Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 282
Filtrar
1.
Clin Transl Sci ; 17(4): e13775, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38651744

RESUMO

This study aimed to evaluate the pharmacokinetics (PKs), safety, and immunogenicity of the biosimilar HEC14028 compared to reference Trulicity® (dulaglutide) in healthy male Chinese subjects. This study was a single-center, randomized, open, single-dose, parallel-controlled comparative Phase I clinical trial, including a screening period of up to 14 days, a 17-day observation period after administration, and a 7-day safety follow-up period. A total of 68 healthy male subjects were randomly assigned (1:1) to the test group (HEC14028) and the reference group (dulaglutide) (single 0.75 mg abdominal subcutaneous dose). The primary objective was to evaluate the pharmacokinetic characteristics of HEC14028 and compare the pharmacokinetic similarities between HEC14028 and dulaglutide. The primary PK endpoints were maximum plasma concentration (Cmax) and area under the blood concentration-time curve from zero time to the estimated infinite time (AUC0-∞). The study results showed that HEC14028 and dulaglutide were pharmacokinetically equivalent: 90% confidence interval (CI) of Cmax and AUC0-∞ geometric mean ratios were 102.9%-122.0% and 97.1%-116.9%, respectively, which were both within the range of 80.00%-125.00%. No grade 3 or above treatment emergent adverse events (TEAEs), serious adverse events (SAEs), TEAEs leading to withdrawal from the trial, or TEAEs leading to death were reported in this study. Both HEC14028 and dulaglutide showed good and similar safety profiles, and no incremental immunogenicity was observed in subjects receiving HEC14028 and dulaglutide.


Assuntos
Medicamentos Biossimilares , Peptídeos Semelhantes ao Glucagon , Peptídeos Semelhantes ao Glucagon/análogos & derivados , Voluntários Saudáveis , Fragmentos Fc das Imunoglobulinas , Proteínas Recombinantes de Fusão , Humanos , Masculino , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/efeitos adversos , Fragmentos Fc das Imunoglobulinas/imunologia , Peptídeos Semelhantes ao Glucagon/farmacocinética , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Peptídeos Semelhantes ao Glucagon/efeitos adversos , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/efeitos adversos , Medicamentos Biossimilares/farmacocinética , Medicamentos Biossimilares/administração & dosagem , Medicamentos Biossimilares/efeitos adversos , Adulto , Adulto Jovem , China , Área Sob a Curva , Povo Asiático , Equivalência Terapêutica , Injeções Subcutâneas , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/efeitos adversos , Pessoa de Meia-Idade , Adolescente , População do Leste Asiático
2.
s,l; CONETEC; 11 nov. 2023.
Não convencional em Espanhol | BRISA/RedTESA | ID: biblio-1523086

RESUMO

INTRODUCCIÓN: La miastenia gravis es un trastorno neuromuscular autoinmune caracterizada por una debilidad motora fluctuante que afecta a los músculos oculares, bulbares, de las extremidades y/o respiratorios.1 La debilidad se debe a un ataque inmunológico mediado por anticuerpos dirigido a proteínas de la membrana post-sináptica de la unión neuromuscular, como los receptores de acetilcolina o proteínas asociadas a receptores. La miastenia gravis generalizada es trastorno crónico y de baja incidencia que se manifiesta predominantemente por debilidad generalizada del músculo esquelético y debilidad inducida por el ejercicio. 1,2 Entre el 80 y 90 % de las personas con miastenia gravis tienen autoanticuerpos contra el receptor de acetilcolina (AChR) detectables en el suero, y se cree que estos anticuerpos desempeñan un papel central en el mecanismo patogénico de la enfermedad.1 A nivel celular, los anticuerpos AChR causan disfunción en la unión neuromuscular al bloquear la unión de ACh al AChR, entrecruzar e internalizar los AChR y activar la destrucción de AChR mediada por el complemento. Se cree que existe una asociación entre la presencia de anticuerpo AChR y la gravedad de los síntomas clínicos de la miastenia gravis, sin embargo, su correlación todavía no está demostrada. TECNOLOGÍA: Efgartigimod alfa (Vyvgart®) es una molécula de fragmento Fc de anticuerpo IgG diseñada para promover la degradación de autoanticuerpos IgG.3 Se administra por via intravenosa a una dosis de 10 mg/kg administrados durante una hora una vez a la semana durante 4 semanas. En personas que pesan 120 kg o más, la dosis recomendada es de 1.200 mg (3 viales). En la actualidad la administración subcutánea del fármaco se encuentra en estudio. OBJETIVO: El objetivo del presente informe es evaluar rápidamente los parámetros de eficacia, seguridad, costos y recomendaciones disponibles acerca del empleo de efgartigimod alfa (Vyvgart®) en adultos con miastenia gravis generalizada. MÉTODOS: Se realizó una búsqueda bibliográfica en las principales bases de datos tales como PUBMED, LILACS, BRISA, COCHRANE, SCIELO, EMBASE, TRIPDATABASE como así también en sociedades científicas, agencias reguladoras, financiadores de salud y agencias de evaluación de tecnologías sanitarias. Se priorizó la inclusión de revisiones sistemáticas, ensayos clínicos controlados aleatorizados, evaluación de tecnología sanitaria y guías de práctica clínica de alta calidad metodológica. La fecha de búsqueda de información fue hasta el 11 de noviembre de 2023. Para la búsqueda en Pubmed se utilizó la siguiente estrategia de búsqueda: efgartigimod alfa [Supplementary Concept] OR efgartigimod alfa [tiab] OR Vyvgart[tiab]. RECOMENDACIONES: No se hallaron guías de práctica clínica actualizadas en Argentina y en el Mundo que recomienden la tecnología en la indicación evaluada, como tampoco es mencionada en el Consenso Internacional para el manejo de la Miastenia Gravis publicado en 2021.14 El Instituto Nacional para la Excelencia en Salud y Atención (NICE, sus siglas del inglés National Institute for Health and Care Excellence) de Reino Undo y la Agencia Canadiense de Medicamentos y Tecnologías en Salud (CADTH, su sigla del inglés Canadian Agency for Drugs and Technologies in Health) recientemente han comenzado a evaluar la tecnología en la indicación evaluada. CONCLUSIONES: La evidencia que sustenta la aprobación de comercialización por parte de los Estados Unidos y Europa de efgartigimod alfa como complemento a la terapia estándar para el tratamiento de adultos con miastenia gravis generalizada que tienen anticuerpos AChR positivos, se basa en un único ensayo clínico aleatorizado de fase III frente a placebo. No se hallaron estudios que comparen efgartigimod alfa frente a otras alternativas farmacológicas disponibles para la enfermedad, como tampoco frente a otros biológicos no disponibles en Argentina. El estudio identificado demostró que los adultos con miastenia gravis generalizada que tienen anticuerpos AChR positivos y reciben un primer ciclo de tratamiento con efgartigimod alfa, en complemento a la terápia estándar, alcanzan mejorías clinicamente significativas para puntuaciones específicas de la patología frente a placebo al corto plazo. No se hallaron estudios publicados que reporten sobre si estos beneficios se mantienen en el tiempo, sin embargo, existe un estudio en curso sin datos publicados aún que podría responder esta pregunta. No se hallaron guías de práctica clínica actualizadas ni evaluaciones económicas en Argentina y en el Mundo que mencionen la tecnología en la indicación evaluada. Según los precios de adquisición relevados, el costo de un ciclo con el fármaco sería de aproximadamente USD 51.201, 28 (ARS 17.915.328 noviembre/23).


Assuntos
Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Miastenia Gravis/tratamento farmacológico , Argentina , Eficácia , Análise Custo-Benefício
3.
N Engl J Med ; 387(5): 433-443, 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-35658022

RESUMO

BACKGROUND: The incidence of type 2 diabetes mellitus is increasing among youths. Once-weekly treatment with dulaglutide, a glucagon-like peptide-1 receptor agonist, may have efficacy with regard to glycemic control in youths with type 2 diabetes. METHODS: In a double-blind, placebo-controlled, 26-week trial, we randomly assigned participants (10 to <18 years of age; body-mass index [BMI], >85th percentile) being treated with lifestyle modifications alone or with metformin, with or without basal insulin, in a 1:1:1 ratio to receive once-weekly subcutaneous injections of placebo, dulaglutide at a dose of 0.75 mg, or dulaglutide at a dose of 1.5 mg. Participants were then included in a 26-week open-label extension study in which those who had received placebo began receiving dulaglutide at a weekly dose of 0.75 mg. The primary end point was the change from baseline in the glycated hemoglobin level at 26 weeks. Secondary end points included a glycated hemoglobin level of less than 7.0% and changes from baseline in the fasting glucose concentration and BMI. Safety was also assessed. RESULTS: A total of 154 participants underwent randomization. At 26 weeks, the mean glycated hemoglobin level had increased in the placebo group (0.6 percentage points) and had decreased in the dulaglutide groups (-0.6 percentage points in the 0.75-mg group and -0.9 percentage points in the 1.5-mg group, P<0.001 for both comparisons vs. placebo). At 26 weeks, a higher percentage of participants in the pooled dulaglutide groups than in the placebo group had a glycated hemoglobin level of less than 7.0% (51% vs. 14%, P<0.001). The fasting glucose concentration increased in the placebo group (17.1 mg per deciliter) and decreased in the pooled dulaglutide groups (-18.9 mg per deciliter, P<0.001), and there were no between-group differences in the change in BMI. The incidence of gastrointestinal adverse events was higher with dulaglutide therapy than with placebo. The safety profile of dulaglutide was consistent with that reported in adults. CONCLUSIONS: Treatment with dulaglutide at a once-weekly dose of 0.75 mg or 1.5 mg was superior to placebo in improving glycemic control through 26 weeks among youths with type 2 diabetes who were being treated with or without metformin or basal insulin, without an effect on BMI. (Funded by Eli Lilly; AWARD-PEDS ClinicalTrials.gov number, NCT02963766.).


Assuntos
Diabetes Mellitus Tipo 2 , Hipoglicemiantes , Adolescente , Glicemia/efeitos dos fármacos , Criança , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/tratamento farmacológico , Método Duplo-Cego , Peptídeos Semelhantes ao Glucagon/análogos & derivados , Hemoglobinas Glicadas/análise , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Injeções Subcutâneas , Insulinas/uso terapêutico , Metformina/uso terapêutico , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/uso terapêutico , Resultado do Tratamento
4.
J Clin Endocrinol Metab ; 107(2): 363-378, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34608929

RESUMO

CONTEXT: Tirzepatide substantially reduced hemoglobin A1c (HbA1c) and body weight in subjects with type 2 diabetes (T2D) compared with the glucagon-like peptide 1 receptor agonist dulaglutide. Improved glycemic control was associated with lower circulating triglycerides and lipoprotein markers and improved markers of beta-cell function and insulin resistance (IR), effects only partially attributable to weight loss. OBJECTIVE: Assess plasma metabolome changes mediated by tirzepatide. DESIGN: Phase 2b trial participants were randomly assigned to receive weekly subcutaneous tirzepatide, dulaglutide, or placebo for 26 weeks. Post hoc exploratory metabolomics and lipidomics analyses were performed. SETTING: Post hoc analysis. PARTICIPANTS: 259 subjects with T2D. INTERVENTION(S): Tirzepatide (1, 5, 10, 15 mg), dulaglutide (1.5 mg), or placebo. MAIN OUTCOME MEASURE(S): Changes in metabolite levels in response to tirzepatide were assessed against baseline levels, dulaglutide, and placebo using multiplicity correction. RESULTS: At 26 weeks, a higher dose tirzepatide modulated a cluster of metabolites and lipids associated with IR, obesity, and future T2D risk. Branched-chain amino acids, direct catabolic products glutamate, 3-hydroxyisobutyrate, branched-chain ketoacids, and indirect byproducts such as 2-hydroxybutyrate decreased compared to baseline and placebo. Changes were significantly larger with tirzepatide compared with dulaglutide and directly proportional to reductions of HbA1c, homeostatic model assessment 2-IR indices, and proinsulin levels. Proportional to metabolite changes, triglycerides and diglycerides were lowered significantly compared to baseline, dulaglutide, and placebo, with a bias toward shorter and highly saturated species. CONCLUSIONS: Tirzepatide reduces body weight and improves glycemic control and uniquely modulates metabolites associated with T2D risk and metabolic dysregulation in a direction consistent with improved metabolic health.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Polipeptídeo Inibidor Gástrico/administração & dosagem , Hipoglicemiantes/administração & dosagem , Adulto , Idoso , Glicemia/análise , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Polipeptídeo Inibidor Gástrico/efeitos adversos , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Peptídeos Semelhantes ao Glucagon/efeitos adversos , Peptídeos Semelhantes ao Glucagon/análogos & derivados , Hemoglobinas Glicadas/análise , Humanos , Hipoglicemiantes/efeitos adversos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/efeitos adversos , Injeções Subcutâneas , Masculino , Metabolômica , Pessoa de Meia-Idade , Receptores dos Hormônios Gastrointestinais/agonistas , Receptores dos Hormônios Gastrointestinais/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/efeitos adversos , Triglicerídeos/sangue , Triglicerídeos/metabolismo , Redução de Peso/efeitos dos fármacos , Adulto Jovem
5.
Lancet Oncol ; 22(12): 1740-1751, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34793719

RESUMO

BACKGROUND: Both innate and adaptive immune responses are important components of anticancer immunity. The CD47-SIRPα interaction could represent an important pathway used by tumour cells to evade immune surveillance. We aimed to evaluate the safety, pharmacokinetics, pharmacodynamics, and anticancer activity of evorpacept (also known as ALX148), a high-affinity CD47-blocking protein with an inactive IgG Fc region in patients with solid tumours. METHODS: We did a first-in-human, open-label, multicentre, phase 1 dose-escalation and dose-expansion study at nine hospitals and one clinic in the USA and Korea. Eligible patients for the dose-escalation and safety lead-in phases were aged 18 years or older with histological or cytological diagnosis of advanced or metastatic solid tumours with no available standard therapy, measurable or unmeasurable disease according to the Response Evaluation Criteria in Solid Tumors version 1.1, and an Eastern Cooperative Oncology Group performance status score of 0 or 1. In the dose-escalation phase, which used a 3 + 3 design, patients received intravenous evorpacept at either 0·3, 1, 3, or 10 mg/kg once per week in 21-day cycles, or 30 mg/kg once every other week in 28-day cycles. In the safety lead-in phase, patients were given the maximum tolerable dose of evorpacept from the dose-escalation phase plus either intravenous pembrolizumab (200 mg administered once every 3 weeks) or intravenous trastuzumab (8 mg/kg loading dose followed by 6 mg/kg once every 3 weeks). In the dose-expansion phase, additional patients aged 18 years or older with second-line or later-line advanced malignancies were enrolled into three parallel cohorts: those with head and neck squamous cell carcinoma (HNSCC) and those with non-small-cell lung cancer (NSCLC) were given the maximum tolerated dose of evorpacept plus intravenous pembrolizumab (200 mg administered once every 3 weeks), and patients with HER2-positive gastric or gastroesophageal junction cancer were given the maximum tolerated dose of evorpacept plus intravenous trastuzumab (8 mg/kg loading dose followed by 6 mg/kg once every 3 weeks) until disease progression, voluntary withdrawal from the study, or unacceptable toxicity. The primary endpoint was the maximum tolerated dose of evorpacept administered as a single agent and in combination with pembrolizumab or trastuzumab, measured by the occurrence of dose-limiting toxicities during the first cycle, and was assessed in all patients who had received at least one dose of evorpacept. Secondary outcomes included the safety, tolerability, and antitumour activity of evorpacept, alone or in combination with pembrolizumab or trastuzumab. The primary outcome, safety, and tolerability were assessed in all patients who had received at least one dose of evorpacept, and antitumour activity was assessed in those who recieved at least one dose of study treatment and underwent at least one post-baseline tumor assessment. This trial is registered with ClinicalTrials.gov, NCT03013218. FINDINGS: Between March 6, 2017, and Feb 21, 2019, 110 patients received single-agent evorpacept (n=28), evorpacept plus pembrolizumab (n=52), or evorpacept plus trastuzumab (n=30), and were included in the safety analysis. Median follow-up was 29·1 months (95% CI not calculable [NC]-NC) in the single-agent cohort, 27·0 months (25·1-28·8) in the evorpacept plus pembrolizumab cohort, and 32·7 months (27·0-32·7) in the evorpacept plus trastuzumab cohort. Two (7%) dose-limiting toxicities in the first cycle were reported in patients who received single-agent evorpacept; neutropenia with an associated infection in one patient with gastroesophageal junction cancer who received 3 mg/kg once per week, and thrombocytopenia with associated bleeding in one patient with pancreatic cancer who received 30 mg/kg once every other week. No maximum tolerated dose was reached; the maximum administered doses were 10 mg/kg once per week or 30 mg/kg once every other week. The 10 mg/kg once per week dose was used in the expansion cohorts in combination with pembrolizumab or trastuzumab. The most common grade 3 or worse treatment-related adverse events were thrombocytopenia with single-agent evorpacept (two [7%] patients) and evorpacept plus pembrolizumab (two [4%]), and thrombocytopenia (two [7%]) and neutropenia (two [7%]) with evorpacept plus trastuzumab. In patients who received single-agent evorpacept, four treatment-related serious adverse events were reported. Five serious treatment-related adverse events related to evorpacept plus pembrolizumab were reported, and one serious adverse event related to evorpacept plus trastuzumab was reported. In response-evaluable patients in the dose-escalation phase (n=15) receiving single-agent evorpacept once per week, four (27%) had a best overall response of stable disease (two received 0·3 mg/kg, one received 3 mg/kg, and one received 10 mg/kg); in the 11 patients who received single-agent evorpacept at the highest dose of 30 mg/kg once every other week, two (18%) had stable disease. In the dose-expansion cohort, overall responses were recorded in four (20·0%; 95% CI 5·7-43·7) of 20 patients with HNSCC who received evorpacept plus pembrolizumab, in one (5·0%; 0·1-24·9) of 20 patients with NSCLC who received evorpacept plus pembrolizumab, and in four (21·1%; 6·1-45·6) of 19 patients with gastric or gastroesophageal junction cancer who received evorpacept plus trastuzumab. INTERPRETATION: The safety findings support the use of evorpacept in combination with pembrolizumab or trastuzumab for patients with advanced solid tumours. Preliminary antitumour activity results support future investigation of evorpacept combined with pembrolizumab or trastuzumab in patients with HNSCC, gastric or gastroesophageal junction cancer, and NSCLC. FUNDING: ALX Oncology.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Idoso , Anticorpos Monoclonais Humanizados/administração & dosagem , Feminino , Seguimentos , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias/patologia , Prognóstico , Trastuzumab/administração & dosagem
6.
Int Immunopharmacol ; 101(Pt A): 108261, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34688134

RESUMO

Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by joint inflammation, synovial hyperplasia, cartilage degeneration, bone erosion, and pannus. Immunoglobulin D (IgD) plays an important role in autoimmune diseases although the content of it in vivo is low. Increased concentrations of anti-IgD autoantibodies have been detected in many RA patients. IgD-Fc-Ig fusion protein is constructed by connecting human IgD Fc domain and IgG1 Fc domain, which specifically block the IgD/ IgDR pathway and regulate the function of cells expressing IgDR to treat RA. The expression levels of Wnt5A and Frizzled 5 are higher in RA synovial tissue specimens. The complex of Wnt5A-Fzd5-LRP5/6-CTHRC1 promotes the expression of hypoxia inducible factor-1α by activating nuclear factor kappa-B (NF-κB), leading to high expression of VEGF and participating in angiogenesis. VEGF is the strongest angiogenic factor found so far. Here, we aimed to explore whether IgD participates in synovitis by binding to IgDR and regulating the activation of Wnt5A-Fzd5-CTHRC1-NF-κB signaling pathway in fibroblast synovial cells (FLSs), whether IgD-Fc-Ig fusion protein inhibits VEGF production in FLS of CIA and explore mechanism. We found that IgDR is expressed on MH7A and FLS. IgD promotes VEGF expression by activating Wnt5A-Fzd5-CTHRC1-NF-κB signaling pathway in MH7A and FLS. After activation of Fzd5 with Wnt5A, IgD-Fc-Ig reduced VEGF-A level in the culture supernatant of MH7A stimulation by IgD. The expressions of CTHRC1, Fzd5, p-P65 and VEGF in MH7A and FLSs were down-regulated after IgD-Fc-Ig treatment. IgD-Fc-Ig suppressed the combination of CTHRC1 and Fzd5 as well. By using the animal model, we demonstrated that IgD-Fc-Ig suppress ankle CTHRC1 and Fzd5 production resulted in inhibition of index of joint inflammation of CIA rats, which were consistent with vitro results. Conclusively, IgD-Fc-Ig inhibits IgD and Wnt5A-induced angiogenesis and joint inflammation by suppressing the combination of CTHRC1 and Fzd5. Our results show that IgD-Fc-Ig exerts its suppressive effect on IgD and Wnt5A by Wnt5A-Fzd5-CTHRC1-NF-κB signaling pathway.


Assuntos
Artrite Experimental/imunologia , Imunoglobulina D/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Membrana Sinovial/patologia , Sinovite/imunologia , Proteína Wnt-5a/antagonistas & inibidores , Animais , Artrite Experimental/tratamento farmacológico , Artrite Experimental/patologia , Colágeno/administração & dosagem , Colágeno/imunologia , Fibroblastos , Receptores Frizzled/metabolismo , Glicoproteínas/metabolismo , Humanos , Imunoglobulina D/administração & dosagem , Imunoglobulina D/genética , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Masculino , NF-kappa B/metabolismo , Ratos , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/imunologia , Sinoviócitos , Sinovite/tratamento farmacológico , Sinovite/patologia , Proteína Wnt-5a/metabolismo
7.
Aging (Albany NY) ; 13(18): 21962-21974, 2021 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-34537761

RESUMO

Dulaglutide, a glucagon-like peptide-1 receptor (GLP-1R) agonist, is widely used to treat diabetes. However, its effects on muscle wasting due to aging are poorly understood. In the current study, we investigated the therapeutic potential and underlying mechanism of dulaglutide in muscle wasting in aged mice. Dulaglutide improved muscle mass and strength in aged mice. Histological analysis revealed that the cross-sectional area of the tibialis anterior (TA) in the dulaglutide-treated group was thicker than that in the vehicle group. Moreover, dulaglutide increased the shift toward middle and large-sized fibers in both young and aged mice compared to the vehicle. Dulaglutide increased myofiber type I and type IIa in young (18.5% and 8.2%) and aged (1.8% and 19.7%) mice, respectively, compared to the vehicle group. Peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis, decreased but increased by dulaglutide in aged mice. The expression of atrophic factors such as myostatin, atrogin-1, and muscle RING-finger protein-1 was decreased in aged mice, whereas that of the myogenic factor, MyoD, was increased in both young and aged mice following dulaglutide treatment. In aged mice, optic atrophy-1 (OPA-1) protein was decreased, whereas Toll-like receptor-9 (TLR-9) and its targeting inflammatory cytokines (interleukin-6 [IL-6] and tumor necrosis factor-α [TNF-α]) were elevated in the TA and quadriceps (QD) muscles. In contrast, dulaglutide administration reversed this expression pattern, thereby significantly attenuating the expression of inflammatory cytokines in aged mice. These data suggest that dulaglutide may exert beneficial effects in the treatment of muscle wasting due to aging.


Assuntos
Envelhecimento/metabolismo , Peptídeos Semelhantes ao Glucagon/análogos & derivados , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Músculo Esquelético/fisiopatologia , Proteínas Recombinantes de Fusão/administração & dosagem , Sarcopenia/tratamento farmacológico , Sarcopenia/imunologia , Receptor Toll-Like 9/imunologia , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Animais , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/imunologia , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Humanos , Hipoglicemiantes/administração & dosagem , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/imunologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/imunologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/imunologia , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/imunologia , Sarcopenia/etiologia , Sarcopenia/genética , Transdução de Sinais/efeitos dos fármacos , Receptor Toll-Like 9/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
9.
Respir Res ; 22(1): 207, 2021 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-34271910

RESUMO

BACKGROUND: Alpha-1 antitrypsin (AAT) is a major serine protease inhibitor. AAT deficiency (AATD) is a genetic disorder characterized by early-onset severe emphysema. In well-selected AATD patients, therapy with plasma-derived AAT (pAAT), "augmentation therapy", provides modest clinical improvement but is perceived as cumbersome with weekly intravenous infusions. Using mouse models of emphysema, we compared the effects of a recombinant AAT-IgG1 Fc-fusion protein (AAT-Fc), which is expected to have a longer half-life following infusion, to those of pAAT. METHODS: In an elastase model of emphysema, mice received a single intratracheal instillation of porcine pancreatic elastase (PPE) or human leucocyte elastase (hLE). AAT-Fc, pAAT, or vehicle was administered intraperitoneally 1 day prior to or 3 weeks following elastase instillation. Lung function and histology assessments were performed at 7 and 32 days after elastase instillation. In a cigarette smoke (CS) model of emphysema, mice were exposed to CS daily, 5 days a week, for 6 months and AAT-Fc, pAAT, or vehicle were administered every 10 days during the last 3 months of CS exposure. Assessments were performed 3 days after the last CS exposure. Immune responses to lung elastin peptide (EP) and the effects of AAT-Fc or pAAT treatment on dendritic cell (DC) function were determined ex vivo. RESULTS: Both elastase instillation and CS exposure triggered emphysema-like alveolar enlargement, increased lung compliance, and increased markers of inflammation compared to controls. Administration of AAT-Fc either prior to or following elastase instillation or during CS exposure provided greater protection than pAAT against alveolar enlargement, lung dysfunction, and airway inflammation. When challenged ex vivo with EP, spleen mononuclear cells from elastase-exposed mice exhibited dose-dependent production of IFNγ and IL-17, suggesting immune reactivity. In co-culture experiments with splenic CD4+ T cells isolated from elastase-exposed mice, AAT-Fc treatment prior to EP-priming of bone marrow-derived dendritic cells inhibited the production of IFNγ and IL-17. CONCLUSIONS: Compared to pAAT, AAT-Fc more effectively prevented or attenuated elastase- and CS-induced models of emphysema. These effects were associated with immunomodulatory effects on DC activity. AAT-Fc may provide a therapeutic option to individuals with AATD- and CS-induced emphysema.


Assuntos
Fragmentos Fc das Imunoglobulinas/administração & dosagem , Exposição por Inalação/efeitos adversos , Elastase Pancreática/toxicidade , Enfisema Pulmonar/induzido quimicamente , Enfisema Pulmonar/tratamento farmacológico , Proteínas Recombinantes de Fusão/administração & dosagem , Fumaça/efeitos adversos , alfa 1-Antitripsina/administração & dosagem , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Elastase Pancreática/administração & dosagem , Enfisema Pulmonar/imunologia , Suínos , Nicotiana
10.
Expert Opin Drug Saf ; 20(8): 937-947, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33877003

RESUMO

Introduction: Beta-thalassemia is an autosomal recessive hereditary anemia characterized by reduced or absent ß-globin chain synthesis, affecting about 60,000 people peryear. Management for ß-thalassemia major includes regular blood transfusions followed by iron chelating therapy and drug targeting ineffective erythropoiesis.Areas covered: The safety of licensed drugs for the management of ß-thalassemia is reviewed, using evidence from clinical trials and observational research. Such drugs include the iron chelators and the erythrocyte maturation agent luspatercept. The safety of emerging treatment, such as hydroxyurea and thalidomide is also reviewed.Expert opinion: Beta-thalassemia is arare disease, and is not surprising that there are limited studies investigating the safety of drugs used in this disease. Indeed, although observational studies are the main source of drug safety information in areal-world setting, only eleven studies were identified for iron-chelators and none of these estimated the risk of agiven safety outcome. Future work should aim to better leverage existing sources of real-world datato investigate drug safety in thalassemia.


Assuntos
Receptores de Activinas Tipo II/efeitos adversos , Fragmentos Fc das Imunoglobulinas/efeitos adversos , Quelantes de Ferro/efeitos adversos , Proteínas Recombinantes de Fusão/efeitos adversos , Talassemia beta/tratamento farmacológico , Receptores de Activinas Tipo II/administração & dosagem , Hematínicos/administração & dosagem , Hematínicos/efeitos adversos , Humanos , Hidroxiureia/administração & dosagem , Hidroxiureia/efeitos adversos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Quelantes de Ferro/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Talidomida/administração & dosagem , Talidomida/efeitos adversos , Talassemia beta/fisiopatologia
11.
J Clin Pharmacol ; 61(7): 889-900, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33719084

RESUMO

Recombinant factor VIII Fc fusion protein (rFVIIIFc) has been indicated for adults and children with hemophilia A. The objective of this article was to build a population pharmacokinetic (PK) model using adult and pediatric data sets and explore relevant dosing scenarios across all ages. The activity-time profiles of rFVIIIFc from 3 clinical studies (all trials registered at https://www.clinicaltrials.gov: NCT01027377, NCT01181128, and NCT01458106) were characterized, and covariates that determine variability of rFVIIIFc PK in children and adults were identified and implemented. Data sets were pooled to estimate population PK parameters. Simulations were conducted to generate activity-time profiles at steady state (SS). The proportion of subjects maintaining SS trough >1 and >3 IU/dL and time >10 IU/dL were estimated. The rFVIIIFc model was a two-compartment model that identified weight and von Willebrand factor as significant covariates. Model-predicted SS peaks and troughs of rFVIIIFc activity-time profiles confirmed the necessity of modifying dosing in pediatric subjects. The model also predicted that the average subject in the adult and adolescent group dosed with 40 IU/kg every 2 days maintained factor VIII activity >10 IU/dL for the entire duration. Children aged <6 years and aged 6 to <12 years receiving this dose maintained factor VIII activity of >10 IU/dL for nearly two-thirds and three-quarters of their time, respectively. In conclusion, these population PK analyses characterize activity-time profiles for rFVIIIFc among pediatric and adult subjects. The model was used for simulation of clinically relevant dosing scenarios, which can provide better protection and better clinical outcomes.


Assuntos
Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacocinética , Adolescente , Fatores Etários , Peso Corporal , Criança , Simulação por Computador , Cálculos da Dosagem de Medicamento , Fator VIII/administração & dosagem , Meia-Vida , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Masculino , Taxa de Depuração Metabólica , Modelos Biológicos , Gravidade do Paciente , Proteínas Recombinantes de Fusão/administração & dosagem , Fator de von Willebrand/efeitos dos fármacos
12.
Toxicol Appl Pharmacol ; 413: 115406, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33434572

RESUMO

This study was conducted to establish the toxicological profile of combination treatment with therapeutic HPV DNA vaccines (GX-188E) and the long-acting form of recombinant human interleukin-7 fused with hybrid Fc (IL-7hyFc). GX-188E was administered intramuscularly by electroporation with or without IL-7hyFc intravaginally once per 2 weeks for 8 weeks (five times) in female Sprague-Dawley rats. Because up-regulation of immune responses and migration of antigen-specific T cells in cervicoviginal tissue were predicted as therapeutic effects, we distinguished adverse effects from therapeutic effects based on the severity of the systemic immune response, reversibility of lymphoid tissue changes, target tissue damage, and off-target immune responses. We observed that the number of neutrophils was increased, and the number of lymphocytes was decreased in the blood. Further, myofiber degeneration, necrosis, fibroplasia, and cell infiltration were observed at the GX-188E administration site. These changes were fully or partially recovered over a 4-week period. Analysis of lymphocytes in spleen revealed that CD4+ T cells and total T cells decreased in rats treated with GX-188E in combination with a high dose of IL-7hyFc (1.25 mg/animal). However, these changes were not considered adverse because they were transient and may have been related to electroporation-mediated DNA delivery or the local migration of lymphocytes induced by IL-7. Therefore, the potential toxicity of the combination of GX-188E and IL-7hyFc treatment was comparable to that of GX-188E treatment alone, and the no observed adverse effect level for GX-188E with IL-7hyFc was considered as 320 µg/animal for GX-188E and 1.25 mg/animal for IL-7hyFc.


Assuntos
Fragmentos Fc das Imunoglobulinas/toxicidade , Interleucina-7/toxicidade , Vacinas contra Papillomavirus/toxicidade , Vacinas de DNA/toxicidade , Administração Intravaginal , Animais , Biomarcadores/sangue , Biomarcadores/urina , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Eletroporação , Feminino , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Interleucina-7/administração & dosagem , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Nível de Efeito Adverso não Observado , Vacinas contra Papillomavirus/administração & dosagem , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/toxicidade , Medição de Risco , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Fatores de Tempo , Vacinas de DNA/administração & dosagem
13.
BMJ Case Rep ; 14(1)2021 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-33514621

RESUMO

Around the world, with the availability of factor concentrates, patients with haemophilia have undergone major and minor surgeries. Inhibitor development in early postoperative period leading to inadequate factor recovery and ongoing bleeding is a nightmare for both operating surgeon as well as haematologists. We describe a case of an elderly man with mild haemophilia A, who was diagnosed with pancreatic carcinoma and underwent Whipple's procedure. After an uneventful procedure, he developed high-titre inhibitors and bleeding a week after surgery posing major challenges in his management. The case highlights the importance of experienced surgeons, trained haematologists, regular monitoring of factor assay/inhibitors, adequate factor and bypassing-agent support while performing such procedures.


Assuntos
Fatores de Coagulação Sanguínea/antagonistas & inibidores , Hemofilia A/imunologia , Neoplasias Pancreáticas/cirurgia , Hemorragia Pós-Operatória/tratamento farmacológico , Idoso , Formação de Anticorpos/imunologia , Fatores de Coagulação Sanguínea/imunologia , Fator VIII/administração & dosagem , Fator VIII/uso terapêutico , Evolução Fatal , Hematologia/normas , Hemofilia A/complicações , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Hemorragias Intracranianas/complicações , Masculino , Neoplasias Pancreáticas/complicações , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/imunologia , Hemorragia Pós-Operatória/etiologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/uso terapêutico , Fatores de Risco , Cirurgiões/estatística & dados numéricos , Neoplasias Pancreáticas
14.
Thromb Haemost ; 121(6): 731-740, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33506481

RESUMO

BACKGROUND: The pharmacokinetic (PK) properties of extended half-life (EHL) factor VIII (FVIII) concentrates differ, leading to variation in the optimal dosing regimen for the individual patient. The aim of this study was to establish these PK differences for various EHL FVIII concentrates by in silico simulations. METHODS: FVIII level over time profiles of rFVIII-SC, BAY 81-8973, rFVIII-Fc, BAX 855, BAY 94-9027, and standard half-life (SHL) rFVIII concentrates were simulated for 1,000 severe hemophilia A patients during steady-state dosing of 40 IU/kg every 72 hours or dosing as advised in the summary of product characteristics (SmPC). RESULTS: Although the elimination half-life values were comparable for rFVIII-FC, BAX 855, and BAY 94-9027, a higher area under the curve (AUC; 2,779 IU/h/dL) for BAY 94-9027 was obtained. During steady-state dosing of 40 IU/kg every 72 hours, 58.5% (rFVIII-SC), 69.3% (BAY 81-8972), 89.0% (rFVIII-Fc), 83.9% (BAX 855), and 93.7% (BAY 94-9027) of the patients maintained a trough level of 1 IU/dL, compared with 56.0% for SHL rFVIII. Following dosing schemes described in the SmPC, between 51.0 and 65.4% or 23.2 and 31.1% of the patients maintained a target trough level of 1 IU/dL or 3 IU/dL, respectively. CONCLUSION: BAY 94-9027 showed the largest increase of AUC and best target attainment compared with SHL rFVIII, followed closely by BAX 855 and rFVIII-Fc. BAY 81-8973 and rFVIII-SC showed smaller PK improvements. Although our analyses increase insight into the PK of these FVIII concentrates, more studies evaluating the relation between factor levels and bleeding risk are needed.


Assuntos
Coagulantes/farmacocinética , Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Modelos Biológicos , Polietilenoglicóis/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Coagulantes/administração & dosagem , Simulação por Computador , Cálculos da Dosagem de Medicamento , Fator VIII/administração & dosagem , Meia-Vida , Hemofilia A/sangue , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Método de Monte Carlo , Polietilenoglicóis/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem
15.
Nat Commun ; 12(1): 444, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33469002

RESUMO

Glioblastoma multiforme (GBM) is the most common and aggressive form of primary brain cancer, for which effective therapies are urgently needed. Chimeric antigen receptor (CAR)-based immunotherapy represents a promising therapeutic approach, but it is often impeded by highly immunosuppressive tumor microenvironments (TME). Here, in an immunocompetent, orthotopic GBM mouse model, we show that CAR-T cells targeting tumor-specific epidermal growth factor receptor variant III (EGFRvIII) alone fail to control fully established tumors but, when combined with a single, locally delivered dose of IL-12, achieve durable anti-tumor responses. IL-12 not only boosts cytotoxicity of CAR-T cells, but also reshapes the TME, driving increased infiltration of proinflammatory CD4+ T cells, decreased numbers of regulatory T cells (Treg), and activation of the myeloid compartment. Importantly, the immunotherapy-enabling benefits of IL-12 are achieved with minimal systemic effects. Our findings thus show that local delivery of IL-12 may be an effective adjuvant for CAR-T cell therapy for GBM.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Imunoconjugados/administração & dosagem , Imunoterapia Adotiva/métodos , Interleucina-12/administração & dosagem , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/imunologia , Encéfalo/patologia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/transplante , Modelos Animais de Doenças , Receptores ErbB/imunologia , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Imunoconjugados/imunologia , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/imunologia , Injeções Intralesionais/métodos , Interleucina-12/imunologia , Imagem por Ressonância Magnética Intervencionista , Camundongos , Receptores de Antígenos Quiméricos/imunologia , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/imunologia , Linfócitos T Reguladores/imunologia , Microambiente Tumoral/imunologia
16.
J Clin Pharmacol ; 61(2): 204-210, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32827162

RESUMO

Eflapegrastim (Rolontis) is a long-acting granulocyte colony-stimulating factor (G-CSF) produced by conjugating a human G-CSF analogue and a human immunoglobulin G4 Fc fragment, linked via a polyethylene glycol linker. Weight-based doses of 45 to 270 µg/kg eflapegrastim (12.3-73.6 µg/kg as G-CSF) were evaluated in a phase 2 study in patients. Based on these results, a fixed dose of 13.2 mg eflapegrastim (3.6 mg G-CSF) was compared with pegfilgrastim (6 mg G-CSF) in 2 phase 3 studies and in a pharmacokinetic single-arm multicenter study. Absolute neutrophil count (ANC) data from these 3 studies were evaluated in patients with early-stage breast cancer who were treated with docetaxel and cyclophosphamide (n = 669). Serum concentrations of eflapegrastim were determined by enzyme-linked immunosorbent assay. Eflapegrastim systemic exposures were higher in cycle 1 than in cycle 3, likely attributable to the higher ANC in cycle 3, increasing neutrophil-mediated clearance. Eflapegrastim elicited a greater effect on ANC than pegfilgrastim in patients at ∼60% of the G-CSF dose. Body weight had no clinically significant effect on response, justifying administration of a fixed dose of eflapegrastim. The results from 2 phase 3 studies demonstrate that eflapegrastim at a fixed dose of 13.2 mg (3.6 mg G-CSF) administered once per chemotherapy cycle is effective in prophylactic treatment of chemotherapy-induced neutropenia.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fármacos Hematológicos/administração & dosagem , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Neutropenia/induzido quimicamente , Neutropenia/prevenção & controle , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Área Sob a Curva , Ciclofosfamida/efeitos adversos , Ciclofosfamida/uso terapêutico , Docetaxel/efeitos adversos , Docetaxel/uso terapêutico , Relação Dose-Resposta a Droga , Feminino , Filgrastim/administração & dosagem , Meia-Vida , Humanos , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Neutrófilos/efeitos dos fármacos , Polietilenoglicóis/administração & dosagem
17.
JCI Insight ; 6(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33332286

RESUMO

CIS43 is a potent neutralizing human mAb that targets a highly conserved "junctional" epitope in the Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP). Enhancing the durability of CIS43 in vivo will be important for clinical translation. Here, 2 approaches were used to improve the durability of CIS43 in vivo while maintaining potent neutralization. First, the Fc domain was modified with the LS mutations (CIS43LS) to increase CIS43 binding affinity for the neonatal Fc receptor (FcRn). CIS43LS and CIS43 showed comparable in vivo protective efficacy. CIS43LS had 9- to 13-fold increased binding affinity for human (6.2 nM versus 54.2 nM) and rhesus (25.1 nM versus 325.8 nM) FcRn at endosomal pH 6.0 compared with CIS43. Importantly, the half-life of CIS43LS in rhesus macaques increased from 22 days to 39 days compared with CIS43. The second approach for sustaining antibody levels of CIS43 in vivo is through adeno-associated virus (AAV) expression. Mice administered once with AAV-expressing CIS43 had sustained antibody levels of approximately 300 µg/mL and mediated protection against sequential malaria challenges up to 36 weeks. Based on these data, CIS43LS has advanced to phase I clinical trials, and AAV delivery provides a potential next-generation approach for malaria prevention.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Substituição de Aminoácidos , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacocinética , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/genética , Anticorpos Antiprotozoários/administração & dosagem , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/genética , Dependovirus/genética , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Macaca mulatta , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Proteínas de Protozoários/imunologia
18.
J Clin Pharm Ther ; 45 Suppl 1: 43-60, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32910487

RESUMO

WHAT IS KNOWN AND OBJECTIVE: In recent years, glucagon-like peptide-1 receptor agonists (GLP-1 RAs) including once-weekly (QW) formulations have been incorporated into type 2 diabetes (T2D) clinical guidelines, making it essential that pharmacists and healthcare professionals (HCPs) have a clear understanding of their safety profiles. Currently, three QW GLP-1 RAs are approved and marketed in the United States for the treatment of T2D: dulaglutide, exenatide extended-release and semaglutide. This review provides pharmacists and HCPs with collated data related to potential safety and tolerability issues when patients use QW GLP-1 RAs, enabling patient education and treatment optimization. METHODS: This is a narrative review comparing the safety and tolerability of the three QW GLP-1 RAs, using data from Phase 3 clinical trials. Extracted safety data included gastrointestinal (GI) adverse events (AEs), hypoglycaemia, injection-site reactions, pancreatitis, neoplasms, gallbladder events, and diabetic retinopathy (DR) and/or its complications (DRCs). RESULTS AND DISCUSSION: A total of 30 trials were identified for inclusion; eight were head-to-head trials involving another GLP-1 RA; of these, six compared GLP-1 RAs with different dosing regimens (QW vs once-daily or twice-daily), and two were direct QW vs QW GLP-1 RA comparisons. The most commonly reported AEs were GI events (notably nausea, vomiting and diarrhoea), but there was variation between the three QW drugs. These were generally mild-to-moderate in severity and transient. Risk of hypoglycaemia, injection-site reactions, pancreatitis, neoplasms and gallbladder events was generally low across the GLP-1 RAs investigated. Overall rates of DR or DRC were low across the trials. Only in one trial (SUSTAIN 6) there were significantly more DRC events reported in patients treated with QW semaglutide (3.0%) compared with placebo (1.8%). This was likely due to the rapid improvement in glucose control in patients with pre-existing DR enrolled within that trial. WHAT IS NEW AND CONCLUSION: This review puts the latest clinical data from the marketed QW GLP-1 RAs into context with results from older Phase 3 trials, to enable pharmacists and HCPs to make informed treatment decisions. Each of the three QW GLP-1 RAs has their own safety profile, which should be considered when choosing the optimal treatment for patients.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Hipoglicemiantes/administração & dosagem , Glicemia/efeitos dos fármacos , Preparações de Ação Retardada , Diabetes Mellitus Tipo 2/fisiopatologia , Esquema de Medicação , Exenatida/administração & dosagem , Exenatida/efeitos adversos , Exenatida/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Peptídeos Semelhantes ao Glucagon/efeitos adversos , Peptídeos Semelhantes ao Glucagon/análogos & derivados , Peptídeos Semelhantes ao Glucagon/farmacologia , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/farmacologia , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/efeitos adversos , Fragmentos Fc das Imunoglobulinas/farmacologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/farmacologia
19.
J Clin Pharm Ther ; 45 Suppl 1: 7-16, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32910488

RESUMO

WHAT IS KNOWN AND OBJECTIVE: As a highly prevalent chronic condition associated with complications and high mortality rates, it is important for pharmacists to have a comprehensive understanding of the impact of type 2 diabetes (T2D) and available treatment options. The use of injectable glucagon-like peptide 1 receptor agonists (GLP-1 RAs) is recommended as an effective and convenient treatment regimen for improving glycaemic control in individuals with T2D, with a good safety profile; however, the wider extent of its potential benefits often are unknown to clinical pharmacists. The objective of this article is to provide an overview of the impact of T2D on individuals and to discuss the multifaceted role of once-weekly (QW) GLP-1 RAs in addressing these challenges. METHODS: This is a narrative review of the published literature regarding the use of injectable GLP-1 RAs in managing health complications in people with T2D. RESULTS AND DISCUSSION: Recent findings reveal additional benefits of GLP-1 RAs in managing T2D complications, including atherosclerotic cardiovascular (CV) disease, retinopathy, neuropathy, and nephropathy. Dulaglutide and semaglutide have been shown to provide additional CV benefit in patients at high risk of CV events compared with standard of care/placebo and may offer renal protection in patients with chronic kidney disease. Cost-effectiveness studies, taking into consideration these different complications, have shown that QW GLP-1 RAs were cost-effective compared with other therapies. GLP-1 RAs may also help to improve overall health-related quality of life, reducing the risk of depression and 'diabetes distress', and limiting the risk of hypoglycaemia. WHAT IS NEW AND CONCLUSION: From the literature, this appears to be the first review of the evidence supporting the multifaceted role of QW GLP-1 RAs in T2D, with particular emphasis on their use in comorbid conditions, as well as associated potential financial and well-being benefits. The results suggest that QW GLP-1 RAs may be an attractive treatment option for improving glycaemic control in T2D, especially in individuals with (or at risk of) additional comorbidities or health complications.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Hipoglicemiantes/administração & dosagem , Diabetes Mellitus Tipo 2/complicações , Esquema de Medicação , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Peptídeos Semelhantes ao Glucagon/análogos & derivados , Peptídeos Semelhantes ao Glucagon/farmacologia , Controle Glicêmico/métodos , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/farmacologia , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/farmacologia , Qualidade de Vida , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacologia
20.
J Clin Pharm Ther ; 45 Suppl 1: 28-42, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32910489

RESUMO

WHAT IS KNOWN AND OBJECTIVE: Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) may confer a range of benefits for people with type 2 diabetes (T2D), which is reflected through their position within diabetes treatment guidelines. The objective of this narrative review is to explore the efficacy data of once-weekly (QW) GLP-1 RAs in terms of glycaemic control, body weight reduction, cardiovascular (CV) outcomes and potential renal protective effects to assist pharmacists and other healthcare professionals (HCPs) in treatment discussions with patients. METHODS: This a narrative review focused on 31 clinical trials involving the Phase 3 clinical programmes of the QW GLP-1 RAs dulaglutide, exenatide extended-release (ER) and semaglutide subcutaneous (s.c.). RESULTS AND DISCUSSION: The clinical trials were divided by their comparator arms and examined for trends. All QW GLP-1 RAs were superior to placebo for reductions in glycated haemoglobin (HbA1c ) and body weight. Data regarding QW GLP-1 RAs versus metformin were limited, likely due to metformin's use as the first-line pharmacologic for T2D. In the robust head-to-head trials of QW versus QW GLP-1 RAs, semaglutide s.c. was superior to both dulaglutide and exenatide ER regarding HbA1c and body weight; however, QW versus once-daily GLP-1 RA trials had mixed results depending on the comparators. Finally, in QW GLP-1 RA versus insulin trials, all QW GLP-1 RAs were as effective as insulin, particularly when hypoglycaemia and body weight were also considered. CV outcome trials demonstrated benefits in major adverse CV events and renal outcomes for semaglutide and dulaglutide. WHAT IS NEW AND CONCLUSION: This review collates recently published data and previously published Phase 3 results to allow pharmacists and other HCPs to understand all of the efficacy data available and the corresponding impact on treatment guidelines. QW GLP-1 RAs are emerging as important therapeutic options for people with T2D as they offer a spectrum of benefits extending beyond glycaemic control, but it is important to be aware of their efficacy differences when prescribing and discussing them with patients.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Hipoglicemiantes/administração & dosagem , Glicemia/efeitos dos fármacos , Preparações de Ação Retardada , Diabetes Mellitus Tipo 2/fisiopatologia , Esquema de Medicação , Exenatida/administração & dosagem , Exenatida/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Peptídeos Semelhantes ao Glucagon/análogos & derivados , Peptídeos Semelhantes ao Glucagon/farmacologia , Humanos , Hipoglicemiantes/farmacologia , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/farmacologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...