1
|
da Silva Araújo A, Silva Pinto AC, de Castro Lobo CL, Figueiredo MS, Menosi Gualandro SF, Olalla Saad ST, Cançado RD. Sickle Cell Disease in Brazil: Current Management. Hemoglobin 2024; 48:218-230. [PMID: 38663998 DOI: 10.1080/03630269.2024.2344790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 04/11/2024] [Accepted: 04/14/2024] [Indexed: 10/24/2024]
Abstract
Sickle cell disease (SCD) comprises inherited red blood cell disorders due to a mutation in the β-globin gene (c20A > T, pGlu6Val) and is characterized by the presence of abnormal hemoglobin, hemoglobin S, hemolysis, and vaso-occlusion. This mutation, either in a homozygous configuration or in compound states with other β-globin mutations, leads to polymerization of hemoglobin S in deoxygenated conditions, causing modifications in red blood cell shape, particularly sickling. Vaso-occlusive crisis (VOC) is the hallmark of the disease, but other severe complications may arise from repeated bouts of VOCs. SCD is considered a global health problem, and its incidence has increased in some areas of the world, particularly the Americas and Africa. Management of the disease varies according to the region of the world, mainly due to local resources and socioeconomic status. This review aimed to describe more recent data on SCD regarding available treatment options, especially in Brazil. New treatment options are expected to be available to all patients, particularly crizanlizumab, which is already approved in the country.
Collapse
Affiliation(s)
- Aderson da Silva Araújo
- Department of Hematology, Fundação de Hematologia e Hemoterapia de Pernambuco, Recife, Brazil
| | - Ana Cristina Silva Pinto
- Department of Medical Imaging, Hematology and Oncology, Hemocentro de Ribeirão Preto, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto - USP, Ribeirão Preto, Brazil
| | - Clarisse Lopes de Castro Lobo
- Department of Clinical Research, Instituto Estadual de Hematologia Arthur de Siqueira Cavalcanti (HEMORIO), Rio de Janeiro, RJ, Brazil
| | - Maria Stella Figueiredo
- Department of Clinical and Experimental Oncology, Escola Paulista de Medicina da Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | | | | | - Rodolfo Delfini Cançado
- Department of Hematology and Oncology, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Barton A, Defoe K, Jupp J, Dersch‐Mills D, Ghosh S, Leaker M. A
PHARMacist
led Initiative for the Management of Hydroxyurea Therapy in Pediatric Sickle Cell Anemia patients attending a Multidisciplinary Tertiary Hemoglobinopathy Clinic: A retrospective cohort study (
PHARMIT‐SCA
). JOURNAL OF THE AMERICAN COLLEGE OF CLINICAL PHARMACY 2021. [DOI: 10.1002/jac5.1516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Amanda Barton
- Inpatient Pharmacy Stollery Children's Hospital Edmonton Canada
| | - Kimberly Defoe
- Inpatient Pharmacy Foothills Medical Center Calgary Canada
| | - Jennifer Jupp
- Inpatient Pharmacy Alberta Children's Hospital Calgary Canada
| | | | - Sunita Ghosh
- Department of Medical Oncology Cross Cancer Institute, University of Alberta Edmonton Canada
| | - Michael Leaker
- Department of Pediatric Hematology Alberta Children's Hospital, University of Calgary Calgary Canada
| |
Collapse
|
3
|
Renella R. Biomarkers for the central nervous system complications of sickle cell disease: are we there yet? Proteomics Clin Appl 2021; 15:e2100026. [PMID: 34160906 DOI: 10.1002/prca.202100026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/19/2021] [Indexed: 11/10/2022]
Abstract
Sickle cell disease (SCD, OMIM #603903), an autosomal recessively inherited β-hemoglobinopathy, was the first human disorder delineated at a molecular level. The putative single nucleotide mutation in the HBB gene generates an abnormal hemoglobin species, which polymerizes in deoxygenated conditions causing irreversible changes in erythrocyte shape and function. Sickling erythrocytes are in turn responsible for microvascular vaso-occlusion, hemolysis and a systemic vasculopathy in patients. SCD has represented an attractive field for proteomic investigation since its methodological infancy. Clinically actionable biomarkers, especially for the prevention of cerebrovascular complications in children with the condition, are urgently needed and their discovery remains a major challenge. In this issue, Lance and colleagues report of their unbiased proteomic studies on samples from the participants of the landmark prospective, randomized, single-blind SIT trial (NEJM 2014). Their results reveal numerous brain-enriched plasma proteins specific for SCD, and for silent cerebral infarcts in this disorder, and further analyses highlight novel cellular mechanisms behind the brain damage in SCD. Although the goal of identifying reliable biomarker candidates for cerebrovascular complications could not be met, the dataset produced by the authors constitutes a significant contribution to the field and opens new horizons for further clinical and laboratory investigation.
Collapse
Affiliation(s)
- Raffaele Renella
- Pediatric Hematology-Oncology Unit, Division of Pediatrics, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
4
|
Yasara N, Premawardhena A, Mettananda S. A comprehensive review of hydroxyurea for β-haemoglobinopathies: the role revisited during COVID-19 pandemic. Orphanet J Rare Dis 2021; 16:114. [PMID: 33648529 PMCID: PMC7919989 DOI: 10.1186/s13023-021-01757-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
Background Hydroxyurea is one of the earliest drugs that showed promise in the management of haemoglobinopathies that include β-thalassaemia and sickle cell disease. Despite this, many aspects of hydroxyurea are either unknown or understudied; specifically, its usefulness in β-thalassaemia major and haemoglobin E β-thalassaemia is unclear. However, during COVID-19 pandemic, it has become a valuable adjunct to transfusion therapy in patients with β-haemoglobinopathies. In this review, we aim to explore the available in vitro and in vivo mechanistic data and the clinical utility of hydroxyurea in β-haemoglobinopathies with a special emphasis on its usefulness during the COVID-19 pandemic. Main body Hydroxyurea is an S-phase-specific drug that reversibly inhibits ribonucleoside diphosphate reductase enzyme which catalyses an essential step in the DNA biosynthesis. In human erythroid cells, it induces the expression of γ-globin, a fetal globin gene that is suppressed after birth. Through several molecular pathways described in this review, hydroxyurea exerts many favourable effects on the haemoglobin content, red blood cell indices, ineffective erythropoiesis, and blood rheology in patients with β-haemoglobinopathies. Currently, it is recommended for sickle cell disease and non-transfusion dependent β-thalassaemia. A number of clinical trials are ongoing to evaluate its usefulness in transfusion dependent β-thalassaemia. During the COVID-19 pandemic, it was widely used as an adjunct to transfusion therapy due to limitations in the availability of blood and logistical disturbances. Thus, it has become clear that hydroxyurea could play a remarkable role in reducing transfusion requirements of patients with haemoglobinopathies, especially when donor blood is a limited resource. Conclusion Hydroxyurea is a well-tolerated oral drug which has been in use for many decades. Through its actions of reversible inhibition of ribonucleoside diphosphate reductase enzyme and fetal haemoglobin induction, it exerts many favourable effects on patients with β-haemoglobinopathies. It is currently approved for the treatment of sickle cell disease and non-transfusion dependent β-thalassaemia. Also, there are various observations to suggest that hydroxyurea is an important adjunct in the treatment of transfusion dependent β-thalassaemia which should be confirmed by randomised clinical trials.
Collapse
Affiliation(s)
- Nirmani Yasara
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, 11010, Sri Lanka
| | - Anuja Premawardhena
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka.,Colombo North Teaching Hospital, Ragama, Sri Lanka
| | - Sachith Mettananda
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, 11010, Sri Lanka. .,Colombo North Teaching Hospital, Ragama, Sri Lanka.
| |
Collapse
|
5
|
Meier ER, Creary SE, Heeney MM, Dong M, Appiah-Kubi AO, Nelson SC, Niss O, Piccone C, Quarmyne MO, Quinn CT, Saving KL, Scott JP, Talati R, Latham TS, Pfeiffer A, Shook LM, Vinks AA, Lane A, McGann PT. Hydroxyurea Optimization through Precision Study (HOPS): study protocol for a randomized, multicenter trial in children with sickle cell anemia. Trials 2020; 21:983. [PMID: 33246482 PMCID: PMC7691962 DOI: 10.1186/s13063-020-04912-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/17/2020] [Indexed: 12/22/2022] Open
Abstract
Background Sickle cell disease (SCD) is a severe and devastating hematological disorder that affects over 100,000 persons in the USA and millions worldwide. Hydroxyurea is the primary disease-modifying therapy for the SCD, with proven benefits to reduce both short-term and long-term complications. Despite the well-described inter-patient variability in pharmacokinetics (PK), pharmacodynamics, and optimal dose, hydroxyurea is traditionally initiated at a weight-based dose with a subsequent conservative dose escalation strategy to avoid myelosuppression. Because the dose escalation process is time consuming and requires frequent laboratory checks, many providers default to a fixed dose, resulting in inadequate hydroxyurea exposure and suboptimal benefits for many patients. Results from a single-center trial of individualized, PK-guided dosing of hydroxyurea for children with SCD suggest that individualized dosing achieves the optimal dose more rapidly and provides superior clinical and laboratory benefits than traditional dosing strategies. However, it is not clear whether these results were due to individualized dosing, the young age that hydroxyurea treatment was initiated in the study, or both. The Hydroxyurea Optimization through Precision Study (HOPS) aims to validate the feasibility and benefits of this PK-guided dosing approach in a multi-center trial. Methods HOPS is a randomized, multicenter trial comparing standard vs. PK-guided dosing for children with SCD as they initiate hydroxyurea therapy. Participants (ages 6 months through 21 years), recruited from 11 pediatric sickle cell centers across the USA, are randomized to receive hydroxyurea either using a starting dose of 20 mg/kg/day (Standard Arm) or a PK-guided dose (Alternative Arm). PK data will be collected using a novel sparse microsampling approach requiring only 10 μL of blood collected at 3 time-points over 3 h. A protocol-guided strategy more aggressive protocols is then used to guide dose escalations and reductions in both arms following initiation of hydroxyurea. The primary endpoint is the mean %HbF after 6 months of hydroxyurea. Discussion HOPS will answer important questions about the clinical feasibility, benefits, and safety of PK-guided dosing of hydroxyurea for children with SCD with potential to change the treatment paradigm from a standard weight-based approach to one that safely and effectively optimize the laboratory and clinical response. Trial registration ClinicalTrials.gov NCT03789591. Registered on 28 December 2018.
Collapse
Affiliation(s)
- Emily R Meier
- Indiana Hemophilia and Thrombosis Center, Indianapolis, IN, USA
| | - Susan E Creary
- Division of Hematology/Oncology/BMT, Nationwide Children's Hospital, Columbus, OH, USA.,Center for Innovation in Pediatric Practice, Nationwide Children's Hospital Research Institute, The Ohio State University, Columbus, OH, USA
| | - Matthew M Heeney
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Abena O Appiah-Kubi
- Pediatric Hematology, Children's Hospitals and Clinics of Minnesota, Minneapolis and St. Paul, MN, USA
| | - Stephen C Nelson
- Division of Hematology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, MLC 7015, Cincinnati, OH, 45229, USA
| | - Omar Niss
- University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA
| | - Connie Piccone
- Case Western Reserve University School of Medicine, Department of Hematology and Oncology, University Hospitals Rainbow Babies and Children's, Cleveland, OH, USA
| | - Maa-Ohui Quarmyne
- Emory University School of Medicine & Sickle Cell Disease Program, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Charles T Quinn
- University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA
| | | | - John P Scott
- Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Teresa S Latham
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA
| | - Amanda Pfeiffer
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA
| | - Lisa M Shook
- University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Adam Lane
- University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA
| | - Patrick T McGann
- University of Cincinnati College of Medicine, Cincinnati, OH, USA. .,Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA.
| |
Collapse
|
6
|
Dong M, McGann PT. Changing the Clinical Paradigm of Hydroxyurea Treatment for Sickle Cell Anemia Through Precision Medicine. Clin Pharmacol Ther 2020; 109:73-81. [PMID: 32869281 DOI: 10.1002/cpt.2028] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022]
Abstract
Sickle cell anemia (SCA) is a common and devastating inherited blood disorder, affecting millions of people across the world. Without treatment, SCA results in tremendous morbidity and early mortality. Hydroxyurea is the primary and most well-established pharmacologic therapy with proven benefits to ameliorate the clinical course of SCA, primarily due to its ability to increase the expression of fetal hemoglobin (HbF), which prevents sickling of red blood cells. The optimal induction of HbF depends upon selection and maintenance of the proper dose that maximizes benefits and minimizes toxicity. Due to the significant interpatient variability in hydroxyurea pharmacokinetics, pharmacodynamics, and dosing, most patients treated with hydroxyurea receive suboptimal doses and have only modest treatment responses. Recognizing this variability, using a precision medicine approach, we developed and prospectively evaluated an individualized dosing model for children with SCA, designed to optimize the hydroxyurea dose and clinical response. We utilize novel laboratory methods and a sparse sampling strategy requiring only 10 μL of blood collected 15 minutes, 60 minutes, and 180 minutes after a test dose. We use Bayesian adaptive control to estimate hydroxyurea exposure and to select an individual, optimal starting dose. This dosing model has resulted in HbF responses >30-40%, levels beyond what is achieved with traditional weight-based dosing and trial and error dose escalation. This hydroxyurea dosing strategy, if widely implemented, has the potential to change the treatment paradigm of hydroxyurea therapy and improve outcomes for the millions of patients with SCA across the world.
Collapse
Affiliation(s)
- Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Patrick T McGann
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
7
|
Combination dose-escalated hydroxyurea and transfusion: an approach to conserve blood during the COVID-19 pandemic. Blood 2020; 135:2320-2322. [PMID: 32403134 PMCID: PMC7316219 DOI: 10.1182/blood.2020006582] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Nickel et al describe a single-arm trial using dose-escalated hydroxyurea and regular transfusions to prevent complications of sickle cell anemia. Preliminary results suggest that a reduction in volume of red cell requirements may be achievable.
Collapse
|
8
|
Abstract
Sickle cell disease (SCD) afflicts millions of people worldwide but is referred to as an orphan disease in the United States. Over the past several decades, there has been an increasing understanding of the pathophysiology of SCD and its complications. While most individuals with SCD in resource-rich countries survive into adulthood, the life expectancy of patients with SCD remains substantially shorter than for the general African-American population. SCD can be cured using hematopoietic stem cell transplantation and possibly gene therapy, but these treatment approaches are not available to most patients, the majority of whom reside in low- and middle-income countries. Until relatively recently, only one drug, hydroxyurea, was approved by the US Food and Drug Administration to ameliorate disease severity. Multiple other drugs (L-glutamine, crizanlizumab, and voxelotor) have recently been approved for the treatment of SCD, with several others at various stages of clinical testing. The availability of multiple agents to treat SCD raises questions related to the choice of appropriate drug therapy, combination of multiple agents, and affordability of recently approved products. The enthusiasm for new drug development provides opportunities to involve patients in low- and middle-income nations in the testing of potentially disease-modifying therapies and has the potential to contribute to capacity building in these environments. Demonstration that these agents, alone or in combination, can prevent or decrease end-organ damage would provide additional evidence for the role of drug therapies in improving outcomes in SCD.
Collapse
Affiliation(s)
- Parul Rai
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kenneth I. Ataga
- Center for Sickle Cell Disease, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
9
|
Salinas Cisneros G, Thein SL. Recent Advances in the Treatment of Sickle Cell Disease. Front Physiol 2020; 11:435. [PMID: 32508672 PMCID: PMC7252227 DOI: 10.3389/fphys.2020.00435] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/08/2020] [Indexed: 12/31/2022] Open
Abstract
Sickle cell anemia (SCA) was first described in the Western literature more than 100 years ago. Elucidation of its molecular basis prompted numerous biochemical and genetic studies that have contributed to a better understanding of its pathophysiology. Unfortunately, the translation of such knowledge into developing treatments has been disproportionately slow and elusive. In the last 10 years, discovery of BCL11A, a major γ-globin gene repressor, has led to a better understanding of the switch from fetal to adult hemoglobin and a resurgence of efforts on exploring pharmacological and genetic/genomic approaches for reactivating fetal hemoglobin as possible therapeutic options. Alongside therapeutic reactivation of fetal hemoglobin, further understanding of stem cell transplantation and mixed chimerism as well as gene editing, and genomics have yielded very encouraging outcomes. Other advances have contributed to the FDA approval of three new medications in 2017 and 2019 for management of sickle cell disease, with several other drugs currently under development. In this review, we will focus on the most important advances in the last decade.
Collapse
Affiliation(s)
- Gabriel Salinas Cisneros
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States.,Division of Hematology and Oncology, Children's National Medical Center, Washington, DC, United States
| | - Swee L Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
10
|
Olubiyi OO, Olagunju MO, Strodel B. Rational Drug Design of Peptide-Based Therapies for Sickle Cell Disease. Molecules 2019; 24:E4551. [PMID: 31842406 PMCID: PMC6943517 DOI: 10.3390/molecules24244551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 11/16/2022] Open
Abstract
Sickle cell disease (SCD) is a group of inherited disorders affecting red blood cells, which is caused by a single mutation that results in substitution of the amino acid valine for glutamic acid in the sixth position of the β-globin chain of hemoglobin. These mutant hemoglobin molecules, called hemoglobin S, can polymerize upon deoxygenation, causing erythrocytes to adopt a sickled form and to suffer hemolysis and vaso-occlusion. Until recently, only two drug therapies for SCD, which do not even fully address the manifestations of SCD, were approved by the United States (US) Food and Drug Administration. A third treatment was newly approved, while a monoclonal antibody preventing vaso-occlusive crises is also now available. The complex nature of SCD manifestations provides multiple critical points where drug discovery efforts can be and have been directed. These notwithstanding, the need for new therapeutic approaches remains high and one of the recent efforts includes developments aimed at inhibiting the polymerization of hemoglobin S. This review focuses on anti-sickling approaches using peptide-based inhibitors, ranging from individual amino acid dipeptides investigated 30-40 years ago up to more promising 12- and 15-mers under consideration in recent years.
Collapse
Affiliation(s)
- Olujide O. Olubiyi
- Institute of Complex Systems: Structural Biochemistry, Forschungszentrum Jülich, 52425 Jülich, Germany; (M.O.O.); (B.S.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife 220282, Nigeria
| | - Maryam O. Olagunju
- Institute of Complex Systems: Structural Biochemistry, Forschungszentrum Jülich, 52425 Jülich, Germany; (M.O.O.); (B.S.)
| | - Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry, Forschungszentrum Jülich, 52425 Jülich, Germany; (M.O.O.); (B.S.)
- Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
11
|
Optimizing Hydroxyurea Treatment for Sickle Cell Disease Patients: The Pharmacokinetic Approach. J Clin Med 2019; 8:jcm8101701. [PMID: 31623213 PMCID: PMC6833033 DOI: 10.3390/jcm8101701] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Hydroxyurea (HU) is a FDA- and EMA-approved drug that earned an important place in the treatment of patients with severe sickle cell anemia (SCA) by showing its efficacy in many studies. This medication is still underused due to fears of physicians and families and must be optimized. Methods: We analyzed our population and identified HU pharmacokinetic (PK) parameters in order to adapt treatment in the future. Working with a pediatric population, we searched for the most indicative sampling time to reduce the number of samples needed. Results: Nine children treated by HU for severe SCA were included for this PK study. HU quantification was made using a validated gas chromatography/mass spectrometry (GC/MS) method. Biological parameters (of effectiveness and compliance) and clinical data were collected. None of the nine children reached the therapeutic target defined by Dong et al. as an area under the curve (AUC) = 115 h.mg/L; four patients were suspected to be non-compliant. Only two patients had an HbF over 20%. The 2 h sample was predictive of the medication exposure (r2 = 0.887). Conclusions: It is urgent to be more efficient in the treatment of SCA, and pharmacokinetics can be an important asset in SCA patients.
Collapse
|
12
|
McGann PT, Niss O, Dong M, Marahatta A, Howard TA, Mizuno T, Lane A, Kalfa TA, Malik P, Quinn CT, Ware RE, Vinks AA. Robust clinical and laboratory response to hydroxyurea using pharmacokinetically guided dosing for young children with sickle cell anemia. Am J Hematol 2019; 94:871-879. [PMID: 31106898 DOI: 10.1002/ajh.25510] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 01/01/2023]
Abstract
Hydroxyurea is FDA-approved and now increasingly used for children with sickle cell anemia (SCA), but dosing strategies, pharmacokinetic (PK) profiles, and treatment responses for individual patients are highly variable. Typical weight-based dosing with step-wise escalation to maximum tolerated dose (MTD) leads to predictable laboratory and clinical benefits, but often takes 6 to 12 months to achieve. The Therapeutic Response Evaluation and Adherence Trial (TREAT, NCT02286154) was a single-center study designed to prospectively validate a novel personalized PK-guided hydroxyurea dosing strategy with a primary endpoint of time to MTD. Enrolled participants received a single oral 20 mg/kg dose of hydroxyurea, followed by a sparse PK sampling approach with three samples collected over three hours. Analysis of individual PK data into a population PK model generated a starting dose that targets the MTD. The TREAT cohort (n = 50) was young, starting hydroxyurea at a median age of 11 months (IQR 9-26 months), and PK-guided starting doses were high (27.7 ± 4.9 mg/kg/d). Time to MTD was 4.8 months (IQR 3.3-9.3), significantly shorter than comparison studies (p < 0.0001), thus meeting the primary endpoint. More remarkably, the laboratory response for participants starting with a PK-guided dose was quite robust, achieving higher hemoglobin (10.1 ± 1.3 g/dL) and HbF (33.3 ± 9.1%) levels than traditional dosing. Though higher than traditional dosing, PK-guided doses were safe without excess hematologic toxicities. Our data suggest early initiation of hydroxyurea, using a personalized dosing strategy for children with SCA, provides laboratory and clinical response beyond what has been seen historically, with traditional weight-based dosing.
Collapse
Affiliation(s)
- Patrick T. McGann
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
| | - Omar Niss
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
| | - Min Dong
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
| | - Anu Marahatta
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
| | - Thad A. Howard
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
| | - Tomoyuki Mizuno
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
| | - Adam Lane
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
| | - Theodosia A. Kalfa
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
| | - Punam Malik
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
| | - Charles T. Quinn
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
| | - Russell E. Ware
- Division of HematologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
| | - Alexander A. Vinks
- Department of PediatricsUniversity of Cincinnati College of Medicine Cincinnati Ohio
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical Center Cincinnati Ohio
| |
Collapse
|
13
|
Inusa BPD, Hsu LL, Kohli N, Patel A, Ominu-Evbota K, Anie KA, Atoyebi W. Sickle Cell Disease-Genetics, Pathophysiology, Clinical Presentation and Treatment. Int J Neonatal Screen 2019; 5:20. [PMID: 33072979 PMCID: PMC7510211 DOI: 10.3390/ijns5020020] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 04/24/2019] [Indexed: 12/28/2022] Open
Abstract
Sickle cell disease (SCD) is a monogenetic disorder due to a single base-pair point mutation in the β-globin gene resulting in the substitution of the amino acid valine for glutamic acid in the β-globin chain. Phenotypic variation in the clinical presentation and disease outcome is a characteristic feature of the disorder. Understanding the pathogenesis and pathophysiology of the disorder is central to the choice of therapeutic development and intervention. In this special edition for newborn screening for haemoglobin disorders, it is pertinent to describe the genetic, pathologic and clinical presentation of sickle cell disease as a prelude to the justification for screening. Through a systematic review of the literature using search terms relating to SCD up till 2019, we identified relevant descriptive publications for inclusion. The scope of this review is mainly an overview of the clinical features of pain, the cardinal symptom in SCD, which present following the drop in foetal haemoglobin as young as five to six months after birth. The relative impact of haemolysis and small-vessel occlusive pathology remains controversial, a combination of features probably contribute to the different pathologies. We also provide an overview of emerging therapies in SCD.
Collapse
Affiliation(s)
- Baba P. D. Inusa
- Paediatric Haematology, Evelina London Children’s Hospital, Guy’s and St Thomas NHS Trust, London SE1 7EH, UK
- Correspondence:
| | - Lewis L. Hsu
- Pediatric Hematology-Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Neeraj Kohli
- Haematology, Guy’s and St Thomas NHS Trust, London SE1 7EH, UK
| | | | - Kilali Ominu-Evbota
- Paediatrics Department, Basildon and Thurrock University Hospitals, NHS Foundation Trust, Basildon SS16 5NL, UK
| | - Kofi A. Anie
- Haematology and Sickle Cell Centre, London North West University Healthcare NHS Trust, London NW10 7NS, UK
| | - Wale Atoyebi
- Department of Clinical Haematology, Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
14
|
Miller RE, Brown DS, Keith SW, Hegarty SE, Setty Y, Campbell CM, McCahan SM, Gayen-Betal S, Byck H, Stuart M. Quantitative sensory testing in children with sickle cell disease: additional insights and future possibilities. Br J Haematol 2019; 185:925-934. [PMID: 30924134 PMCID: PMC6563447 DOI: 10.1111/bjh.15876] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022]
Abstract
Quantitative sensory testing (QST) is used in a variety of pain disorders to characterize pain and predict prognosis and response to specific therapies. In this study, we aimed to confirm results in the literature documenting altered QST thresholds in sickle cell disease (SCD) and assess the test-retest reliability of results over time. Fifty-seven SCD and 60 control subjects aged 8-20 years underwent heat and cold detection and pain threshold testing using a Medoc TSAII. Participants were tested at baseline and 3 months; SCD subjects were additionally tested at 6 months. An important facet of our study was the development and use of a novel QST modelling approach, allowing us to model all data together across modalities. We have not demonstrated significant differences in thermal thresholds between subjects with SCD and controls. Thermal thresholds were consistent over a 3- to 6-month period. Subjects on whom hydroxycarbamide (HC) was initiated shortly before or after baseline testing (new HC users) exhibited progressive decreases in thermal sensitivity from baseline to 6 months, suggesting that thermal testing may be sensitive to effective therapy to prevent vasoocclusive pain. These findings inform the use of QST as an endpoint in the evaluation of preventative pain therapies.
Collapse
Affiliation(s)
- Robin E Miller
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Dawn S Brown
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Scott W Keith
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sarah E Hegarty
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yamaja Setty
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Claudia M Campbell
- Department of Psychiatry and Behavioral Health, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Suzanne M McCahan
- Bioinformatics Core Facility, Nemours Biomedical Research, Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Suhita Gayen-Betal
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Hal Byck
- Department of Pediatrics, Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Marie Stuart
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| |
Collapse
|
15
|
Wienert B, Martyn GE, Funnell APW, Quinlan KGR, Crossley M. Wake-up Sleepy Gene: Reactivating Fetal Globin for β-Hemoglobinopathies. Trends Genet 2018; 34:927-940. [PMID: 30287096 DOI: 10.1016/j.tig.2018.09.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/23/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022]
Abstract
Disorders in hemoglobin (hemoglobinopathies) were the first monogenic diseases to be characterized and remain among the most common and best understood genetic conditions. Moreover, the study of the β-globin locus provides a textbook example of developmental gene regulation. The fetal γ-globin genes (HBG1/HBG2) are ordinarily silenced around birth, whereupon their expression is replaced by the adult β-globin genes (HBB primarily and HBD). Over 50 years ago it was recognized that mutations that cause lifelong persistence of fetal γ-globin expression ameliorate the debilitating effects of mutations in β-globin. Since then, research has focused on therapeutically reactivating the fetal γ-globin genes. Here, we summarize recent discoveries, focusing on the influence of genome editing technologies, including CRISPR-Cas9, and emerging gene therapy approaches.
Collapse
Affiliation(s)
- Beeke Wienert
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW) Sydney, Sydney, New South Wales, Australia; Innovative Genomics Institute, University of California, Berkeley, CA, USA; Present address: Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Gabriella E Martyn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW) Sydney, Sydney, New South Wales, Australia
| | - Alister P W Funnell
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW) Sydney, Sydney, New South Wales, Australia; Present address: Altius Institute for Biomedical Sciences, Seattle, WA, USA
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW) Sydney, Sydney, New South Wales, Australia
| | - Merlin Crossley
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW) Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
16
|
Inusa BPD, Wale A, Hassan AA, Idhate T, Dogara L, Ijei I, Qin Y, Anie K, Lawson JO, Hsu L. Low-dose hydroxycarbamide therapy may offer similar benefit as maximum tolerated dose for children and young adults with sickle cell disease in low-middle-income settings. F1000Res 2018; 7. [PMID: 30228870 PMCID: PMC6124375 DOI: 10.12688/f1000research.14589.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2018] [Indexed: 12/14/2022] Open
Abstract
The multiple clinical benefits of hydroxycarbamide in sickle cell disease are supported by a large body of evidence. The maximum tolerated dose (MTD) is the regimen recommended by guidelines from a panel of National Heart, Lung, and Blood Institute (NHLBI) experts, but other dosage regimens have been used in babies (BABY-HUG) 9 to 18 months old (20 mg/kg per day) and developing countries such as India (10 mg/kg per day); however, there has been no direct comparison of the efficacy, effectiveness, or cost-effectiveness of these different regimens. The purpose of this review was to investigate the current situation with various hydroxycarbamide regimens with particular relevance to low-middle-income countries. In regard to methodology, a literature review was undertaken by using multiple databases in PubMed and Google and the search terms included sickle cell disease, hydroxyurea, hydroxycarbamide, sickle cell anaemia, low-middle-income countries, Sub-Saharan Africa, and India. Although MTD regimens have been widely used in research, especially within North America, clinical trials elsewhere tend to use fixed-dose regimens. In a survey of haematologists across Europe and Africa, 60% (75% response rate) did not use the MTD regimen for hydroxycarbamide treatment of sickle cell disease. The recommendations are (1) for practical purposes to commence using fixed-dose hydroxycarbamide in line with BABY-HUG recommendations and then (2) to consider or propose a trial comparing MTD escalation with various fixed doses and to include as end points health-related quality of life, haemoglobin F levels, adherence, and cost-effectiveness.
Collapse
Affiliation(s)
- Baba Psalm Duniya Inusa
- Paediatric Haematology, Evelina London Children's Hospital, Guy's and St Thomas NHS Foundation Trust, London, UK
| | - Atoyebi Wale
- Department of Haematology, Oxford University Teaching Hospital, Oxford, UK
| | - Abdul Aziz Hassan
- Department of Haematology & Blood Transfusion, Faculty of Basic Clinical Sciences, College of Health Sciences, Ahmadu Bello University & ABU Teaching Hospital, Zaria, Nigeria
| | - Tushar Idhate
- Division of Paediatric Haematology and Oncology, Department of Paediatrics, Mahatma Gandhi Mission Medical College and Hospital, Aurangabad, India
| | - Livingstone Dogara
- Haematology and Blood Transfusion, Faculty of Clinical Sciences, Kaduna State University College of Medicine, Kaduna State University, Kaduna, Nigeria
| | - Ifeoma Ijei
- Haematology and Blood Transfusion, Faculty of Clinical Sciences, Kaduna State University College of Medicine, Kaduna State University, Kaduna, Nigeria
| | - Yewen Qin
- Paediatrics Department, University Hospital, Lewisham and Greenwich NHS Trust, King's College London, London, UK
| | - Kofi Anie
- Haematology and Sickle Cell Centre, London North West University Healthcare NHS Trust, London, UK.,Imperial College London, London, UK
| | | | - Lewis Hsu
- Pediatric Sickle Cell Center, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
17
|
Guilliams KP, Fields ME, Ragan DK, Eldeniz C, Binkley MM, Chen Y, Comiskey LS, Doctor A, Hulbert ML, Shimony JS, Vo KD, McKinstry RC, An H, Lee JM, Ford AL. Red cell exchange transfusions lower cerebral blood flow and oxygen extraction fraction in pediatric sickle cell anemia. Blood 2018; 131:1012-1021. [PMID: 29255068 PMCID: PMC5833262 DOI: 10.1182/blood-2017-06-789842] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 12/04/2017] [Indexed: 01/13/2023] Open
Abstract
Blood transfusions are the mainstay of stroke prevention in pediatric sickle cell anemia (SCA), but the physiology conferring this benefit is unclear. Cerebral blood flow (CBF) and oxygen extraction fraction (OEF) are elevated in SCA, likely compensating for reduced arterial oxygen content (CaO2). We hypothesized that exchange transfusions would decrease CBF and OEF by increasing CaO2, thereby relieving cerebral oxygen metabolic stress. Twenty-one children with SCA receiving chronic transfusion therapy (CTT) underwent magnetic resonance imaging before and after exchange transfusions. Arterial spin labeling and asymmetric spin echo sequences measured CBF and OEF, respectively, which were compared pre- and posttransfusion. Volumes of tissue with OEF above successive thresholds (36%, 38%, and 40%), as a metric of regional metabolic stress, were compared pre- and posttransfusion. Transfusions increased hemoglobin (Hb; from 9.1 to 10.3 g/dL; P < .001) and decreased Hb S (from 39.7% to 24.3%; P < .001). Transfusions reduced CBF (from 88 to 82.4 mL/100 g per minute; P = .004) and OEF (from 34.4% to 31.2%; P < .001). At all thresholds, transfusions reduced the volume of peak OEF found in the deep white matter, a location at high infarct risk in SCA (P < .001). Reduction of elevated CBF and OEF, both globally and regionally, suggests that CTT mitigates infarct risk in pediatric SCA by relieving cerebral metabolic stress at patient- and tissue-specific levels.
Collapse
Affiliation(s)
| | | | | | - Cihat Eldeniz
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; and
| | - Michael M Binkley
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, St. Louis, MO
| | | | | | | | | | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; and
| | - Katie D Vo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; and
| | - Robert C McKinstry
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; and
| | - Hongyu An
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; and
| | - Jin-Moo Lee
- Department of Neurology
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; and
| | | |
Collapse
|
18
|
Dembélé A, Toure B, Sarro Y, Guindo A, Fané B, Offredo L, Kené S, Conaré I, Tessougué O, Traoré Y, Badiaga Y, Sidibé M, Diabaté D, Coulibaly M, Kanta M, Ranque B, Diallo D. Prévalence et facteurs de risque de la rétinopathie drépanocytaire dans un centre de suivi drépanocytaire d’Afrique subsaharienne. Rev Med Interne 2017; 38:572-577. [DOI: 10.1016/j.revmed.2017.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/12/2017] [Accepted: 01/23/2017] [Indexed: 11/30/2022]
|
19
|
Dong M, Mizuno T, Vinks AA. Opportunities for model-based precision dosing in the treatment of sickle cell anemia. Blood Cells Mol Dis 2017; 67:143-147. [PMID: 28807656 DOI: 10.1016/j.bcmd.2017.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 08/07/2017] [Indexed: 12/16/2022]
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.
Collapse
Affiliation(s)
- Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
20
|
Marahatta A, Ware RE. Hydroxyurea: Analytical techniques and quantitative analysis. Blood Cells Mol Dis 2017; 67:135-142. [PMID: 28847416 DOI: 10.1016/j.bcmd.2017.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 08/07/2017] [Indexed: 11/25/2022]
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.
Collapse
Affiliation(s)
- Anu Marahatta
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Russell E Ware
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
21
|
Hulbert ML, Panepinto JA, Scott JP, Liem RI, Cook LJ, Simmons T, Brousseau DC. Red blood cell transfusions during sickle cell anemia vaso-occlusive crises: a report from the magnesium in crisis (MAGiC) study. Transfusion 2017; 57:1891-1897. [PMID: 28500682 DOI: 10.1111/trf.14155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/27/2017] [Accepted: 04/03/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Little is known about red blood cell (RBC) transfusion practices for children hospitalized for a sickle cell vaso-occlusive pain crisis (VOC). We hypothesized that transfusion would be associated with the development of acute chest syndrome (ACS), lower hemoglobin (Hb) concentration, and lack of hydroxyurea therapy. STUDY DESIGN AND METHODS This is a secondary analysis of all children admitted for a sickle cell pain crisis enrolled in the Magnesium in Crisis (MAGiC) randomized trial; all had HbSS or S-β0 thalassemia. ACS development and transfusion administration were prospectively collected during the parent trial. All Hb values during the hospitalization were recorded, as was parent report of child receiving hydroxyurea. Relative risks (RRs) of transfusion were compared between groups. RESULTS Of 204 enrolled children, 40 (19.6%) received a transfusion. Of the 30 children who developed ACS, 22 (73.3%) received transfusions compared to 18 of 174 (10.3%) without ACS: the RR of transfusion in children with ACS was 7.1 (95% confidence interval [CI], 4.4-11.5). Among those without ACS, the lowest Hb was most strongly associated with transfusions: RR was 3.1 (95% CI 2.0 - 4.7) for each 1 g/dL decrease in lowest Hb. In a binary recursive partitioning model for those without ACS, a lowest recorded Hb level of less than 6.3 g/dL was significantly associated with transfusion during admission (p < 0.01). Hydroxyurea use was not associated with transfusions in any analysis. CONCLUSION ACS increased the RR of transfusion in children hospitalized for VOC sevenfold. In children without ACS, transfusion was associated with lowest Hb concentration, particularly Hb concentration of less than 6.3 g/dL.
Collapse
Affiliation(s)
- Monica L Hulbert
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Julie A Panepinto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - J Paul Scott
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Robert I Liem
- Department of Pediatrics, Northwestern University School of Medicine, Chicago, Illinois
| | - Lawrence J Cook
- University of Utah/Pediatric Emergency Care Applied Research Network Data Coordinating Center, Salt Lake City, Utah
| | - Timothy Simmons
- University of Utah/Pediatric Emergency Care Applied Research Network Data Coordinating Center, Salt Lake City, Utah
| | - David C Brousseau
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | |
Collapse
|
22
|
Treating sickle cell disease by targeting HbS polymerization. Blood 2017; 129:2719-2726. [PMID: 28385699 DOI: 10.1182/blood-2017-02-765891] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/21/2017] [Indexed: 01/11/2023] Open
Abstract
Although the root cause of sickle cell disease is the polymerization of hemoglobin S (HbS) to form fibers that make red cells less flexible, most drugs currently being assessed in clinical trials are targeting the downstream sequelae of this primary event. Less attention has been devoted to investigation of the multiple ways in which fiber formation can be inhibited. In this article, we describe the molecular rationale for 5 distinct approaches to inhibiting polymerization and also discuss progress with the few antipolymerization drugs currently in clinical trials.
Collapse
|
23
|
Cannas G, Poutrel S, Thomas X. Hydroxycarbamine: from an Old Drug Used in Malignant Hemopathies to a Current Standard in Sickle Cell Disease. Mediterr J Hematol Infect Dis 2017; 9:e2017015. [PMID: 28293403 PMCID: PMC5333733 DOI: 10.4084/mjhid.2017.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/20/2017] [Indexed: 01/31/2023] Open
Abstract
While hydroxycarbamide (hydroxyurea, HU) has less and fewer indications in malignant hemopathies, it represents the only widely used drug which modifies sickle cell disease pathogenesis. Clinical experience with HU for patients with sickle cell disease has been accumulated over the past 25 years in Western countries. The review of the literature provides increasing support for safety and efficacy in both children and adults for reducing acute vaso-occlusive events including pain episodes and acute chest syndrome. No increased incidence of leukemia and teratogenicity was demonstrated. HU has become the standard-of-care for sickle cell anemia but remains underused. Barriers to its use should be identified and overcome.
Collapse
Affiliation(s)
- Giovanna Cannas
- Hospices Civils de Lyon, Department of Internal Medicine, Edouard Herriot Hospital, Lyon, France
- Claude Bernard University Lyon 1, Laboratoire Interuniversitaire de Biologie de la Motricité EA7424, Equipe ‘Vascular biology and red blood cell’, Villeurbanne, France
| | - Solène Poutrel
- Hospices Civils de Lyon, Department of Internal Medicine, Edouard Herriot Hospital, Lyon, France
| | - Xavier Thomas
- Hospices Civils de Lyon, Hematology Department, Lyon-Sud Hospital, Pierre Bénite, France
| |
Collapse
|
24
|
Green NS, Manwani D, Qureshi M, Ireland K, Sinha A, Smaldone AM. Decreased fetal hemoglobin over time among youth with sickle cell disease on hydroxyurea is associated with higher urgent hospital use. Pediatr Blood Cancer 2016; 63:2146-2153. [PMID: 27573582 PMCID: PMC5072999 DOI: 10.1002/pbc.26161] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/28/2016] [Accepted: 06/30/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Hydroxyurea (HU) induces dose-dependent increased fetal hemoglobin (HbF) for sickle cell disease (SCD). Large deviation from historical personal best (PBest) HbF, a clinic-based version of maximum dose, may identify a subset with suboptimal HU adherence over time. PROCEDURE Retrospective clinical data from youth ages 10-18 years prescribed HU at two centers were extracted from medical records at three time points: pre-HU initiation, PBest and a recent assessment. Decrease from PBest HbF of 20% or more at recent assessment despite stable dosing was designated as high deviation from PBest. Acute hospital use was compared between 1-year periods, pre-HU and ±6 months for PBest and recent assessment. Groups were compared using descriptive and bivariate nonparametric statistics. RESULTS Seventy-five youth, mean HU duration 5.9 years, met eligibility criteria. Mean ages of HU initiation, PBest and recent assessment were 8.0, 10.9 and 13.9 years, respectively. Despite stable dosing, average HbF of 19.5% at PBest overall declined by 31.8% at recent assessment. PBest HbF declined by 11.7 and 40.1% in two groups, the latter comprised 70.7% of the sample, had lower pre-HU and recent HbF and higher dosing. They experienced more urgent hospital use during the year framing recent assessment than during PBest; these findings were supported by sensitivity analysis. CONCLUSIONS Decline from PBest HbF is a novel approach to assess HU effectiveness, is common among youth and may represent suboptimal adherence. Larger prospective studies using additional adherence measures are needed to confirm our approach of tracking HbF deviation over time and to define an appropriate cutoff.
Collapse
Affiliation(s)
- Nancy S. Green
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, Columbia University Medical Center, New York, New York
| | - Deepa Manwani
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Albert Einstein Medical School and Montefiore Hospital, New York, New York
| | - Mahvish Qureshi
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, Columbia University Medical Center, New York, New York
| | - Karen Ireland
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Albert Einstein Medical School and Montefiore Hospital, New York, New York
| | - Arpan Sinha
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Albert Einstein Medical School and Montefiore Hospital, New York, New York
| | - Arlene M. Smaldone
- School of Nursing, Columbia University Medical Center, New York, New York,College of Dental Medicine, CUMC
| |
Collapse
|
25
|
Sickle cell anemia in sub-Saharan Africa: advancing the clinical paradigm through partnerships and research. Blood 2016; 129:155-161. [PMID: 27821508 DOI: 10.1182/blood-2016-09-702324] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/03/2016] [Indexed: 01/12/2023] Open
|
26
|
Bello-Manga H, DeBaun MR, Kassim AA. Epidemiology and treatment of relative anemia in children with sickle cell disease in sub-Saharan Africa. Expert Rev Hematol 2016; 9:1031-1042. [PMID: 27677923 DOI: 10.1080/17474086.2016.1240612] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Sickle cell disease (SCD) is the most common inherited hemoglobinopathy in the world, with the majority of cases in sub-Saharan Africa. Concomitant nutritional deficiencies, infections or exposure to environmental toxins exacerbate chronic anemia in children with SCD. The resulting relative anemia is associated with increased risk of strokes, poor cognitive function and impaired growth. It may also attenuate optimal response to hydroxyurea therapy, the only effective and practical treatment option for SCD in sub-Saharan Africa. This review will focus on the epidemiology, clinical sequelae, and treatment of relative anemia in children with SCD living in low and middle-income countries in sub-Saharan Africa. Areas covered: The causes and treatment of relative anemia in children with SCD in sub-Saharan Africa. The MEDLINE database was searched using medical subject headings (MeSH) and keywords for articles regarding relative anemia in children with SCD in sub-Saharan Africa. Expert commentary: Anemia due to nutritional deficiencies and infectious diseases such as helminthiasis and malaria are prevalent in sub-Saharan Africa. Their co-existence in children with SCD increases morbidity and mortality. Therefore, preventing, diagnosing and treating the underlying cause of this relative anemia will improve SCD-related outcomes in children in sub-Saharan Africa.
Collapse
Affiliation(s)
- Halima Bello-Manga
- a Department of Hematology and Blood Transfusion , Barau Dikko Teaching hospital/Kaduna State University , Kaduna , Nigeria
| | - Michael R DeBaun
- b Department of Pediatrics, Division of Hematology/Oncology, Vanderbilt-Meharry Center for Excellence in Sickle Cell Disease , Vanderbilt University Medical Center , Nashville , TN , USA
| | - Adetola A Kassim
- c Department of Medicine, Division of Hematology/Oncology, Vanderbilt-Meharry Center for Excellence in Sickle Cell Disease , Vanderbilt University Medical Center , Nashville , TN , USA
| |
Collapse
|
27
|
Genetic Modifiers of White Blood Cell Count, Albuminuria and Glomerular Filtration Rate in Children with Sickle Cell Anemia. PLoS One 2016; 11:e0164364. [PMID: 27711207 PMCID: PMC5053442 DOI: 10.1371/journal.pone.0164364] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/24/2016] [Indexed: 11/19/2022] Open
Abstract
Discovery and validation of genetic variants that influence disease severity in children with sickle cell anemia (SCA) could lead to early identification of high-risk patients, better screening strategies, and intervention with targeted and preventive therapy. We hypothesized that newly identified genetic risk factors for the general African American population could also impact laboratory biomarkers known to contribute to the clinical disease expression of SCA, including variants influencing the white blood cell count and the development of albuminuria and abnormal glomerular filtration rate. We first investigated candidate genetic polymorphisms in well-characterized SCA pediatric cohorts from three prospective NHLBI-supported clinical trials: HUSTLE, SWiTCH, and TWiTCH. We also performed whole exome sequencing to identify novel genetic variants, using both a discovery and a validation cohort. Among candidate genes, DARC rs2814778 polymorphism regulating Duffy antigen expression had a clear influence with significantly increased WBC and neutrophil counts, but did not affect the maximum tolerated dose of hydroxyurea therapy. The APOL1 G1 polymorphism, an identified risk factor for non-diabetic renal disease, was associated with albuminuria. Whole exome sequencing discovered several novel variants that maintained significance in the validation cohorts, including ZFHX4 polymorphisms affecting both the leukocyte and neutrophil counts, as well as AGGF1, CYP4B1, CUBN, TOR2A, PKD1L2, and CD163 variants affecting the glomerular filtration rate. The identification of robust, reliable, and reproducible genetic markers for disease severity in SCA remains elusive, but new genetic variants provide avenues for further validation and investigation.
Collapse
|
28
|
Marahatta A, Megaraj V, McGann PT, Ware RE, Setchell KDR. Stable-Isotope Dilution HPLC-Electrospray Ionization Tandem Mass Spectrometry Method for Quantifying Hydroxyurea in Dried Blood Samples. Clin Chem 2016; 62:1593-1601. [PMID: 27694393 DOI: 10.1373/clinchem.2016.263715] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/23/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND Sickle cell anemia (SCA) is a life-threatening blood disorder characterized by the presence of sickle-shaped erythrocytes. Hydroxyurea is currently the only US Food and Drug Administration-approved treatment and there is a need for a convenient method to monitor compliance and hydroxyurea concentrations, especially in pediatric SCA patients. METHODS We describe a novel approach to the determination of hydroxyurea concentrations in dried whole blood collected on DMPK-C cards or volumetric absorptive microsampling (VAMS) devices. Hydroxyurea was quantified by electrospray ionization LC-MS/MS using [13C15N2]hydroxyurea as the internal standard. Calibrators were prepared in whole blood applied to DMPK-C cards or VAMS devices. RESULTS Calibration curves for blood hydroxyurea measured from DMPK-C cards and VAMS devices were linear over the range 0.5-60 μg/mL. Interassay and intraassay CVs were <15% for blood collected by both methods, and the limit of detection was 5 ng/mL. Whole blood hydroxyurea was stable for up to 60 days on DMPK-C cards and VAMS devices when frozen at -20 °C or -80 °C. Whole blood hydroxyurea concentrations in samples collected on DMPK-C cards or VAMS devices from SCA patients were in close agreement. CONCLUSIONS This tandem mass spectrometry method permits measurement of hydroxyurea concentrations in small volumes of dried blood applied to either DMPK-C cards or VAMS devices with comparable performance. This method for measuring hydroxyurea from dried blood permits the evaluation of therapeutic drug monitoring, individual pharmacokinetics, and medication adherence using heel/finger-prick samples from pediatric patients with SCA treated with hydroxyurea.
Collapse
Affiliation(s)
- Anu Marahatta
- Division of Hematology, Department of Pediatrics, and
| | - Vandana Megaraj
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | | | - Kenneth D R Setchell
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| |
Collapse
|
29
|
Han J, Bhat S, Gowhari M, Gordeuk VR, Saraf SL. Impact of a Clinical Pharmacy Service on the Management of Patients in a Sickle Cell Disease Outpatient Center. Pharmacotherapy 2016; 36:1166-1172. [PMID: 27639254 DOI: 10.1002/phar.1834] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ambulatory care clinical pharmacy services have expanded beyond primary care settings, but literature supporting the benefits of clinical pharmacy involvement with patients who have rare diseases such as sickle cell disease (SCD) is lacking. Hydroxyurea is the only agent approved by the U.S. Food and Drug Administration for the treatment of SCD; full benefit in controlling pain episodes and other complications is achieved through monitored escalation to a maximum tolerated dose. The primary objective of this analysis was to evaluate the impact of a newly implemented clinical pharmacy service on the management of patients with SCD. We performed a retrospective cross-sectional analysis of 385 adults with SCD who received care between January 1, 2014, and December 31, 2014, at a single Sickle Cell Outpatient Center that implemented a clinical pharmacy service in August 2013. Data were collected on hydroxyurea dose escalation, immunization completion rates, and health maintenance metrics (screening for nephropathy with microalbuminuria testing, retinopathy with annual retinal examinations, and pulmonary hypertension with echocardiography). The impact of the clinical pharmacy service on quality measurements was evaluated by using univariate and multivariate analyses. The number of pharmacist encounters, defined as a clinic visit when a clinical pharmacist interacted with a patient as documented in the medical records, was associated with an improved hydroxyurea dose escalation rate (odds ratio [OR] 1.48, 95% confidence interval [CI] 1.07-2.05, p=0.02). Immunization rates for the 23-valent pneumococcal polysaccharide vaccine, the 13-valent pneumococcal conjugate vaccine, and influenza vaccine were 66%, 47%, and 62%, respectively. The number of pharmacist encounters was associated with improved immunization completion rates (OR 1.38, 95% CI 1.17-1.62, p<0.001). Improved screening for microalbuminuria (OR 2.14, 95% CI 1.60-2.86, p<0.001) and sickle cell retinopathy (OR 1.16, 95% CI 1.00-1.35, p=0.05) were also associated with the number of pharmacist encounters. A new clinical pharmacy service implemented in managing a rare disease, SCD, was associated with an improved hydroxyurea dose escalation rate, immunization completion rates, and health maintenance metrics.
Collapse
Affiliation(s)
- Jin Han
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois.,Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Shubha Bhat
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Michel Gowhari
- Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Victor R Gordeuk
- Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Santosh L Saraf
- Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
30
|
Rankine-Mullings AE, Little CR, Reid ME, Soares DP, Taylor-Bryan C, Knight-Madden JM, Stuber SE, Badaloo AV, Aldred K, Wisdom-Phipps ME, Latham T, Ware RE. EXpanding Treatment for Existing Neurological Disease (EXTEND): An Open-Label Phase II Clinical Trial of Hydroxyurea Treatment in Sickle Cell Anemia. JMIR Res Protoc 2016; 5:e185. [PMID: 27619954 PMCID: PMC5037317 DOI: 10.2196/resprot.5872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/21/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cerebral vasculopathy in sickle cell anemia (SCA) begins in childhood and features intracranial arterial stenosis with high risk of ischemic stroke. Stroke risk can be reduced by transcranial doppler (TCD) screening and chronic transfusion therapy; however, this approach is impractical in many developing countries. Accumulating evidence supports the use of hydroxyurea for the prevention and treatment of cerebrovascular disease in children with SCA. Recently we reported that hydroxyurea significantly reduced the conversion from conditional TCD velocities to abnormal velocities; whether hydroxyurea can be used for children with newly diagnosed severe cerebrovascular disease in place of starting transfusion therapy remains unknown. OBJECTIVE The primary objective of the EXpanding Treatment for Existing Neurological Disease (EXTEND) trial is to investigate the effect of open label hydroxyurea on the maximum time-averaged mean velocity (TAMV) after 18 months of treatment compared to the pre-treatment value. Secondary objectives include the effects of hydroxyurea on serial TCD velocities, the incidence of neurological and non-neurological events, quality of life (QOL), body composition and metabolism, toxicity and treatment response, changes to brain magnetic resonance imaging (MRI) and magnetic resonance angiography (MRA), genetic and serologic markers of disease severity, and cognitive and pulmonary function. METHODS This prospective Phase II trial will enroll children with SCA in Jamaica, between the ages of 2 and 17 years, with either conditional (170-199 cm/sec) or abnormal (≥ 200 cm/sec) TCD velocities. Oral hydroxyurea will be administered daily and escalated to the maximum tolerated dose (MTD). Participants will be seen in the Sickle Cell Unit (SCU) in Kingston, Jamaica monthly until achieving MTD, and then every 3 months. TCD will be performed every 6 months. RESULTS Currently, 43 participants have been enrolled out of a projected 50. There was one withdrawal due to immigration, with no permanent screen failures. Of the 43 enrolled, 37 participants have initiated study treatment. CONCLUSIONS This trial investigates the effects of hydroxyurea treatment at MTD in children with conditional or abnormal TCD velocities before transfusion therapy and may represent an important advance towards establishing a suitable non-transfusion protocol for stroke prevention in children with SCA. The trial outcomes will have profound significance in developing countries where the disease burden is highest. CLINICALTRIAL ClinicalTrials.gov NCT02556099; https://clinicaltrials.gov/ct2/show/NCT02556099 (Archived by WebCite at http://www.webcitation.org/6k1yMAa9G).
Collapse
Affiliation(s)
- Angela E Rankine-Mullings
- Sickle Cell Unit, Tropical Medicine Research Institute, University of the West Indies, Kingston, Jamaica.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Renella R. Clinically-oriented proteomic investigation of sickle cell disease: Opportunities and challenges. Proteomics Clin Appl 2016; 10:816-30. [DOI: 10.1002/prca.201500133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/05/2016] [Accepted: 05/02/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Raffaele Renella
- Department of Pediatrics; Centre Hospitalier Universitaire Vaudois; Lausanne Switzerland
| |
Collapse
|
32
|
Karacaoglu PK, Asma S, Korur A, Solmaz S, Buyukkurt NT, Gereklioglu C, Kasar M, Ozbalcı D, Unal S, Kaya H, Gurkan E, Yeral M, Sariturk Ç, Boga C, Ozdogu H. East Mediterranean region sickle cell disease mortality trial: retrospective multicenter cohort analysis of 735 patients. Ann Hematol 2016; 95:993-1000. [PMID: 27068408 DOI: 10.1007/s00277-016-2655-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/24/2016] [Indexed: 01/21/2023]
Abstract
Sickle cell disease (SCD), one of the most common genetic disorders worldwide, is characterized by hemolytic anemia and tissue damage from the rigid red blood cells. Although hydroxyurea and transfusion therapy are administered to treat the accompanying tissue injury, whether either one prolongs the lifespan of patients with SCD is unknown. SCD-related mortality data are available, but there are few studies on mortality-related factors based on evaluations of surviving patients. In addition, ethnic variability in patient registries has complicated detailed analyses. The aim of this study was to investigate mortality and mortality-related factors among an ethnically homogeneous population of patients with SCD. The 735 patients (102 children and 633 adults) included in this retrospective cohort study were of Eti-Turk origin and selected from 1367 patients seen at 5 regional hospitals. A central population management system was used to control for records of patient mortality. Data reliability was checked by a data supervision group. Mortality-related factors and predictors were identified in univariate and multivariate analyses using a Cox regression model with stepwise forward selection. The study group included patients with homozygous hemoglobin S (Hgb S) disease (67 %), Hb S-β(0) thalassemia (17 %), Hgb S-β(+) thalassemia (15 %), and Hb S-α thalassemia (1 %). They were followed for a median of 66 ± 44 (3-148) months. Overall mortality at 5 years was 6.1 %. Of the 45 patients who died, 44 (6 %) were adults and 1 (0.1 %) was a child. The mean age at death was 34.1 ± 10 (18-54) years for males, 40.1 ± 15 (17-64) years for females, and 36.6 ± 13 (17-64) years overall. Hydroxyurea was found to have a notable positive effect on mortality (p = 0.009). Mortality was also significantly related to hypertension and renal damage in a univariate analysis (p = 0.015 and p = 0.000, respectively). Acute chest syndrome, splenic sequestration, and prolonged painful-crisis-related multiorgan failure were the most common causes of mortality. In a multivariate analysis of laboratory values, only an elevated white blood cell count was related to mortality (p = 0.009). These data show that despite recent progress in the treatment of SCD, disease-related factors continue to result in mortality in young adult patients. Our results highlight the importance of evaluating curative treatment options for patients who have an appropriate stem cell donor in addition to improving patient care and patient education.
Collapse
Affiliation(s)
| | - Suheyl Asma
- Department of Internal Medicine, Family Medicine Division, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Aslı Korur
- Department of Internal Medicine, Family Medicine Division, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Soner Solmaz
- Department of Internal Medicine, Hematology Division, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Nurhilal Turgut Buyukkurt
- Department of Internal Medicine, Hematology Division, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Cigdem Gereklioglu
- Department of Internal Medicine, Family Medicine Division, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Mutlu Kasar
- Department of Internal Medicine, Hematology Division, Baskent University Faculty of Medicine, Ankara, Turkey
| | | | - Selma Unal
- Department of Internal Medicine, Pediatric Hematology Division, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Hasan Kaya
- Department of Internal Medicine, Hematology Division, Mustafa Kemal University Faculty of Medicine, Antakya, Turkey
| | - Emel Gurkan
- Department of Internal Medicine, Hematology Division, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Mahmut Yeral
- Department of Internal Medicine, Hematology Division, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Çagla Sariturk
- Medical statistics, Baskent University Faculty of Medicine, Adana, Turkey
| | - Can Boga
- Department of Internal Medicine, Hematology Division, Baskent University Faculty of Medicine, Ankara, Turkey.
| | - Hakan Ozdogu
- Department of Internal Medicine, Hematology Division, Baskent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
33
|
Ware RE, Davis BR, Schultz WH, Brown RC, Aygun B, Sarnaik S, Odame I, Fuh B, George A, Owen W, Luchtman-Jones L, Rogers ZR, Hilliard L, Gauger C, Piccone C, Lee MT, Kwiatkowski JL, Jackson S, Miller ST, Roberts C, Heeney MM, Kalfa TA, Nelson S, Imran H, Nottage K, Alvarez O, Rhodes M, Thompson AA, Rothman JA, Helton KJ, Roberts D, Coleman J, Bonner MJ, Kutlar A, Patel N, Wood J, Piller L, Wei P, Luden J, Mortier NA, Stuber SE, Luban NLC, Cohen AR, Pressel S, Adams RJ. Hydroxycarbamide versus chronic transfusion for maintenance of transcranial doppler flow velocities in children with sickle cell anaemia-TCD With Transfusions Changing to Hydroxyurea (TWiTCH): a multicentre, open-label, phase 3, non-inferiority trial. Lancet 2016; 387:661-670. [PMID: 26670617 PMCID: PMC5724392 DOI: 10.1016/s0140-6736(15)01041-7] [Citation(s) in RCA: 327] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND For children with sickle cell anaemia and high transcranial doppler (TCD) flow velocities, regular blood transfusions can effectively prevent primary stroke, but must be continued indefinitely. The efficacy of hydroxycarbamide (hydroxyurea) in this setting is unknown; we performed the TWiTCH trial to compare hydroxyurea with standard transfusions. METHODS TWiTCH was a multicentre, phase 3, randomised, open-label, non-inferiority trial done at 26 paediatric hospitals and health centres in the USA and Canada. We enrolled children with sickle cell anaemia who were aged 4-16 years and had abnormal TCD flow velocities (≥ 200 cm/s) but no severe vasculopathy. After screening, eligible participants were randomly assigned 1:1 to continue standard transfusions (standard group) or hydroxycarbamide (alternative group). Randomisation was done at a central site, stratified by site with a block size of four, and an adaptive randomisation scheme was used to balance the covariates of baseline age and TCD velocity. The study was open-label, but TCD examinations were read centrally by observers masked to treatment assignment and previous TCD results. Participants assigned to standard treatment continued to receive monthly transfusions to maintain 30% sickle haemoglobin or lower, while those assigned to the alternative treatment started oral hydroxycarbamide at 20 mg/kg per day, which was escalated to each participant's maximum tolerated dose. The treatment period lasted 24 months from randomisation. The primary study endpoint was the 24 month TCD velocity calculated from a general linear mixed model, with the non-inferiority margin set at 15 cm/s. The primary analysis was done in the intention-to-treat population and safety was assessed in all patients who received at least one dose of assigned treatment. This study is registered with ClinicalTrials.gov, number NCT01425307. FINDINGS Between Sept 20, 2011, and April 17, 2013, 159 patients consented and enrolled in TWiTCH. 121 participants passed screening and were then randomly assigned to treatment (61 to transfusions and 60 to hydroxycarbamide). At the first scheduled interim analysis, non-inferiority was shown and the sponsor terminated the study. Final model-based TCD velocities were 143 cm/s (95% CI 140-146) in children who received standard transfusions and 138 cm/s (135-142) in those who received hydroxycarbamide, with a difference of 4·54 (0·10-8·98). Non-inferiority (p=8·82 × 10(-16)) and post-hoc superiority (p=0·023) were met. Of 29 new neurological events adjudicated centrally by masked reviewers, no strokes were identified, but three transient ischaemic attacks occurred in each group. Magnetic resonance brain imaging and angiography (MRI and MRA) at exit showed no new cerebral infarcts in either treatment group, but worsened vasculopathy in one participant who received standard transfusions. 23 severe adverse events in nine (15%) patients were reported for hydroxycarbamide and ten serious adverse events in six (10%) patients were reported for standard transfusions. The most common serious adverse event in both groups was vaso-occlusive pain (11 events in five [8%] patients with hydroxycarbamide and three events in one [2%] patient for transfusions). INTERPRETATION For high-risk children with sickle cell anaemia and abnormal TCD velocities who have received at least 1 year of transfusions, and have no MRA-defined severe vasculopathy, hydroxycarbamide treatment can substitute for chronic transfusions to maintain TCD velocities and help to prevent primary stroke. FUNDING National Heart, Lung, and Blood Institute, National Institutes of Health.
Collapse
Affiliation(s)
- Russell E Ware
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Barry R Davis
- University of Texas School of Public Health, Houston, TX, USA
| | | | | | - Banu Aygun
- Cohen Children's Medical Center, New Hyde Park, NY, USA
| | | | - Isaac Odame
- Hospital for Sick Children, Toronto, ON, Canada
| | - Beng Fuh
- East Carolina University, Greenville, NC, USA
| | - Alex George
- Baylor College of Medicine, Houston, TX, USA
| | - William Owen
- Children's Hospital of the King's Daughters, Norfolk, VA, USA
| | | | | | | | | | | | | | | | | | - Scott T Miller
- State University of New York-Downstate, Brooklyn, NY, USA
| | | | | | | | - Stephen Nelson
- Children's Hospitals and Clinics of Minnesota, Minneapolis, MN, USA
| | | | - Kerri Nottage
- St Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | - Alexis A Thompson
- Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | | | | | - Donna Roberts
- Medical University of South Carolina, Charleston, SC, USA
| | - Jamie Coleman
- St Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | - Niren Patel
- Georgia Regents University, Augusta, GA, USA
| | - John Wood
- Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Linda Piller
- University of Texas School of Public Health, Houston, TX, USA
| | - Peng Wei
- University of Texas School of Public Health, Houston, TX, USA
| | - Judy Luden
- Medical University of South Carolina, Charleston, SC, USA
| | - Nicole A Mortier
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Susan E Stuber
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Alan R Cohen
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sara Pressel
- University of Texas School of Public Health, Houston, TX, USA
| | - Robert J Adams
- Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|