. Eligible subjects were evenly randomized to 5 groups: 240, 370, 500, or 630 mg of once-daily oral vadadustat or placebo for 6 weeks. All subjects received low-dose supplemental oral iron (50 mg daily). The primary endpoint was the mean absolute change in hemoglobin (Hb) from baseline to the end of treatment. Secondary endpoints included iron indices, safety, and tolerability. RESULTS: Ninety-three subjects were randomized. Compared with placebo, vadadustat significantly increased Hb after 6 weeks in a dose-dependent manner (analysis of variance; p < 0.0001). Vadadustat increased the total iron-binding capacity and decreased concentrations of ferritin and hepcidin. The proportion of subjects with at least 1 treatment-emergent adverse event was similar between vadadustat- and placebo-treated groups. No significant changes in blood pressure, vascular endothelial growth factor, C-reactive protein, or total cholesterol were observed. Limitations of this study included its small sample size and short treatment duration. CONCLUSIONS: Vadadustat increased Hb levels and improved biomarkers of iron mobilization and utilization in patients with anemia secondary to stage 3 or 4 CKD. Global multicenter, randomized phase 3 trials are ongoing in non-dialysis-dependent and dialysis-dependent patients.


Assuntos
Anemia/tratamento farmacológico , Glicina/análogos & derivados , Prolina Dioxigenases do Fator Induzível por Hipóxia/antagonistas & inibidores , Ácidos Picolínicos/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Adulto , Idoso , Anemia/sangue , Anemia/etiologia , Biomarcadores/sangue , Proteína C-Reativa/análise , Colesterol/sangue , Relação Dose-Resposta a Droga , Método Duplo-Cego , Eritropoetina/sangue , Feminino , Ferritinas/sangue , Ferritinas/metabolismo , Glicina/farmacologia , Glicina/uso terapêutico , Hemoglobinas/análise , Hepcidinas/sangue , Hepcidinas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Ácidos Picolínicos/farmacologia , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/complicações , Fator A de Crescimento do Endotélio Vascular/sangue
15.
Oncotarget ; 7(35): 56408-56421, 2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27486763

RESUMO

In the present work we show that multiple lung cancer cell lines contain cisplatin resistant cell subpopulations expressing the Colony-Stimulating-Factor-Receptor-1 (CSF-1R) and surviving chemotherapy-induced stress. By exploiting siRNA-mediated knock down in vitro and the use of an investigational CSF-1R TKI (JNJ-40346527) in vitro and in vivo, we show that expression and function of the receptor are required for the clonogenicity and chemoresistance of the cell lines. Thus, inhibition of the kinase activity of the receptor reduced the levels of EMT-associated genes, stem cell markers and chemoresistance genes. Additionally, the number of high aldehyde dehydrogenase (ALDH) expressing cells was reduced, consequent to the lack of cisplatin-induced increase of ALDH isoforms. This affected the collective chemoresistance of the treated cultures. Treatment of tumor bearing mice with JNJ-40346527, at pharmacologically relevant doses, produced strong chemo-sensitizing effects in vivo. These anticancer effects correlated with a reduced number of CSF-1Rpos cells, in tumors excised from the treated mice. Depletion of the CD45pos cells within the treated tumors did not, apparently, play a major role in mediating the therapeutic response to the TKI. Thus, lung cancer cells express a functional CSF-1 and CSF-1R duo which mediates pro-tumorigenic effects in vivo and in vitro and can be targeted in a therapeutically relevant way. These observations complement the already known role for the CSF-1R at mediating the pro-tumorigenic properties of tumor-infiltrating immune components.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Aldeído Desidrogenase/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzotiazóis/farmacologia , Benzotiazóis/uso terapêutico , Carcinogênese/patologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Regulação para Baixo , Sinergismo Farmacológico , Técnicas de Silenciamento de Genes , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Antígenos Comuns de Leucócito/metabolismo , Neoplasias Pulmonares/patologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fosforilação , Ácidos Picolínicos/farmacologia , Ácidos Picolínicos/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Science ; 352(6288): aad3018, 2016 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-27199435

RESUMO

Macrophages accumulate with glioblastoma multiforme (GBM) progression and can be targeted via inhibition of colony-stimulating factor-1 receptor (CSF-1R) to regress high-grade tumors in animal models of this cancer. However, whether and how resistance emerges in response to sustained CSF-1R blockade is unknown. We show that although overall survival is significantly prolonged, tumors recur in >50% of mice. Gliomas reestablish sensitivity to CSF-1R inhibition upon transplantation, indicating that resistance is tumor microenvironment-driven. Phosphatidylinositol 3-kinase (PI3K) pathway activity was elevated in recurrent GBM, driven by macrophage-derived insulin-like growth factor-1 (IGF-1) and tumor cell IGF-1 receptor (IGF-1R). Combining IGF-1R or PI3K blockade with CSF-1R inhibition in recurrent tumors significantly prolonged overall survival. Our findings thus reveal a potential therapeutic approach for treating resistance to CSF-1R inhibitors.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzotiazóis/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/tratamento farmacológico , Imidazóis/uso terapêutico , Neoplasias Experimentais/terapia , Ácidos Picolínicos/uso terapêutico , Pirazinas/uso terapêutico , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Microambiente Tumoral/imunologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzotiazóis/farmacologia , Glioblastoma/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Imidazóis/farmacologia , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos , Fatores de Transcrição NFATC/metabolismo , Recidiva Local de Neoplasia/metabolismo , Neoplasias Experimentais/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Ácidos Picolínicos/farmacologia , Pirazinas/farmacologia , Receptor IGF Tipo 1/antagonistas & inibidores , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais
17.
J Med Chem ; 58(21): 8373-86, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26505898

RESUMO

Pan proviral insertion site of Moloney murine leukemia (PIM) 1, 2, and 3 kinase inhibitors have recently begun to be tested in humans to assess whether pan PIM kinase inhibition may provide benefit to cancer patients. Herein, the synthesis, in vitro activity, in vivo activity in an acute myeloid leukemia xenograft model, and preclinical profile of the potent and selective pan PIM kinase inhibitor compound 8 (PIM447) are described. Starting from the reported aminopiperidyl pan PIM kinase inhibitor compound 3, a strategy to improve the microsomal stability was pursued resulting in the identification of potent aminocyclohexyl pan PIM inhibitors with high metabolic stability. From this aminocyclohexyl series, compound 8 entered the clinic in 2012 in multiple myeloma patients and is currently in several phase 1 trials of cancer patients with hematological malignancies.


Assuntos
Leucemia Mieloide Aguda/tratamento farmacológico , Ácidos Picolínicos/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Amidas/síntese química , Amidas/química , Amidas/uso terapêutico , Animais , Linhagem Celular Tumoral , Halogenação , Humanos , Leucemia Mieloide Aguda/metabolismo , Camundongos , Modelos Moleculares , Ácidos Picolínicos/síntese química , Ácidos Picolínicos/química , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/metabolismo
18.
Ann Nutr Metab ; 67(1): 42-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26279073

RESUMO

BACKGROUND: The aim of the present study was to evaluate the beneficial effects of chromium intake on markers of insulin metabolism and lipid profiles in women with polycystic ovary syndrome (PCOS). METHODS: In a prospective, randomized, double-blind, placebo-controlled trial, 64 women with PCOS were randomized to receive 200 µg chromium picolinate supplements (n = 32) or placebo (n = 32) for 8 weeks. Fasting blood samples were obtained at baseline and 8 weeks after the intervention to quantify markers of insulin metabolism and lipid concentrations. RESULTS: Chromium supplementation in women with PCOS resulted in significant decreases in serum insulin levels (-3.6 ± 7.4 vs. +3.6 ± 6.2 µIU/ml, p < 0.001), homeostasis model of assessment-insulin resistance (HOMA-IR; -0.8 ± 1.6 vs. +0.9 ± 1.5, p < 0.001), homeostatic model assessment-beta cell function (HOMA-B; -15.5 ± 32.3 vs. +13.6 ± 23.1, p < 0.001), and a significant increase in quantitative insulin sensitivity check index (QUICKI) score (+0.02 ± 0.03 vs. -0.008 ± 0.02, p = 0.001) compared with the placebo. In addition, a trend toward a significant effect of chromium supplementation on decreasing serum triglycerides (-12.4 ± 74.4 vs. +15.2 ± 32.4 mg/dl, p = 0.05), very low-density lipoprotein-cholesterol (-2.5 ± 14.9 vs. +3.0 ± 6.5 mg/dl, p = 0.05), and cholesterol concentrations (-8.6 ± 21.9 vs. +0.7 ± 22.4 mg/dl, p = 0.09) was seen. CONCLUSIONS: Eight weeks of chromium supplementation among PCOS women had favorable effects on markers of insulin metabolism.


Assuntos
Suplementos Nutricionais , Resistência à Insulina , Quelantes de Ferro/uso terapêutico , Ácidos Picolínicos/uso terapêutico , Síndrome do Ovário Policístico/tratamento farmacológico , Adulto , Colesterol/sangue , VLDL-Colesterol/sangue , Método Duplo-Cego , Jejum/sangue , Feminino , Homeostase/fisiologia , Humanos , Insulina/sangue , Células Secretoras de Insulina/fisiologia , Síndrome do Ovário Policístico/sangue , Estudos Prospectivos , Triglicerídeos/sangue , Adulto Jovem
19.
Sci Rep ; 4: 6031, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25112436

RESUMO

Antagonizing vascular endothelial growth factor receptor 2 (VEGFR2) to block angiogenesis has been applied toward cancer therapy for its role in promoting cancer growth and metastasis. However, most these clinical anticancer drugs have unexpected side effects. Development of novel VEGFR2 inhibitors with less toxicity remains an urgent need. In this study, we describe a novel, well-tolerated, and orally active VEGFR2 inhibitor, YLT192, which inhibits tumor angiogenesis and growth. YLT192 significantly inhibited kinase activity of VEGFR2 and suppressed proliferation, migration, invasion, and tube formation of human umbilical vascular endothelial cells (HUVEC) in vitro. In addition, it inhibited VEGF-induced phosphorylation of VEGFR2 and its downstream signaling regulator in HUVEC. Zebrafish embryonic models and alginate-encapsulated tumor cell assays indicated YLT192 also inhibited angiogenesis in vivo. Moreover, YLT192 could directly inhibit proliferation and induce apoptosis of cancer cells in vitro and in vivo. Oral administration of YLT192 at a dose of 100 mg/kg/day could markedly inhibited human tumor xenograft growth without causing obvious toxicities. It decreased microvessel densities (MVD) in tumor sections. It also shows good safety profiles in the studies with mice and rats. Taken together, these preclinical evaluations suggest that YLT192 inhibits angiogenesis and may be a promising anticancer drug candidate.


Assuntos
Inibidores da Angiogênese/farmacologia , Benzamidas/farmacologia , Ácidos Picolínicos/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/química , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/farmacologia , Benzamidas/química , Benzamidas/uso terapêutico , Sítios de Ligação , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Feminino , Células HCT116 , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Ácidos Picolínicos/química , Ácidos Picolínicos/uso terapêutico , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/crescimento & desenvolvimento
20.
Expert Opin Ther Pat ; 24(7): 829-37, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24392786

RESUMO

Cystic fibrosis (CF) is a genetic disease caused by defects in the CF transmembrane regulator (CFTR) gene, which encodes an epithelial chloride channel. The most common mutation, Δ508CFTR, produces a protein that is misfolded and does not reach the cell membrane. The discovery that partial chloride channel function could be restored by exposure to exogenous chemical stimuli led to the search for selective CFTR modulators. Vertex has led the way in developing N-aryl-1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropanecarboxamide derivatives such as lumacaftor and VX-661, which correct trafficking of Δ508CFTR and partially restore chloride channel activity. Novartis had identified similar activity in a series of picolinamide derivatives such as 3-amino-N-[(2S)-3,3,3-trifluoro-2-hydroxy-2-methylpropyl]-5,6-bis(trifluoromethyl)pyridine-2-carboxamide. A series of five filings from Novartis has expanded on the activity of compounds based on this scaffold and provided compounds with nanomolar potency in cellular assays.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/efeitos dos fármacos , Fibrose Cística/tratamento farmacológico , Ácidos Picolínicos/uso terapêutico , Amidas/química , Amidas/farmacologia , Amidas/uso terapêutico , Animais , Humanos , Patentes como Assunto , Ácidos Picolínicos/química , Ácidos Picolínicos/farmacologia , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA