Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
2.
Toxicol Appl Pharmacol ; 449: 116113, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35691369

ABSTRACT

Hydroxyurea (HU) is the key drug to treat Sickle cell anemia (SCA). However, its treatment is associated with the liability of myelosuppression. The present study aimed to investigate the potential of epicatechin as a supplementation therapy for the symptomatic management of SCA under HU therapy. A panel of experiments were performed at first to observe epicatechin's effect on sickling and hemolytic behaviour using SCA patient's blood (ex vivo). Thereafter, the effect of HU in the presence or absence of epicatechin was investigated on cytokine inhibition in rat splenocytes (ex vivo) as well as alterations in hematological parameters and kidney function tests in rats (in vivo). Then, any effect of epicatechin on pharmacokinetic modulation of HU in rats was elucidated along with the underlying mechanism using a battery of in vitro and in vivo models. Epicatechin exhibited potent action on anti-sickling, polymerization inhibition, and erythrocyte membrane stability. It did not show any inherent hemolytic activity and reduced TNF-α level during concomitant administration with HU. Based on hematological changes in rats, epicatechin treatment aided to the beneficial effect of HU and prevented the treatment-linked disadvantageous effects of HU like neutropenia. The plasma exposure of HU was significantly augmented in rats upon simultaneous oral administration of epicatechin with HU. Down-regulation of Oatp1b2 and catalase possibly contributed to the pharmacokinetic interaction of HU. Epicatechin is found to be a promising candidate and should be explored at a reduced dose level of HU towards offsetting the dose-dependent myelosuppressive effect of HU under the frame of supplementation therapy in SCA.


Subject(s)
Anemia, Sickle Cell , Catechin , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Animals , Catechin/pharmacology , Catechin/therapeutic use , Cytokines , Erythrocyte Membrane , Hydroxyurea/pharmacokinetics , Hydroxyurea/toxicity , Rats
3.
Br J Haematol ; 194(3): 617-625, 2021 08.
Article in English | MEDLINE | ID: mdl-34227124

ABSTRACT

Hydroxyurea (hydroxycarbamide) is an effective treatment for sickle cell anaemia (SCA), but clinical responses depend primarily upon the degree of fetal haemoglobin (HbF) induction and the heterogeneity of HbF expression across erythrocytes. The number and characteristics of HbF-containing cells (F-cells) are not assessed by traditional HbF measurements. Conventional hydroxyurea dosing (e.g. fixed doses or low starting doses with stepwise escalation) produces a moderate heterocellular HbF induction, but haemolysis and clinical complications continue. Robust, pancellular HbF induction is needed to minimise or fully inhibit polymerisation of sickle haemoglobin. We treated children with hydroxyurea using an individualised, pharmacokinetics-guided regimen starting at predicted maximum tolerated dose (MTD). We observed sustained HbF induction (mean >30%) for up to 6 years, which was not dependent on genetic determinants of HbF expression. Nearly 70% of patients had ≥80% F-cells (near-pancellular), and almost half had ≥90% F-cells (pancellular). The mean HbF/F-cell content was ~12 pg. Earlier age of initiation and better medication adherence were associated with high F-cell responses. In summary, early initiation of hydroxyurea using pharmacokinetics-guided starting doses at predicted MTD can achieve sustained near-pancellular or pancellular HbF expression and should be considered an achievable goal for children with SCA treated with hydroxyurea at optimal doses. Clinical trial registration number: NCT02286154 (clinicaltrials.gov).


Subject(s)
Anemia, Sickle Cell/drug therapy , Antisickling Agents/therapeutic use , Fetal Hemoglobin/analysis , Hydroxyurea/therapeutic use , Adolescent , Antisickling Agents/administration & dosage , Antisickling Agents/pharmacokinetics , Child , Child, Preschool , Dose-Response Relationship, Drug , Drug Monitoring , Female , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/pharmacokinetics , Male , Precision Medicine
5.
Pediatr Blood Cancer ; 68(6): e29008, 2021 06.
Article in English | MEDLINE | ID: mdl-33742510

ABSTRACT

BACKGROUND: Without early initiation of disease-modifying therapy, the acute and chronic complications of sickle cell anemia (SCA) begin early in childhood and progress throughout life. Hydroxyurea is a safe and effective medication that reduces or prevents most SCA-related complications. Despite recommendations to prescribe hydroxyurea for all children with SCA as young as 9 months, utilization remains low. PROCEDURE: We completed a retrospective review of hydroxyurea-prescribing practices and associated clinical outcomes at our institution over a 10-year period before and after the 2014 National Heart, Lung, and Blood Institute (NHLBI) recommendations to use hydroxyurea for all children with SCA. RESULTS: Hydroxyurea use more than doubled within our pediatric SCA population from 43% in 2010 to 95% in 2019. The age of hydroxyurea initiation was significantly younger during 2014-2019 compared to 2010-2013 (median 2 years vs. 6 years, p ≤ .001). With this change in clinical practice, nearly all (69/71 = 97%) children born after 2013 received disease-modifying therapy by the end of 2019, primarily hydroxyurea (93%). Concurrently, the number of SCA-related admissions significantly decreased from 67/100 patient-years in 2010 to 39/100 patient-years in 2019 (p < .001). CONCLUSION: The early and universal prescription of hydroxyurea for children with SCA is the standard of care. Here, we demonstrate that a careful and deliberate commitment to follow this guideline in clinical practice is feasible and results in measurable improvements in clinical outcomes. Our approach and improved outcomes can serve as a model for other programs to expand their hydroxyurea use for more children with SCA.


Subject(s)
Anemia, Sickle Cell/drug therapy , Antisickling Agents/therapeutic use , Hydroxyurea/therapeutic use , Practice Patterns, Physicians'/statistics & numerical data , Adolescent , Antisickling Agents/adverse effects , Antisickling Agents/pharmacokinetics , Biological Transport , Child , Child, Preschool , Humans , Hydroxyurea/adverse effects , Hydroxyurea/pharmacokinetics , Infant , Infant, Newborn , Male , Retrospective Studies , Young Adult
6.
Exp Hematol ; 97: 47-56.e5, 2021 05.
Article in English | MEDLINE | ID: mdl-33677043

ABSTRACT

Polycythemia vera (PV) is a myeloproliferative neoplasm marked by hyperproliferation of the myeloid lineages and the presence of an activating JAK2 mutation. Hydroxyurea (HU) is a standard treatment for high-risk patients with PV. Because disease-driving mechanisms are thought to arise in PV stem cells, effective treatments should target primarily the stem cell compartment. We tested for the antiproliferative effect of patient treatment with HU in fluorescence-activated cell sorting-isolated hematopoietic stem/multipotent progenitor cells (HSC/MPPs) and more committed erythroid progenitors (common myeloid/megakaryocyte-erythrocyte progenitors [CMP/MEPs]) in PV using RNA-sequencing and gene set enrichment analysis. HU treatment led to significant downregulation of gene sets associated with cell proliferation in PV HSCs/MPPs, but not in PV CMP/MEPs. To explore the mechanism underlying this finding, we assessed for expression of solute carrier membrane transporters, which mediate transmembrane movement of drugs such as HU into target cells. The active HU uptake transporter OCTN1 was upregulated in HSC/MPPs compared with CMP/MEPs of untreated patients with PV, and the HU diffusion facilitator urea transporter B (UTB) was downregulated in HSC/MPPs compared with CMP/MEPs in all patient and control groups tested. These findings indicate a higher accumulation of HU within PV HSC/MPPs compared with PV CMP/MEPs and provide an explanation for the differential effects of HU in HSC/MPPs and CMP/MEPs of patients with PV. In general, the findings highlight the importance of transporter expression in linking therapeutics with human disease.


Subject(s)
Antineoplastic Agents/pharmacology , Hematopoietic Stem Cells/drug effects , Hydroxyurea/pharmacology , Membrane Transport Proteins/genetics , Polycythemia Vera/genetics , Antineoplastic Agents/pharmacokinetics , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Hematopoietic Stem Cells/pathology , Humans , Hydroxyurea/pharmacokinetics , Organic Cation Transport Proteins/genetics , Polycythemia Vera/drug therapy , Polycythemia Vera/pathology , Symporters/genetics , Tumor Cells, Cultured , Urea Transporters
7.
Am J Hematol ; 96(5): 538-544, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33534136

ABSTRACT

Neurologic complications are common in patients with sickle cell anemia (SCA), but conventional tools such as MRI and transcranial Doppler ultrasonography (TCD) do not fully assess cerebrovascular pathology. Cerebral tissue oximetry measures mixed oxygen saturation in the frontal lobes (SCT O2 ) and provides early prognostic information about tissue at risk of ischemic injury. Untreated patients with SCA have significantly lower SCT O2 than healthy controls that declines with age. Hydroxyurea is effective in preventing many SCA-related complications, but the degree to which it preserves normal neurophysiology is unclear. We analyzed participants enrolled in the Therapeutic Response Evaluation and Adherence Trial (TREAT, NCT02286154), which enrolled participants initiating hydroxyurea using individualized dosing (new cohort) and those previously taking hydroxyurea (old cohort) and was designed to monitor the long-term benefits of hydroxyurea. Cerebral oximetry was performed at baseline and annually. For the new cohort (median starting age = 12 months, n = 55), mean baseline SCT O2 was normal before starting hydroxyurea (mean 65%, 95% CI 58-72%) and significantly increased after 2 years (mean 72%, 95% CI 65-79%, p < .001). The SCT O2 for patients receiving long-term hydroxyurea (median age = 9.6 years) was normal at study entry (mean 66%, 95% CI 58-74%) and remained stable across 2 years. Both cohorts had significantly higher SCT O2 than published data from predominantly untreated SCA patients. Cerebral oximetry is a non-invasive method to assess cerebrovascular pathology that complements conventional imaging. Our results indicate that hydroxyurea suggests protection against neurophysiologic changes seen in untreated SCA.


Subject(s)
Anemia, Sickle Cell/drug therapy , Antisickling Agents/therapeutic use , Cerebrovascular Circulation/drug effects , Hydroxyurea/therapeutic use , Oximetry/methods , Adolescent , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/physiopathology , Antisickling Agents/administration & dosage , Antisickling Agents/pharmacokinetics , Antisickling Agents/pharmacology , Child , Child, Preschool , Dose-Response Relationship, Drug , Early Medical Intervention , Female , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/pharmacokinetics , Hydroxyurea/pharmacology , Infant , Male , Oximetry/instrumentation , Oxygen/blood , Oxyhemoglobins/analysis , Precision Medicine , Prospective Studies , Young Adult
8.
J Clin Pharmacol ; 61(4): 547-554, 2021 04.
Article in English | MEDLINE | ID: mdl-33029796

ABSTRACT

Total pancreatectomy with islet autotransplantation is a complex surgical approach for acute recurrent or chronic pancreatitis that frequently triggers extreme thrombocytosis (platelets ≥ 1000 × 109 /L). Thrombocytosis can be prothrombotic, so cytoreductive hydroxyurea is often initiated after this surgery; however, optimal dosing strategy and efficacy are unknown. This prospective pilot study characterized the pharmacokinetics of hydroxyurea after this procedure in children. It also compared them with previously published pediatric parameters in sickle cell anemia (SCA), the disease in which pediatric hydroxyurea pharmacokinetics have primarily been studied. Plasma hydroxyurea levels were quantified in 14 participants aged 4-19 years using high-performance liquid chromatography. Blood collections were scheduled 20 minutes, 1 hour, and 4 hours after the first dose, on pharmacokinetic day 1 (PK1), and again 2-3 months later if still on hydroxyurea (PK2). Six participants had PK1 and PK2 data at all 3 postdose timed collections, 5 only had PK1 samples, and 3 only had PK2 samples. Total pancreatectomy with islet autotransplantation participants had reduced and delayed absorption compared with sickle cell anemia participant data from the Hydroxyurea Study of Long-Term Effects, regardless of timing or dosing methodology. Total pancreatectomy with islet autotransplantation participants had different pharmacokinetic profiles at PK1 versus PK2, with lower dose-normalized exposures than previously reported in sickle cell anemia. These results suggest variability exists in hydroxyurea absorption and bioavailability in total pancreatectomy with islet autotransplantation patients, suspected to be primarily because of Roux-en-Y reconstruction, and suggest that more pharmacokinetic data are needed for scenarios when hydroxyurea is prescribed to children without sickle cell anemia.


Subject(s)
Hydroxyurea/administration & dosage , Hydroxyurea/pharmacokinetics , Islets of Langerhans Transplantation/adverse effects , Pancreatectomy/adverse effects , Thrombocytosis/etiology , Thrombocytosis/prevention & control , Adolescent , Anemia, Sickle Cell/drug therapy , Child , Child, Preschool , Female , Humans , Hydroxyurea/blood , Male , Prospective Studies , Transplantation, Autologous , Young Adult
9.
Article in English | MEDLINE | ID: mdl-32862024

ABSTRACT

Hydroxyurea (HU) is the first-ever approved drug by the United States Food and Drug Administration (USFDA) for the management of sickle cell anemia (SCA). However, its treatment is associated with severe liabilities like myelosuppression. Therefore, the aim of the present investigation was to identify phytotherapeutics through assessment of the pharmacokinetic interaction of HU with dietary bioflavonoids followed by elucidation of the same phytoconstituents for their ability to protect HU-induced toxicity in hematological profile. In this direction, we developed a sensitive ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method to estimate HU in rat plasma at first and then validated as per USFDA guidelines as there is no such precedent in the literature. A simple plasma protein precipitation method was employed for plasma sample processing. The separation was achieved in gradient mode using Syncronis HILIC column (100 × 4.6 mm, 3 µm) with a mobile phase composition of water containing 0.1% (v/v) formic acid and acetonitrile. Ionization was carried out in positive heated-electrospray ionization (H-ESI) mode. Detection was done in selected reaction monitoring (SRM) mode with m/z 77.1 > 44.4 and m/z 75.1 > 58.2 for HU and methylurea (internal standard), respectively. All the validation parameters were within the acceptable criteria. This bioanalytical method was found to be useful in assessing the preclinical pharmacokinetic interaction of HU. Concomitant administration of chrysin or quercetin with HU in rats significantly enhanced the oral exposure of HU. Lowering of total red blood cells (RBC) and hemoglobin (Hb) level by HU in rats was significantly improved in the presence of chrysin, quercetin, and naringenin. Overall, both chrysin and quercetin showed potential to be a promising phytotherapeutics for concomitant therapy with HU to combat its dose-dependent side effects.


Subject(s)
Chromatography, High Pressure Liquid/methods , Hydroxyurea/blood , Hydroxyurea/pharmacokinetics , Tandem Mass Spectrometry/methods , Animals , Drug Interactions , Flavonoids/blood , Flavonoids/pharmacokinetics , Hydroxyurea/chemistry , Linear Models , Male , Rats , Rats, Wistar , Reproducibility of Results , Sensitivity and Specificity
10.
J Pharmacol Exp Ther ; 374(3): 469-478, 2020 09.
Article in English | MEDLINE | ID: mdl-32631869

ABSTRACT

The complex pathophysiology of sickle cell anemia (SCA) involves intravascular hemolytic processes and recurrent vaso-occlusion, driven by chronic vascular inflammation, which result in the disease's severe clinical complications, including recurrent painful vaso-occlusive episodes. Hydroxyurea, the only drug frequently used for SCA therapy, is a cytostatic agent, although it appears to exert nitric oxide/soluble guanylyl cyclase (sGC) modulating activity. As new drugs that can complement or replace the use of hydroxyurea are sought to further reduce vaso-occlusive episode frequency in SCA, we investigated the effects of the sGC agonists BAY 60-2770 (sGC activator) and BAY 41-2272 (sGC stimulator) in the presence or absence of hydroxyurea on SCA vaso-occlusive mechanisms and cell recruitment both ex vivo and in vivo. These agents significantly reduced stimulated human SCA neutrophil adhesive properties ex vivo in association with the inhibition of surface ß2-integrin activation. A single administration of BAY 60-2770 or BAY 41-2272 decreased tumor necrosis factor cytokine-induced leukocyte recruitment in a mouse model of SCA vaso-occlusion. Importantly, the in vivo actions of both agonists were significantly potentiated by the coadministration of hydroxyurea. Erythroid cell fetal hemoglobin (HbF) elevation is also a major goal for SCA therapy. BAY 41-2272 but not BAY 60-2770 at the concentrations employed significantly induced γ-globin gene transcription in association with HbF production in cultured erythroleukemic cells. In conclusion, sGC agonist drugs could represent a promising approach as therapy for SCA, for use either as stand-alone treatments or in combination with hydroxyurea. SIGNIFICANCE STATEMENT: This preclinical study demonstrates that stimulators and activators of sGC are potent inhibitors of the adhesion and recruitment of leukocytes from humans and in mice with sickle cell anemia (SCA) and may represent a promising approach for diminishing vaso-occlusive episode frequency in SCA. Hydroxyurea, a drug already frequently used for treating SCA, was found to potentiate the beneficial effects of sGC agonists in in vivo studies, implying that these classes of compounds could be used alone or in combination therapy.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/metabolism , Hydroxyurea/pharmacokinetics , Soluble Guanylyl Cyclase/metabolism , Animals , Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Disease Models, Animal , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Fetal Hemoglobin/metabolism , Humans , Hydrocarbons, Fluorinated/pharmacology , K562 Cells , Male , Mice , Mice, Inbred C57BL , Pyrazoles/pharmacology , Pyridines/pharmacology , Vascular Diseases/drug therapy , Vascular Diseases/metabolism , Vasodilator Agents/pharmacology
11.
J Pediatr ; 222: 236-239, 2020 07.
Article in English | MEDLINE | ID: mdl-32171562

ABSTRACT

Lactation is contraindicated for women with sickle cell anemia receiving hydroxyurea therapy, despite sparse pharmacokinetics data. In 16 women who were lactating volunteers, we documented hydroxyurea transferred into breastmilk with a relative infant dosage of 3.4%, which is below the recommended 5%-10% safety threshold. Breastfeeding should be permitted for women taking daily oral hydroxyurea.


Subject(s)
Anemia, Sickle Cell/drug therapy , Hydroxyurea/pharmacokinetics , Lactation/drug effects , Milk, Human/metabolism , Adult , Anemia, Sickle Cell/metabolism , Antineoplastic Agents/pharmacokinetics , Antisickling Agents , Female , Humans , Milk, Human/drug effects
12.
Clin Adv Hematol Oncol ; 16(11): 750-757, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30543590

ABSTRACT

Polycythemia vera (PV) is the most common Philadelphia chromosome-negative myeloproliferative neoplasm. Whereas low-risk patients are treated with aspirin and phlebotomy, high-risk patients receive cytoreductive therapy, which most commonly consists of hydroxyurea in the United States. Concerns about the long-term safety of hydroxyurea, as well as a desire for more efficacious and targeted therapy, have led to the development of novel therapies for high-risk patients with PV. Pegylated interferon (IFN) has shown promise in phase 2 studies of PV, and preliminary data from ongoing phase 3 studies suggest noninferiority as a frontline therapy. Efficient count control, tolerability, and even molecular responses as a salvage therapy have been demonstrated. Ropeginterferon-α-2b, a monopegylated IFN with a longer half-life and less frequent dose interval compared with recombinant or pegylated IFN, is an impressive agent in development. Ruxolitinib has a proven role as second-line therapy for PV, but an ongoing trial combining ruxolitinib and IFN as salvage therapy is under way. Early-phase clinical trials have also suggested that MDM2 inhibitors such as idasanutlin and histone deacetylase inhibitors should continue in their development. If these novel agents are able to modify the natural history of PV, the treatment paradigm in newly diagnosed patients will evolve from risk-adapted or reactive treatment toward early interventions.


Subject(s)
Interferon alpha-2/therapeutic use , Interferon-alpha/therapeutic use , Polycythemia Vera/drug therapy , Polyethylene Glycols/therapeutic use , Pyrazoles/therapeutic use , Pyrrolidines/therapeutic use , para-Aminobenzoates/therapeutic use , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Half-Life , Humans , Hydroxyurea/pharmacokinetics , Hydroxyurea/therapeutic use , Interferon alpha-2/pharmacokinetics , Interferon-alpha/pharmacokinetics , Nitriles , Polycythemia Vera/metabolism , Polycythemia Vera/pathology , Polyethylene Glycols/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyrimidines , Pyrrolidines/pharmacokinetics , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Salvage Therapy/methods , para-Aminobenzoates/pharmacokinetics
13.
Br J Clin Pharmacol ; 84(7): 1478-1485, 2018 07.
Article in English | MEDLINE | ID: mdl-28884840

ABSTRACT

AIMS: The purposes of this work were to: (1) compare pharmacokinetic (PK) parameters for hydroxycarbamide in children receiving their first dose (HCnew ) vs. those receiving chronic therapy (HCchronic ), (2) assess the external validity of a published PK dosing strategy, and (3) explore the accuracy of dosing strategies based on a limited number of HC measurements. METHODS: Utilizing data from two prospective, multicenter trials of hydroxycarbamide (Pharmacokinetics of Liquid Hydroxyurea in Pediatric Patients with Sickle Cell Anemia; NCT01506544 and Single-Dose (SD) and Steady-State (SS) Pharmacokinetics of Hydroxyurea in Children and Adolescents with Sickle Cell Disease), plasma drug concentration vs. time profiles were evaluated with a model independent approach in the HCnew and HCchronic groups. Various predictive scenarios were analysed to evaluate whether systemic exposure with hydroxycarbamide could be accurately predicted. RESULTS: Absorption of hydroxycarbamide was rapid, variable and dose independent. Dose-normalized peak plasma concentrations and drug exposure (AUC) were higher, and weight-normalized apparent oral clearance was lower in the HCnew group. We assessed a PK-guided dosing strategy along with other predictive scenarios and found that inclusion of plasma samples only slightly improved the accuracy of AUC predictions when compared to a population-based method. CONCLUSIONS: Children naïve to hydroxycarbamide exhibit a different PK profile compared to children receiving chronic therapy. Accuracy of population-based dosing is sufficient to target AUCs in individual patients. Further clearance/bioavailability studies are needed to address the factors responsible for variability in the disposition of hydroxycarbamide.


Subject(s)
Anemia, Sickle Cell/drug therapy , Antisickling Agents/pharmacokinetics , Hydroxyurea/pharmacokinetics , Models, Biological , Adolescent , Anemia, Sickle Cell/blood , Antisickling Agents/administration & dosage , Area Under Curve , Biological Availability , Child , Child, Preschool , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Hydroxyurea/administration & dosage , Male , Prospective Studies
14.
Blood Cells Mol Dis ; 67: 143-147, 2017 09.
Article in English | MEDLINE | ID: mdl-28807656

ABSTRACT

Hydroxyurea is the primary pharmacotherapy to prevent complications of sickle cell anemia (SCA). Accumulated clinical experience across multiple age ranges has suggested that the use of an individualized maximum tolerated dose (MTD) will achieve optimal benefit of hydroxyurea treatment. However, the current empirical and trial-and-error approach for dose escalation often results in a lengthy titration process and is not strictly implemented in many clinics. Opportunities exist for pharmacokinetics model-based precision dosing of hydroxyurea to quickly achieve individual MTD. This review intends to introduce the use of a quantitative modeling approach including a Bayesian adaptive control strategy for the precision dosing of hydroxyurea. The rationale and practical considerations for the implementation of this approach are discussed. Future research directions with a focus on integrating specific safety and other clinical outcome endpoints into dose selection decision making are also discussed.


Subject(s)
Anemia, Sickle Cell/drug therapy , Antisickling Agents/therapeutic use , Hydroxyurea/therapeutic use , Animals , Antisickling Agents/administration & dosage , Antisickling Agents/adverse effects , Antisickling Agents/pharmacokinetics , Bayes Theorem , Dose-Response Relationship, Drug , Drug Dosage Calculations , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/adverse effects , Hydroxyurea/pharmacokinetics , Maximum Tolerated Dose , Models, Biological
15.
Blood Cells Mol Dis ; 67: 135-142, 2017 09.
Article in English | MEDLINE | ID: mdl-28847416

ABSTRACT

Hydroxyurea is a potent disease-modifying therapeutic agent with efficacy for the treatment of sickle cell anemia. When administered at once-daily oral doses that lead to mild marrow suppression, hydroxyurea leads to substantial and sustained fetal hemoglobin induction, which effectively inhibits erythrocyte sickling. When escalated to maximum tolerated dose, hydroxyurea has proven laboratory and clinical effects for both children and adults with sickle cell anemia. However, there is substantial inter-patient variability with regard to the optimal dosing regimen, as well as differences in treatment-related toxicities and responses that may be explained by hydroxyurea pharmacokinetics and pharmacogenetics. Addressing the safety and efficacy of hydroxyurea treatment requires quantitative and accurate drug analysis, and various laboratory techniques have been established. We review the historical and current analytical techniques for measuring hydroxyurea concentrations accurately, and discuss clinical settings where quantitative analysis can increase understanding and safety of this important therapeutic agent, and ultimately improve patient outcomes.


Subject(s)
Anemia, Sickle Cell/drug therapy , Antisickling Agents/pharmacokinetics , Drug Monitoring/methods , Hydroxyurea/pharmacokinetics , Animals , Antisickling Agents/administration & dosage , Antisickling Agents/therapeutic use , Chromatography, High Pressure Liquid/methods , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/therapeutic use , Mass Spectrometry/methods
16.
Pharmacogenomics ; 18(10): 995-967, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28639471

ABSTRACT

AIM: To identify the BCL11A intron-2 enhancer linkage disequilibrium (LD) block, harboring two previously identified SNPs, associating with the hydroxyurea response in ß-thalassemia patients and the functional significance of this region. MATERIALS & METHODS: Several neighboring SNPs were genotyped in our cohort. The associating LD block was identified, and its function studied in K562 erythroid cells via CRISPR/Cas9 genome editing. RESULTS: A haplotype harboring three tag SNPs correlated significantly with the HU-response and BCL11A transcript levels in the patients' reticulocytes. Two deletions encompassing this LD block significantly reduced BCL11A transcript levels in K562 cells. CONCLUSION: Our data suggest an essential role for this LD block in BCL11A expression levels and the response to hydroxyurea in ß-thalassemia patients.


Subject(s)
Carrier Proteins/genetics , Hydroxyurea/therapeutic use , Nuclear Proteins/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , beta-Thalassemia/drug therapy , Cohort Studies , Enhancer Elements, Genetic , Gene Frequency , Genotype , HEK293 Cells , Haplotypes , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/pharmacokinetics , K562 Cells , Linkage Disequilibrium , Repressor Proteins , Reticulocytes/metabolism , Transfection , beta-Thalassemia/blood , beta-Thalassemia/genetics
17.
J Pharm Biomed Anal ; 137: 213-219, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28131939

ABSTRACT

Hydroxyurea (HU) has been used in the treatment of chronic myeloid leukaemia (CML) and other myeloproliferative malignancies. Considering patient's wide variation in clinical response to HU, a new and simple liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated to monitor patients' compliance to treatment and investigate the pharmacokinetics of HU in patients with CML. Stable isotope labeled HU-13C1,15N2 was used as internal standard. Plasma samples were treated with acetonitrile to precipitate protein. The supernatant was injected directly without derivatization and separated on a hydrophilic interaction liquid chromatography column. HU was quantitatively analyzed with a mobile phase of acetonitrile-1.5mM ammonium formate (90:10, V:V) within 3min. The proposed method provided a linearity range of 1-200µg/mL. The coefficients of variation for intra- and inter-day precision were less than 2.07% and 4.28%, respectively, while the accuracy (bias) was in the range of -3.77 to 2.96%. This method was satisfactorily applied to the determination of HU in two patients with CML. It is suitable for supporting pharmacokinetic studies and clinical therapeutic monitoring.


Subject(s)
Acetonitriles/chemistry , Hydroxyurea/blood , Hydroxyurea/pharmacokinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Chromatography, High Pressure Liquid/methods , Humans , Hydrophobic and Hydrophilic Interactions , Hydroxyurea/chemistry , Limit of Detection , Reproducibility of Results , Tandem Mass Spectrometry/methods
18.
OMICS ; 20(10): 565-574, 2016 10.
Article in English | MEDLINE | ID: mdl-27636225

ABSTRACT

Sickle cell disease (SCD) is a blood disease caused by a single nucleotide substitution (T > A) in the beta globin gene on chromosome 11. The single point mutation (Glu6Val) promotes polymerization of hemoglobin S (HbS) and causes sickling of erythrocytes. Vaso-occlusive painful crises are associated with recurrent and long-term use of analgesics/opioids and hydroxyurea (HU) by people living with SCD. The present analysis offers a state-of-the-art expert review of the effectiveness of pharmacogenomics/genetics of pain management in SCD, with specific focus on HU and opioids. The literature search used the following keywords: SCD, pharmacogenomics, pharmacogenetics, pain, antalgics, opioids, morphine, and HU. The literature was scanned until March 2016, with specific inclusion of targeted landmark and background articles on SCD. Surprisingly, our review identified only a limited number of studies that addressed the genetic/genomic basis of variable responses to pain (e.g., variants in OPRM1, HMOX-1, GCH1, VEGFA COMT genes), and pharmacogenomics of antalgics and opioids (e.g., variants in OPRM1, STAT6, ABCB1, and COMT genes) in SCD. There has been greater progress made toward identifying the key genomic variants, mainly in BCL11A, HBS1L-MYB, or SAR1, which contribute to response to HU treatment. However, the complete picture on pharmacogenomic determinants of the above therapeutic phenotypes remains elusive. Strikingly, no study has been conducted in sub-Saharan Africa where majority of the patients with SCD live. This alerts the broader global life sciences community toward the existing disparities in optimal and ethical targeting of research and innovation investments for SCD specifically and precision medicine and pharmacology research broadly.


Subject(s)
Anemia, Sickle Cell/genetics , Pain/genetics , Pharmacogenomic Variants , Precision Medicine , Analgesics/adverse effects , Analgesics/pharmacokinetics , Analgesics/therapeutic use , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/therapeutic use , Anemia, Sickle Cell/drug therapy , Humans , Hydroxyurea/adverse effects , Hydroxyurea/pharmacokinetics , Hydroxyurea/therapeutic use , Pain Management , Pharmacogenetics
19.
J Clin Pharmacol ; 56(3): 298-306, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26201504

ABSTRACT

Hydroxyurea (HU) is a crucial therapy for children with sickle cell anemia, but its off-label use is a barrier to widespread acceptance. We found HU exposure is not significantly altered by liquid vs capsule formulation, and weight-based dosing schemes provide consistent exposure. HU is recommended for all children starting as young as 9 months of age with sickle cell anemia (SCA; HbSS and HbSßspan(0) thalassemia); however; a paucity of pediatric data exists regarding the pharmacokinetics (PK) or the exposure-response relationship of HU. This trial aimed to characterize the PK of HU in children and to evaluate and compare the bioavailability of a liquid vs capsule formulation. This multicenter; prospective; open-label trial enrolled 39 children with SCA who provided 682 plasma samples for PK analysis following administration of HU. Noncompartmental and population PK models are described. We report that liquid and capsule formulations of HU are bioequivalent; weight-based dosing schemes provide consistent drug exposure; and age-based dosing schemes are unnecessary. These data support the use of liquid HU in children unable to swallow capsules and in those whose weight precludes the use of fixed capsule formulations. Taken with existing safety and efficacy literature; these findings should encourage the use of HU across the spectrum of age and weight in children with SCA; and they should facilitate the expanded use of HU as recommended in the National Heart; Lung; and Blood Institute guidelines for individuals with SCA.


Subject(s)
Anemia, Sickle Cell/blood , Hydroxyurea/chemistry , Hydroxyurea/pharmacokinetics , Adolescent , Antisickling Agents/blood , Antisickling Agents/pharmacokinetics , Capsules , Child , Child, Preschool , Female , Humans , Hydroxyurea/blood , Male , Prospective Studies , Solutions , Therapeutic Equivalency
20.
Br J Clin Pharmacol ; 81(4): 742-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26615061

ABSTRACT

AIMS: Hydroxyurea has emerged as the primary disease-modifying therapy for patients with sickle cell anaemia (SCA). The laboratory and clinical benefits of hydroxyurea are optimal at maximum tolerated dose (MTD), but the current empirical dose escalation process often takes up to 12 months. The purpose of this study was to develop a pharmacokinetic-guided dosing strategy to reduce the time required to reach hydroxyurea MTD in children with SCA. METHODS: Pharmacokinetic (PK) data from the HUSTLE trial (NCT00305175) were used to develop a population PK model using non-linear mixed effects modelling (nonmem 7.2). A D-optimal sampling strategy was developed to estimate individual PK and hydroxyurea exposure (area under the concentration-time curve (AUC)). The initial AUC target was derived from HUSTLE clinical data and defined as the mean AUC at MTD. RESULTS: PK profiles were best described by a one compartment with Michaelis-Menten elimination and a transit absorption model. Body weight and cystatin C were identified as significant predictors of hydroxyurea clearance. The following clinically feasible sampling times are included in a new prospective protocol: pre-dose (baseline), 15-20 min, 50-60 min and 3 h after an initial 20 mg kg(-1) oral dose. The mean target AUC(0,∞) for initial dose titration was 115 mg l(-1)  h. CONCLUSION: We developed a PK model-based individualized dosing strategy for the prospective Therapeutic Response Evaluation and Adherence Trial (TREAT, ClinicalTrials.gov NCT02286154). This approach has the potential to optimize the dose titration of hydroxyurea therapy for children with SCA, such that the clinical benefits at MTD are achieved more quickly.


Subject(s)
Anemia, Sickle Cell/drug therapy , Antisickling Agents/pharmacokinetics , Hydroxyurea/pharmacokinetics , Patient-Specific Modeling , Antisickling Agents/administration & dosage , Antisickling Agents/therapeutic use , Area Under Curve , Child , Clinical Trials as Topic , Computer Simulation , Dose-Response Relationship, Drug , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/therapeutic use , Metabolic Clearance Rate , Precision Medicine , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL