Your browser doesn't support javascript.
loading
Oral tetrahydrouridine and decitabine for non-cytotoxic epigenetic gene regulation in sickle cell disease: A randomized phase 1 study.
Molokie, Robert; Lavelle, Donald; Gowhari, Michel; Pacini, Michael; Krauz, Lani; Hassan, Johara; Ibanez, Vinzon; Ruiz, Maria A; Ng, Kwok Peng; Woost, Philip; Radivoyevitch, Tomas; Pacelli, Daisy; Fada, Sherry; Rump, Matthew; Hsieh, Matthew; Tisdale, John F; Jacobberger, James; Phelps, Mitch; Engel, James Douglas; Saraf, Santhosh; Hsu, Lewis L; Gordeuk, Victor; DeSimone, Joseph; Saunthararajah, Yogen.
Afiliação
  • Molokie R; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • Lavelle D; Jesse Brown VA Medical Center, Chicago, Illinois, United States of America.
  • Gowhari M; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • Pacini M; Jesse Brown VA Medical Center, Chicago, Illinois, United States of America.
  • Krauz L; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • Hassan J; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • Ibanez V; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • Ruiz MA; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • Ng KP; Jesse Brown VA Medical Center, Chicago, Illinois, United States of America.
  • Woost P; Jesse Brown VA Medical Center, Chicago, Illinois, United States of America.
  • Radivoyevitch T; Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
  • Pacelli D; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America.
  • Fada S; Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, United States of America.
  • Rump M; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • Hsieh M; Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
  • Tisdale JF; Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
  • Jacobberger J; Molecular and Clinical Hematology Section, National Institutes of Health, Bethesda, Maryland, United States of America.
  • Phelps M; Molecular and Clinical Hematology Section, National Institutes of Health, Bethesda, Maryland, United States of America.
  • Engel JD; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America.
  • Saraf S; College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America.
  • Hsu LL; Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America.
  • Gordeuk V; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • DeSimone J; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
  • Saunthararajah Y; Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America.
PLoS Med ; 14(9): e1002382, 2017 Sep.
Article em En | MEDLINE | ID: mdl-28880867
ABSTRACT

BACKGROUND:

Sickle cell disease (SCD), a congenital hemolytic anemia that exacts terrible global morbidity and mortality, is driven by polymerization of mutated sickle hemoglobin (HbS) in red blood cells (RBCs). Fetal hemoglobin (HbF) interferes with this polymerization, but HbF is epigenetically silenced from infancy onward by DNA methyltransferase 1 (DNMT1). METHODS AND

FINDINGS:

To pharmacologically re-induce HbF by DNMT1 inhibition, this first-in-human clinical trial (NCT01685515) combined 2 small molecules-decitabine to deplete DNMT1 and tetrahydrouridine (THU) to inhibit cytidine deaminase (CDA), the enzyme that otherwise rapidly deaminates/inactivates decitabine, severely limiting its half-life, tissue distribution, and oral bioavailability. Oral decitabine doses, administered after oral THU 10 mg/kg, were escalated from a very low starting level (0.01, 0.02, 0.04, 0.08, or 0.16 mg/kg) to identify minimal doses active in depleting DNMT1 without cytotoxicity. Patients were SCD adults at risk of early death despite standard-of-care, randomized 32 to THU-decitabine versus placebo in 5 cohorts of 5 patients treated 2X/week for 8 weeks, with 4 weeks of follow-up. The primary endpoint was ≥ grade 3 non-hematologic toxicity. This endpoint was not triggered, and adverse events (AEs) were not significantly different in THU-decitabine-versus placebo-treated patients. At the decitabine 0.16 mg/kg dose, plasma concentrations peaked at approximately 50 nM (Cmax) and remained elevated for several hours. This dose decreased DNMT1 protein in peripheral blood mononuclear cells by >75% and repetitive element CpG methylation by approximately 10%, and increased HbF by 4%-9% (P < 0.001), doubling fetal hemoglobin-enriched red blood cells (F-cells) up to approximately 80% of total RBCs. Total hemoglobin increased by 1.2-1.9 g/dL (P = 0.01) as reticulocytes simultaneously decreased; that is, better quality and efficiency of HbF-enriched erythropoiesis elevated hemoglobin using fewer reticulocytes. Also indicating better RBC quality, biomarkers of hemolysis, thrombophilia, and inflammation (LDH, bilirubin, D-dimer, C-reactive protein [CRP]) improved. As expected with non-cytotoxic DNMT1-depletion, platelets increased and neutrophils concurrently decreased, but not to an extent requiring treatment holds. As an early phase study, limitations include small patient numbers at each dose level and narrow capacity to evaluate clinical benefits.

CONCLUSION:

Administration of oral THU-decitabine to patients with SCD was safe in this study and, by targeting DNMT1, upregulated HbF in RBCs. Further studies should investigate clinical benefits and potential harms not identified to date. TRIAL REGISTRATION ClinicalTrials.gov, NCT01685515.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Azacitidina / Tetra-Hidrouridina / Regulação da Expressão Gênica / Epigênese Genética / Inibidores Enzimáticos / Anemia Falciforme Tipo de estudo: Clinical_trials / Prognostic_studies Limite: Adult / Female / Humans / Male / Middle aged Idioma: En Revista: PLoS Med Assunto da revista: MEDICINA Ano de publicação: 2017 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Azacitidina / Tetra-Hidrouridina / Regulação da Expressão Gênica / Epigênese Genética / Inibidores Enzimáticos / Anemia Falciforme Tipo de estudo: Clinical_trials / Prognostic_studies Limite: Adult / Female / Humans / Male / Middle aged Idioma: En Revista: PLoS Med Assunto da revista: MEDICINA Ano de publicação: 2017 Tipo de documento: Article País de afiliação: Estados Unidos