Hydroxycarbamine: from an Old Drug Used in Malignant Hemopathies to a Current Standard in Sickle Cell Disease

Giovanna Cannas1,2, Solène Poutrel1 and Xavier Thomas3

1 Hospices Civils de Lyon, Department of Internal Medicine, Edouard Herriot Hospital, Lyon, France.
2 Claude Bernard University Lyon 1, Laboratoire Interuniversitaire de Biologie de la Motricité EA7424, Equipe ‘Vascular biology and red blood cell’, Villeurbanne, France.
3 Hospices Civils de Lyon, Hematology Department, Lyon-Sud Hospital, Pierre Bénite, France.

Corresponding author: Xavier Thomas, M.D., Ph.D., Hospices Civils de Lyon, Hematology  Department, Lyon-Sud Hospital, Bât. 1G, 165 chemin du Grand Revoyet, 69495 Pierre Bénite, France. Tel: +33 478862235. Fax: +33 4 72678880. E-mail: xavier.thomas@chu-lyon.fr

Published: February 15, 2017
Received: December 12, 2016
Accepted: January 20, 2017
Mediterr J Hematol Infect Dis 2017, 9(1): e2017015 DOI 10.4084/MJHID.2017.015
This article is available on PDF format at: 

This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(
https://creativecommons.org/licenses/by-nc/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

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.



Introduction

Hydroxycarbamide (Hydroxyurea, HU) was first synthesized in Germany in 1869,[1] but its potential biologic significance was not recognized until 1928.[2] It is a simple compound of the formula, H2NCONHOH, which the =C−NHOH moiety is responsible for its biological activity (Figure 1). HU is a potent inhibitor of DNA synthesis. It is antimitotic and cytotoxic depending upon the used concentration, the duration of exposure, and the sensitivity of the organism. HU is active mainly in the S-phase of the cell cycle. In the 1950s the drug was evaluated in a large number of experimental tumor models and was found to have broad anti-tumor activity against both leukemia and solid tumors.[3] Clinical trials began in the 1960s.[4] As an antineoplastic drug, HU has some advantages. It may be used with ambulatory patients and has relatively few side effects, which are relieved almost immediately after withdrawal of the drug. The drug is readily absorbed from the gastrointestinal tract following oral administration. Peak serum concentrations are reached in 1 to 2 hours, and the serum half-life is about 5.5 hours. It is rapidly excreted in the urine, and it is reported that up to 70% of the dose is excreted unchanged.[5] At present, HU has only a limited medical use in acute leukemia, consisting in reducing and controlling white blood cell count in patients with hyperleukocytosis. The principal use of HU has been as a myelosuppressive agent in the myeloproliferative syndromes. The efficacy of HU as initial therapy for chronic myeloid leukemia (CML) has been known for a number of years.[6] Since the introduction of tyrosine kinase inhibitors in the treatment of CML, hydroxyurea is essentially used in the BCR-ABL1-negative myeloproliferative neoplasms, including polycythemia vera, essential thrombocythemia, and primary myelofibrosis.[7] In high-risk patients with polycythemia vera or essential thrombocythemia, HU remains the first-line cytoreductive drug of choice, the second-line choice being represented by interferon-alpha and busulfan.[8,9] Survival is relatively long in these diseases, and risk of leukemic transformation low. Treatment with HU has not been shown to modify these favorable outcomes, while controlled clinical trials have shown increased risk of acute leukemia with the use of chlorambucil, radiophosphorus and pipobroman, and increased risk of fibrotic transformation with the use of anagrelide.[7] The introduction of new drugs should, therefore, be careful. This is particularly important when considering the use of JAK inhibitor ruxolitinib, which was recently approved for use in these pathologies. HU also remains the first-line drug of choice for myelofibrosis-associated splenomegaly, while hydroxyurea-refractory splenomegaly is often managed with ruxolitinib therapy or splenectomy.[10] In addition to its use as an anti-cancer agent, HU has found some marginal applications in dermatology.[11]

Table 1 Figure 1. Structure of hydroxycarbamine (hydroxyurea, HU).

While HU is an old drug that can still be used to control essential thrombocythemia and polycythemia vera in patients with high-risk disease, it has emerged over the last decades as the primary disease-modifying therapy for sickle cell anemia, a non-malignant inherited disease. The purpose of this short review is to provide the reader a comprehensive understanding of HU and to reinforce the fact that HU is a safe and effective medication for the treatment of sickle cell disease.

Sickle Cell Disease: Historical Considerations

Sickle cell anemia, first described by James B Herrick in 1910,[12] is the first inherited disease identified at the molecular level. In 1949, Linus Pauling confirmed an intrinsic dissimilarity in the hemoglobin from patients with sickle cell anemia on electrophoretic mobility patterns.[13] Because of the heterozygote state, sickle cell trait, appeared to persist in some populations with prevalence as high as 20%-40% and the sickle cell trait allele frequency overlapped with malarial endemicity, AC Allison hypothesized that sickle hemoglobin (HbS) must confer a selective advantage of malarial resistance in the carrier state.[14] A recent meta-analysis confirmed a strong protective advantage of sickle cell trait for Plasmodium falciparum malaria, suggesting that HbS does not protect against infection itself, but rather to progression to clinical malaria and its childhood associated-mortality.[15] Although not elucidated, the suggested mechanisms involved in this epidemiologic observation comprise a protective effect through enhanced immunity, increased clearance of infected erythrocytes, and reduced parasite growth.  In 1956, VM Ingram discovered a single amino acid substitution in HbS.[16] The genetic basis for the abnormal hemoglobin was a single base-pair change (A → T) in the β-globin gene, resulting in a substitution of a valine for glutamic acid at position 6. Structural changes promote polymerization into long fibrils, distorting the red cell into a sickle shape, leading to erythrocytes dehydrated, rigid and prone to hemolysis, and so to occluding the microvasculature causing acute and chronic tissue ischemia and injury. It took then until the 1970s for systematic research into the laboratory screening techniques and clinical sequelae of sickling disorders to be prioritized.[17] At that time, only 50% of afflicted children survived into adulthood.[18] As a result of the institution of the National Sickle Cell Anemia Control Act, a Hemoglobinopathy Reference Laboratory was created to standardize techniques and elaborate screening programs.[19] By the 1990s, widespread mandatory newborn screening and the routine administration of penicillin to prevent pneumococcal sepsis increased childhood survival to over 90%.[20] Currently, the most common screening techniques include sickle solubility testing, hemoglobin electrophoresis, high-performance liquid chromatography, and isoelectric focusing, each with their own advantages and limitations. Recent advances in technology have also allowed for detection of sickle cell trait from DNA through exome sequencing.[21,22] Indeed misclassification of individuals with sickle cell trait and sickle cell disease in early case reports led to confusing series in which sickle cell disease complications were ascribed to individuals with sickle cell trait.
No specific therapy was available until the 1970s when it was recognized that patients with increased red blood cell HbF had fewer adverse clinical events. First described as a potential therapy for sickle-cell anemia in 1984, HU enhances the production of fetal hemoglobin production in sickle erythrocytes.[23] The two most common acute morbidities in sickle cell anemia are vaso-occlusive pain crises and acute chest syndrome, corresponding to the occlusion of small vessels in the bone marrow and lungs, respectively.[24,25] Other pulmonary complications of sickle cell disease include pulmonary hypertension, pulmonary artery thrombosis, and pulmonary fibrosis, with an increased prevalence of reactive airways disease, increased tricuspid regurgitant jet velocity, sleep-disordered breathing, and nocturnal hypoxemia.[26] On a chronic basis, vaso-occlusion may damage the lungs, kidney or brain accounting ultimately for most deaths in patients with sickle cell disease.[27] Clinical studies with HU demonstrated a decreased rate of vaso-occlusive disease and acute chest syndrome, and an improved survival.[28] Consequently, HU became in 1998 the only US Food and Drug Administration-approved therapy for sickle cell disease. The European Medicines Agency authorized HU in 2007 for pediatric and adult patients with sickle cell anemia. In 2008, the Agency for Healthcare Research and Quality published a comprehensive review,[29] and a consensus conference on HU in the treatment of sickle cell disease was organized by the National Institute of Health.[30]

HU Mechanisms of Action in Sickle Cell Anemia

In sickle cell anemia, the red cells almost contain only HbS. Only a smaller population of red cells comes directly from immature progenitors, which contain the fetal hemoglobin (HbF). These nearly normal cells mitigate the damage caused by HbS.[31] Cells with high levels of HbS lose deformability when deoxygenated, leading to vascular obstruction and ischemia. Membrane damage shortens the life span of the cell leading to chronic intravascular and extravascular hemolysis. Damage red cells showed an increased adherence to vascular endothelium leading to vaso-occlusion and proliferative lesions involving many cells and factors underlying large-vessel stroke.[32] Shifting hemoglobin production from HbS to HbF represents then a major therapeutic approach to sickle cell anemia. Low level of HbF is one of the strongest predictors of morbidity and mortality in sickle cell disease.[27] The cytotoxic effect of HU reduces the production of red cells containing a high level of HbS, which tend to arise from rapidly dividing precursors, and favors the production of cells containing a high level of HbF.[32] The exact mechanism by which HU induces HbF remains unclear. The increase in HbF appears to interfere with HbS polymerization both by preventing contact between adjacent HbS molecules and by forming mixed hybrids with HbS that have greater solubility than HbS polymers.[33] HU may increase HbF indirectly by killing dividing late erythroid cells, causing recruitment of more primitive erythroid precursors which produce high levels of HbF, or by acting directly on the primitive precursors stimulating HbF production.[34] However, induction of HbF was unlikely to explain all the clinical effects of HU. Prior to any rise in HbF, sickle erythrocytes show reduced adhesion to endothelial cells. HU reduces adhesion molecule expression on sickle erythrocytes, including very late activation antigen-4 and CD36.[35] Other rheological properties of sickle erythrocytes, including erythrocyte hydration status and whole cell deformability, can be increased by HU. HU also reduces white blood cells and platelets reducing their roles in vascular injury. Neutrophilia has long been identified as a marker of severity in sickle cell disease.[27] Neutrophils release pro-inflammatory mediators involved in endothelial damage and cytokine release, which trigger sickling, and contribute to slow transit time via their adhesive properties and an increase in blood viscosity.[36] The drug also produces nitric oxide, which stimulates soluble guanylate cyclase (an enzyme containing heme iron) resulting in the production of HbF.[37] Some of the clinical effects are mediated by nitric oxide-induced vasodilatation or reduced platelet activation.

The Use of HU in Sickle Cell Disease placebo.

HU was initially tested in anemic baboons.[38] The first patients were tested in 1984 showing a response within 72 hours after therapy with an elevated level of HbF.[23] Subsequent prospective studies confirmed the efficacy and tolerability of HU in this setting. Recent reviews of the literature on HU therapy in sickle cell disease showed that HU was consistently associated with overall increases in HbF, a reduction of vaso-occlusive crises, decreased rates of hospitalization, and prevention of pulmonary complications.[39,40] The benefit of HU regarding the frequency of acute clinical events was demonstrated in randomized studies (Table 1),[28,41-47] but also in observational studies (uncontrolled longitudinal studies, retrospective case series, or prospective cohort studies using historical controls) (Table 2).[48-64]

Table 1 Table 1.  Randomized trials comparing HU with placebo.

Table 3 Table 2. Observational studies addressing acute clinical events with HU in sickle cell anemia.

Treatment with HU in adults: Most studies included both children and adults. Among specific studies for adult patients, the most important was a multicenter, double-blind, randomized controlled study that ran from 1992 to 1994 and that was stopped early after inclusion of 299 patients, because of a significant reduction of events in the Hu arm.[28] HU improved the clinical course of sickle cell disease by significantly reducing the annual rate of crises, increasing the median time to the first and second crisis, reducing the incidence of acute chest syndrome, and reducing transfusion requirements. Furthermore, the recommended dose of HU was not always needed to achieve a clinical response. Among the other randomized studies, no difference was noted in terms of frequency of vaso-occlusive crises in three studies.[42,44,45] In one study, there were equivalent liver iron contents and similar rates of stroke in both arms.[42] However, two of these three trials terminated earlier due to poor accrual. In the third study (SWiTCH trial), given the low rates of acute chest syndrome observed in the trial, the number of patients was not sufficient to determine whether there was a true difference between acute chest syndrome in the two arms.[45] Although studies of various designs showed that HU decreased the occurrence of acute chest disease, most studies provided lower-quality evidence for such effect.[49,58,62-64] Regarding pulmonary hypertension and tricuspid regurgitant velocity, the lack of randomization and prospective follow-up makes interpretation of results difficult. If most studies showed no difference among groups[26,65-70] or even higher proportion of patients with prior exposure to HU in a group with increased tricuspid regurgitant velocity,[71] some studies tended to provide evidence of HU effect.[72,73] Evidence for primary stroke prevention was limited to observational data.[50,58] While current evidence supports the use of chronic blood transfusions to prevent progressive disease and especially clinical stroke, HU represents an attractive alternative treatment option in order to avoid indefinite blood transfusion therapy which can lead to serious complications such as infections, iron overload, transfusion reactions, and erythrocyte allo- and auto-antibody formation.[74] The therapeutic switch from transfusions to HU should follow an overlap period of dual therapy because the benefits of HU have a slow onset and treatment should reach a stable maximum tolerated dose. After the switch, the problem of hemosiderosis persists. Despite effective oral chelators, the greatest challenge of serial phlebotomy in patients with sickle cell anemia is the underlying anemia, but HU therapy at a stable maximum tolerated dose typically raises the hemoglobin concentration, allowing a safe procedure.[75] Before the era of HU, the average life expectancy was in the 40s.[27] HU was associated with decreased mortality in symptomatic patients compared with those receiving only short-term HU or no HU.[57] It typically takes less than 6 months for patients to be stabilized on a dose that defines their maximal tolerated dose. Before the maximal tolerated dose is established, the number of cells with high HbF levels increases.[44,50,53,56] At 6 months, the HbF level is typically doubled, the hemoglobin level is increased, and the absolute reticulocyte count, bilirubin level, and lactate dehydrogenase level are reduced.[32] Patients should receive HU therapy as a continuous treatment unless adverse events occur. The optimal dose is still a source of debate. Dose escalation has been suggested toward the maximum tolerated dose (MTD). However, the stepwise approach of dose escalation generally requires several months and patients receiving HU have variable pharmacokinetics and pharmacodynamics.[76] Creatinine, reticulocyte count, and body mass index are among the simplest parameters that best predicted the HU maximum tolerated dose. It has been demonstrated a near linear dose response to HU. The treatment dose correlated positively with both the plasma drug concentration and the percentage of HbF response.[77] However, the dose does not need to be titrated to a particular HbF threshold. The dose can be escalated simply to reach an acceptable nontoxic degree of marrow suppression with target counts for both neutrophils and reticulocytes.[78] It has been suggested that HU may have benefits for the less common genotypes, especially HbSC or HbS/β+ thalassemia.[79] Because the primary effect of HU is damaging DNA replication by inhibiting ribonucleotide reductase, concerns have been raised about an oncogenic potential, especially after prolonged use. Although fears have been amplified by its original use as chemotherapy for chronic myeloproliferative diseases, which could evolve to acute leukemia, oncogenicity of HU is probably quite low or non-existent. Only a few cases of acute leukemia have been reported, but do not appear more frequent than in the untreated population.[80] Similarly, the benefits and harms of HU therapy in women with sickle cell disease during pregnancy and lactation represent a relevant issue.[79] No clear teratogenic phenotype exists for HU, but more data should be collected. Women with sickle cell anemia receiving HU have had successful normal pregnancies.[81] A variety of factors can lead to treatment failure. Poor adherence is recognized as a common problem and seems in part related to adverse events of the drug and inconvenience associated with monitoring.
Treatment with HU in children: The use of HU in children brings theoretically the best satisfactions regarding prevention of end-organ damage. However, it also carries potential risks in terms of growth and development and remains questionable for the risk of secondary malignancy after exposure to the drug for long periods. Observational studies in children have noted significant improvements in splenic uptake, glomerular filtration rate, renal hypertrophy, the ability to concentrate urine, microalbuminuria, and retinopathy.[82-86] As in adults studies, beneficial results with HU were reported showing a reduction in hospital admissions and days spent in the hospital, and potentially a reduced frequency of acute chest syndrome.[62,87-89] Evidence on benefits of HU use in children below 5 years is that it is associated with decreased pain crises and dactylitis.[41,90] However, most studies provided lower-quality evidence for the occurrence of acute chest syndrome.[46,47,60,61] There were few pieces of evidence that HU prevents stroke or the recurrence of stroke in children.[91] As in adults, evidence for primary stroke prevention was limited to observational data.[51] HU treatment was shown to lower secondary stroke rates in children with previous stroke.[92] Transcranial Doppler screening is used for primary stroke prevention. Abnormal velocities are the most common indication to commence chronic transfusion therapy in children. HU can lower transcranial Doppler velocities[93] and its utility in this setting is under investigations.[94] Although case reports have shown a reversal of splenic dysfunction after HU therapy, larger studies demonstrated that HU is clearly not enough to completely prevent the major complications of the disease.[95] Growth and development appeared to be unaffected in all studies.[95,96] HU should, therefore, be offered early and routinely as a preventive treatment for sickle cell anemia in children.

Distribution of HbF

More than 75% of the hemoglobin of the newborn is HbF. It diminishes over a period of several months to adult levels. HbF is becoming <2% by one year-of-age and <1% by 2 years. In most patients with sickle cell disease, HbF levels are increased. HbF is produced by a small number of erythroid precursors: the F-cells. Both HbF concentration and its distribution among erythrocytes are heritable. A correlation has been demonstrated between the number of F-cells and the percent of HbF in the hemolysate.[97] The concentration of HbF in each F-cell (HbF/F-cell) is changing during maturation.[98] However, quantitative methods for measuring this amount and plotting the distribution of HbF among F-cells are not available. The distribution of HbF concentrations among F-cells is the most critical element in the physiopathology of sickle cell anemia.[99] Compound heterozygosity for HbS and gene deletion hereditary persistence of HbF (HPFH) represents a condition in which the typical HbF level is 30%.[100] In this setting, HbF is distributed among all erythrocytes, each cell containing about 10 pg of HbF. Patients with sickle cell anemia have individually characteristic distributions of HbF/F-cell regardless of their total HbF level.[101] HU can induce HbF in most patients, but the HbF response to HU is highly variable.[102] Higher HbF levels were associated with a reduced rate of painful episodes, fewer leg ulcers, less osteonecrosis, and less frequent acute chest syndromes. However, HbF level had a weak or no clear association with priapism, urine albumin excretion, stroke and silent cerebral infarction, systemic blood pressure, and tricuspid regurgitant velocity.[103] HbF is  unevenly  distributed when high levels are successfully induced with HU. Total HbF and F-cell percentages are generally not good predictors of disease severity since they provide no information on F-cells with sufficient levels of HbF to protect against polymer-induced damage. Very few protected F-cells are present when HbF levels are about 5%, but more cells are possible when they reach 10%.[99] The calculated mean HbF/F-cell in HU-treated sickle cell anemia is about 8 pg. Early starting treatment seems to retard the fall in HbF, but many F-cells will continue to be poorly protected from polymer-induced damage even with 20% HbF.[99]

A Clinical and Economic Problem

It is estimated that 7% of the world population are carriers for hemoglobin disorders. Sickle cell disease is the most important potentially devastating, recessively inherited condition. The β-globin gene point mutation resulting in HbS has undergone evolutionary selection in the world because of its overwhelming malaria protective effects. High prevalence areas include Africa, the Middle-East, and Indian subcontinent with sickle cell trait affecting up to 300 million individuals worldwide.[104] In Africa, one on 14 persons with sickle cell anemia is an asymptomatic carrier.[32] One in 700 newborns is affected.[105] However, recent studies suggest that only 16% of polled individuals are aware of their sickle cell trait status, and only 37% of parents report having received notification of the sickle cell trait status of their children.[106,107] Sickle cell disease represents then an emerging global health burden in limited-resource countries, in which the development of sickle cell disease strategies should include sickle cell awareness, early detection, and the development of care and treatment programs.[108-110] The main recommendation is to educate all patients and their families about HU therapy. Although Food and Drug administration-label recommends treatment only for adults with sickle cell anemia severely affected with at least 3 painful crises over the prior 12 months, there are strong recommendations to treat adults with common clinical symptoms and to offer HU to children after age 9 months, regardless of clinical symptoms.[111] HU is relatively cheap. Especially in limited-resource countries without a safe and adequate blood supply, HU may represent a clinically useful and cost-effective therapeutic strategy for preventing cerebrovascular disease.[112] In the United States, it was reported per year 113,000 hospitalizations for sickle cell disease generating total hospital costs of about $488 million.[113] The average cost of HU was estimated at about $1,000 per year plus $400 per year for visits and tests.[28] This cost was offset by reduced costs for hospitalization, emergency room visits, and transfusions. The net savings was estimated at about $5,000 per patient per year.[114]

Beyond HU Therapy

HU is currently the only US Food and Drug Administration-approved medication to modify the disease course in sickle cell disease. However, elucidation of the multiple pathophysiologic mechanisms leading to vaso-occlusion and tissue injury in sickle cell disease is currently resulting in the identification of new treatment modalities.[115] In addition to HU, a number of drugs have been proposed: histone deacetylase inhibitors,[116] decitabine,[117] thalidomide and related compounds,[118] pomalidomide.[119] Optimally efficient induction of HbF may require combined use of drugs.[120] Carbon monoxide is also a potent antisickling agent that attaches to Hb and therefore reverse HbS polymerization. Shifting the oxyhemoglobin to the left or preventing cell dehydration can ameliorate sickling.[121] Sanguinate is a bovine pegylated Hb product designed to reduce sickling by delivering carbon monoxide to HbS and then carrying O2.[122] Because adhesive cell interactions contribute to vaso-occlusion, drugs targeting either red blood cell or leukocyte adhesion appear as attractive therapeutic modalities. Drugs targeting selectin-mediated adhesion are being especially investigated including the selectin inhibitors GMI-1070 (rivipansel)[123] and the humanized monoclonal antibody SelG1.[124] Heparin derivatives, such as sevuparin or tinzaparin, also have a well-known ability to inhibit adhesive interactions via P-selectin.[125,126] Targeting signaling pathways that activate adhesion molecules is another potential therapeutic modality. This can be achieved via beta-blockers administration[127] or through the use of MEK inhibitors[128] that might reduce red blood cell adhesion. Poloxamer 188, a nonspecific inhibitor of adhesion is also currently being studied.[129] Vaso-occlusion can engender an inflammatory response typical of hypoxia/reperfusion injury. Down-regulation of inflammatory pathways can, therefore, represent another approach to ameliorate sickle cell disease. Invariant NKT cells are involved in this pathogenesis. Their activation can be down-regulated by activation of the adenosine A2A receptor. Regadenoson is a partially selective adenosine A2A receptor agonist. It has been proposed in the treatment of vaso-occlusive crisis, which involves invariant NKT cells as contributors to the inflammatory component.[130] A humanized monoclonal antibody against invariant NKT cells has also recently shown some efficacy.[131] Because inflammatory pathways are important to both vaso-occlusion and tissue injury, targeting inflammatory mediators, such as leukotrienes, has also been proposed as a promising approach for the development of novel therapies in sickle cell disease.[132] Intravenous γ globulin infusion can also reduce inflammation via inhibition of neutrophil adhesion.[133] Statins that decrease endothelial inflammation have also been studied in sickle cell disease.[134] Drugs that increase HbF levels are the archetypal antisickling agents, because HbF interferes with polymerization of HbS, thereby lessening hemolytic rate and resulting in total Hb levels seen with HU therapy. The interference lengthens the delay time, allowing cells to avoid getting stuck in the microvasculature, even if hemolysis does not happen. Despite promising results, high mortality rates in patients older than 16 years and a paucity of suitable HLA-identical donors have limited the implementation of allogeneic stem cell transplantation in this patient population.[135] In the future, correction of the β-globin gene may be the ideal approach to curing sickle cell disease. However, there are still many concerns regarding this approach.
Despite the development of these many new treatment modalities and the promising results of the initial trials, HU remains a well-tolerated, safe, cheap, and efficacious for most patients with sickle cell disease, and should currently be considered standard-of-care for this disease.

Conclusions

HU is a remarkably effective drug for a large proportion of patients with sickle cell disease and appears to be the best currently available treatment option in this setting. Treatment is indicated for patients with “frequent pain episodes, history of acute chest syndrome, other severe vaso-occlusive events, or severe symptomatic anemia”.[32] Treatment endpoints remain “less pain and improved well-being, increased HbF to 15%-20%, increased hemoglobin level, and acceptable myelotoxicity”.[32] However, studies regarding a better understanding of HU effects, the ability to predict individual response, and the clinical applications for modifying disease effects are still ongoing. Two decades after the approval of HU, most patients with sickle cell disease are suboptimally treated with it, or not treated at all since this disease has continued to be treated with analgesics for pain relief. HU remains underutilized for a variety of reasons. It is likely that optimal therapy will only be achieved with a multi-targeted approach. However, any of the new therapies may be similarly underused, which may be the most difficult problem. HU is currently prescribed only sparingly and therefore has only limited effectiveness. Early initiation and broader use of HU should alter the natural history of sickle cell anemia. HU should be extended to low-resource settings, where the burden of the disease and the need for such a drug is the greatest.

References   

  1. Dresler WFC, Stein R. Über den hydroxylharnstoff. Justus Liebigs Ann Chem 1869; 150:242-52. https://doi.org/10.1002/jlac.18691500212  
  2. Rosenthal F, Wislicki L, Koller L. Über die beziehungen von schwersten blutgiften zu abbauprodukten des eiweisses: ein beitrag zum entstehungmechanismus der pernizosen anemie. KlinWochenschr 1928; 7:972-7. https://doi.org/10.1007/BF01716922 .   
  3. Stearns B, Losee KA, Bernstein J. Hydroxyurea: a new type of potential antitumor agent. J Med Chem 1963; 6:201. https://doi.org/10.1021/jm00338a026 PMid:14188794    
  4. Fishbein WN, Carbone PP, Freireich EJ, et al. Clinical trials of hydroxyurea in patients with cancer and leukemia.Clin Pharmacol Ther 1964; 5:574-80. https://doi.org/10.1002/cpt196455574  
  5. Becloff GL. Pharmacological, metabolic and clinical experience with hydroxyurea. Clin Trials J 1967; 4:873-83.  
  6. Kennedy BJ. Hydroxyurea therapy in chronic myelogenous leukemia.Cancer 1972; 29:1052-6. https://doi.org/10.1002/1097-0142(197204)29:4<1052::AID-CNCR2820290454>3.0.CO;2-7  
  7. Tefferi A, Pardanani A. Myeloproliferative neoplasms. A contemporary review. JAMA Oncol 2015; 1:97-105. https://doi.org/10.1001/jamaoncol.2015.89 PMid:26182311    
  8. Cortelazzo S, Finazzi G, Specchia G, et al. Hydroxyurea for patients with essential thrombocythemia and a high risk of thrombosis. N Engl J Med 1995; 332:1132-6. https://doi.org/10.1056/NEJM199504273321704 PMid:7700286    
  9. Fruchtman SM, Mack K, Kaplan ME, et al. From efficacy to safety: a Polycytemia Vera Study Group report on hydroxyurea in patients with polycytemia vera. Semin Hematol 1997; 34:17-23. PMid:9025158    
  10. Tefferi A. Primary myelofibrosis:2014 update on diagnosis, risk-stratification, and management. Am J Hematol 2014; 89:915-25. https://doi.org/10.1002/ajh.23703 PMid:25124313    
  11. Leavell UW, Yarbro JW. Hydroxyurea. A new treatment for psoriasis. Arch Dermatol 1970; 102:144-50. https://doi.org/10.1001/archderm.1970.04000080016003  
  12. Herrick JB. Peculiar elongated and sickle-shaped red blood corpuscules in a case of severe anemia. Arch Intern Med 1910; 6:517-21. https://doi.org/10.1001/archinte.1910.00050330050003  
  13. Pauling L, Itano HA.Sickle cell anemia, a molecular disease. Science 1949; 109:443. https://doi.org/10.1126/science.110.2865.543  
  14. Allison AC.The distribution of the sickle-cell trait in East Africa and elsewhere, and its apparent relationship to the incidence of subtertianmalaria.Trans R SocTrop Med Hyg 1954; 48:312-8. https://doi.org/10.1016/0035-9203(54)90101-7  
  15. Taylor SM, Parobeck CM, Fairhurst RM. Haemoglobinopathies and the clinical epidemiology of malaria: a systematic review and meta-analysis. Lancet Infect Dis 2012; 12:457-68. https://doi.org/10.1016/S1473-3099(12)70055-5  
  16. Ingram VM. A specific chemical difference between the globins of normal human and sickle-cell anaemiahaemoglobin.Nature 1956; 178:792-4. https://doi.org/10.1038/178792a0 PMid:13369537   
  17. Schmidt RM, Brosious EM. Evaluation of proficiency in the performance of tests for abnormal hemoglobins.Am J ClinPathol 1974;62:664-9. https://doi.org/10.1093/ajcp/62.5.664 PMid:4413315    
  18. Scott RB. Health care priority and sickle cell anemia. JAMA 1970; 214:731-4. https://doi.org/10.1001/jama.1970.03180040039008 PMid:5536114    
  19. Benson JM, Therrell BL Jr. History and current status of newborn screening for hemoglobinopathies. SeminPerinatol 2010; 34: 134-44. https://doi.org/10.1053/j.semperi.2009.12.006 PMid:20207263   
  20. Quinn CT, Rogers ZR, McCavit TL, Buchanan GR. Improved survival of children and adolescents with sickle cell disease. Blood 2010; 115:3447-52. https://doi.org/10.1182/blood-2009-07-233700 PMid:20194891 PMCid:PMC2867259  
  21. Naik RP, Derebail VK, Grams ME, et al. Association of sickle cell trait with chronic kidney disease and albuminuria in African Americans. JAMA 2014; 312:2115-25. https://doi.org/10.1001/jama.2014.15063 PMid:25393378 PMCid:PMC4356116  
  22. Auer PL, Johnsen JM, Johnson AD, et al. Imputation of exome sequence variants into population-based samples and blood-cell-trait associated loci in African Americans: NHLBI GO Exome Sequencing Project. Am J Hum Genet 2012; 91:794-808. https://doi.org/10.1016/j.ajhg.2012.08.031 PMid:23103231 PMCid:PMC3487117  
  23. Platt OS, Orkin SH, Dover G, et al. Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia. J Clin Invest 1984; 74 :652-6. https://doi.org/10.1172/JCI111464 PMid:6205021 PMCid:PMC370519  
  24. Platt OS, Thorington BD, Brambilla DJ, et al. Pain in sickle cell disease: rates and risk factors. N Engl J Med 1991; 325:11-6. https://doi.org/10.1056/NEJM199107043250103 PMid:1710777  
  25. Castro O, Brambilla DJ, Thorington BD, et al. The acute chest syndrome in sickle cell disease: incidence and risk factors.The Cooperative Study of Sickle Cell Disease.Blood 1994; 84:643-9. PMid:7517723    
  26. Gladwin MT, Vichinsky E. Pulmonary complications of sickle cell disease. N Engl J Med 2008; 359:2254-65. https://doi.org/10.1056/NEJMra0804411 PMid:19020327    
  27. Platt OS, Brambilla DJ, Rosse WF, et al. Mortality in sickle cell disease: life expectancy and risk factors for early death. N Engl J Med 1994; 330:1639-44. https://doi.org/10.1056/NEJM199406093302303 PMid:7993409    
  28. Charache S, Terrin ML, Moore RD, et al. Effects of hydroxyurea on the frequency of painful crises in sickle cell anemia. N Engl J Med 1995; 332:1317-22. https://doi.org/10.1056/NEJM199505183322001 PMid:7715639    
  29. Segal JB, Strouse JJ, Beach MC, et al. Hydroxyurea for the treatment of sickle cell disease. Evid Rep Technol Assess 2008; 165:1-95.  
  30. Brawley OW, Cornelius LJ, Edwards LR, et al. National Institutes of Health Consensus Development Conference statement: hydroxyurea treatment for sickle cell disease. Ann Intern Med 2008; 148:932-8. https://doi.org/10.7326/0003-4819-148-12-200806170-00220 PMid:18458271    
  31. Franco RS, Yasin Z, Palascak MB, et al. The effect of fetal hemoglobin on the survival characteristics of sickle cells.Blood 2006; 108:1073-6. https://doi.org/10.1182/blood-2005-09-008318 PMid:16861353 PMCid:PMC1895865  
  32. Platt OS.Hydroxyurea for the treatmet of sickle cell anemia. N Engl J Med 2008; 358:1362-9. https://doi.org/10.1056/NEJMct0708272 PMid:18367739    
  33. Halsey C, Roberts IAG. The role of hydroxyurea in sickle cell disease. Br J Haematol 2003; 120:177-86. https://doi.org/10.1046/j.1365-2141.2003.03849.x PMid:12542474    
  34. Kolata G. Globin gene studies create a puzzle. Science 1984; 223:470-1. https://doi.org/10.1126/science.6197757 PMid:6197757    
  35. Styles LA, Lubin B, Vichinsky E, et al. Decrease of very late activation antigen-4 and CD36 on reticulocytes in sickle cell patients treated with hydroxyurea. Blood 1997; 89:2554-9. PMid:9116302    
  36. Nogochi CT, Schechter AN, Rodgers GP. Sickle cell disease pathophysiology. Bailliere's Clinical Haematology 1993; 6:57-91. https://doi.org/10.1016/S0950-3536(05)80066-6  
  37. Cokic VP, Smith RD, Beleslin-Cokic BB, et al. Hydroxyurea induces fetal hemoglobin by the nitric oxide-dependent activation of soluble guanylylcyclase. J Clin Invest 2003; 111:231-9. https://doi.org/10.1172/JCI200316672 PMid:12531879 PMCid:PMC151872  
  38. Letvin NL, Linch DC, Beardsley GP, McIntyre KW, Nathan DG. Augmentation of fetal-hemoglobin production in anemic monkeys by hydroxyurea. N Engl J Med 1984; 310:869-73. https://doi.org/10.1056/NEJM198404053101401 PMid:6199670    
  39. Wong TE, Brandow AM, Lim W, Lottenberg R. Update on the use of hydroxyurea therapy in sickle cell disease. Blood 2014; 124:3850-7. https://doi.org/10.1182/blood-2014-08-435768 PMid:25287707 PMCid:PMC4271176  
  40. Buckner TW, Ataga KI. Does hydroxyurea prevent pulmonary complications of sickle cell disease? Hematology (Educational Sessions of the American Society of Hematology) 2014; 432-7. https://doi.org/10.1182/asheducation-2014.1.432 PMid:25696890    
  41. Wang WC, Ware RE, Miller ST, et al. Hydroxycarbamine in very young children with sickle-cell anaemia: a multicentre, randomized, controlled trial (BABY HUG). Lancet 2011; 377:1663-72. https://doi.org/10.1016/S0140-6736(11)60355-3  
  42. Ware RE, Helms RW. Stroke with transfusions changing to hydroxyurea (SWiTCH). Blood 2012; 119:3925-32. https://doi.org/10.1182/blood-2011-11-392340 PMid:22318199 PMCid:PMC3350359  
  43. Jain DL, Sarathi V, Desai S, et al. Low fixed-dose hydroxyurea in severely affected Indian children with sickle cell disease. Hemoglobin 2012; 36:323-32. https://doi.org/10.3109/03630269.2012.697948 PMid:22734586    
  44. Wang W, Brugnara C, Snyder C, et al. The effects of hydroxycarbamine and magnesium on haemoglobin SC disease: results of the multicentre CHAMPS trial. Br J Haematol 2011; 152:771-6. https://doi.org/10.1111/j.1365-2141.2010.08523.x PMid:21275961    
  45. Alvarez O, Yovetich NA, Scott JP, et al. Pain and other non-neurological adverse events in children with sickle cell anemia and previous stroke who received hydroxyurea and phlebotomy or chronic transfusions and chelation: results from the SWiTCH clinical trial. Am J Hematol 2013; 88:932-8. https://doi.org/10.1002/ajh.23547 PMid:23861242 PMCid:PMC4631259  
  46. Lebensburger JD, Miller ST, Howard TH, et al. Influence of severity of anemia on clinical findings in infants with sickle cell anemia: analyses from the BABY HUG study. Pediatr Blood Cancer 2012; 59:675-8. https://doi.org/10.1002/pbc.24037 PMid:22190441 PMCid:PMC3337342  
  47. Thornburg CD, Files BA, Luo Z, et al. Impact of hydroxyurea on clinical events in the BABY HUG trial. Blood 2012; 120:4304-10. https://doi.org/10.1182/blood-2012-03-419879 PMid:22915643 PMCid:PMC3507142  
  48. Ali SB, Moosang M, King L, et al. Stroke recurrence in children with sickle cell disease treated with hydroxyurea following first clinical stroke. Am J Hematol 2011; 86:846-50. https://doi.org/10.1002/ajh.22142 PMid:21898530    
  49. Gilmore A, Cho G, Howard J, et al. Feasibility and benefit of hydroxycarbamide as a long-term treatment for sickle cell disease patients: results from the North West London Sickle Cell Disease Registry. Am J Hematol 2011; 86:958-61. https://doi.org/10.1002/ajh.22146 PMid:21948113    
  50. Italia K, Jain D, Gattani S, et al. Hydroxyurea in sickle cell disease – a study of clinic-pharmacological efficacy in the Indian haplotype. Blood Cells Mol Dis 2009; 42:25-31. https://doi.org/10.1016/j.bcmd.2008.08.003 PMid:18954999    
  51. Lobo CL, Pinto JF, Nascimento EM, et al.The effect of hydroxycarbamine therapy on survival of children with sickle cell disease. Br J Haematol 2013; 161:852-60. https://doi.org/10.1111/bjh.12323 PMid:23590693    
  52. Nzouakou R, Bachir D, Lavaud A, et al. Clinical follow-up of hydroxyurea-treated adults with sickle cell disease. Acta Haematol 2011; 125:145-52. https://doi.org/10.1159/000322248  
  53. Patel DK, Mashon RS, Patel S, et al. Low dose hydroxyurea is effective in reducing the incidence of painful crisis and frequency of blood transfusion in sickle cell anemia patients from eastern India. Hemoglobin 2012; 36:409-20.  https://doi.org/10.3109/03630269.2012.709897 PMid:22881992    
  54. Rigano P, Pecoraro A, Calvaruso G, et al. Cerebrovascular events in sickle cell-beta thalassemia treated with hydroxyurea: a single center prospective survey in adult Italians. Am J Hematol 2013; 88:E261-4. https://doi.org/10.1002/ajh.23531 PMid:23828131    
  55. Sharef SW, Al-Hajri M, Beshlawi I, et al. Optimizing hydroxyurea use in children with sickle cell disease: low dose regimen is effective. Eur J Haematol 2013; 90:519-24. https://doi.org/10.1111/ejh.12103 PMid:23489171    
  56. Singh H, Dulhani N, Kumar BN, et al. Effective control of sickle cell disease with hydroxyurea therapy. Indian J Pharmacol 2010; 42:32-5. https://doi.org/10.4103/0253-7613.62409 PMid:20606834 PMCid:PMC2885637  
  57. Steinberg MH, McCarthy WF, Castro O, et al. The risks and benefits of long-term use of hydroxyurea in sickle cell anemia: a 17.5 year follow-up. Am J Hematol 2010; 85:403-8. https://doi.org/10.1002/ajh.21699  
  58. Voskaridou E, Christoulas D, Bilalis A, et al. The effect of prolonged administration of hydroxyurea on morbidity and mortality in adult patients with sickle cell syndromes: results of a 17-year, single-center trial (LaSHS). Blood 2010; 115:2354-63. https://doi.org/10.1182/blood-2009-05-221333 PMid:19903897    
  59. Gulbis B, Haberman D, Dufour D, et al. Hydroxyurea for sickle cell disease in children and for prevention of cerebrovascular events: the Belgian experience. Blood 2005; 105:2685-90. https://doi.org/10.1182/blood-2004-07-2704 PMid:15604217    
  60. Hankins JS, Ware RE, Rogers ZR, et al. Long-term hydroxyurea therapy for infants with sickle cell anemia: the HUSOFT extension study. Blood 2005; 106:2269-75. https://doi.org/10.1182/blood-2004-12-4973 PMid:16172253 PMCid:PMC1895275  
  61. Jain DL, Apte M, Colah R, et al. Efficacy of fixed low dose hydroxyurea in Indian children with sickle cell anemia: a single centre experience. Indian Pediatr 2013; 50:929-33. https://doi.org/10.1007/s13312-013-0264-0 PMid:23798623    
  62. Koren A, Segal-Kupershmit D, Zalman L, et al. Effect of hydroxyurea in sickle cell anemia: a clinical trial in children and teenagers with severe sickle cell beta-thalassemia. Pediatr Hematol Oncol 1999; 16:221-32. https://doi.org/10.1080/088800199277272 PMid:10326220    
  63. Olivieri NF, Vichinsky EP. Hydroxyurea in children with sickle cell disease: impact on splenic function and compliance with therapy. J Pediatr Hematol Oncol 1998; 20:26-31. https://doi.org/10.1097/00043426-199801000-00004  
  64. Silva-Pinto AC, Angulo IL, Brunetta DM, et al. Clinical and hematological effects of hydroxyurea therapy in sickle cell patients: a single-center experience in Brazil. Sao Paulo Med J 2013; 131:238-43. https://doi.org/10.1590/1516-3180.2013.1314467 PMid:24141294     
  65. Desai PC, May RC, Jones SK, et al. Longitudinal study of echocardiography-derived tricuspid regurgitant jet velocity in sickle cell disease. Br J Haematol 2013; 162:836-41. https://doi.org/10.1111/bjh.12453 PMid:23829561 PMCid:PMC3759564  
  66. Gordeuk VR, Campbell A, Rana S, et al. Relationship of erythropoietin, fetal hemoglobin, and hydroxyurea treatment to tricuspid regurgitation velocity in children with sickle cell disease. Blood 2009; 114:4639-44. https://doi.org/10.1182/blood-2009-04-218040 PMid:19724057 PMCid:PMC2780300  
  67. Pashankar FD, Carbonella J, Bazzy-Asaad A, et al. Prevalence and risk factors of elevated pulmonary artery pressures in children with sickle cell disease. Pediatrics 2008; 121:777-82. https://doi.org/10.1542/peds.2007-0730 PMid:18381543    
  68. Voskaridou E, Tsetsos G, Tsoutsias A, et al. Pulmonary hypertension in patients with sickle cell/beta thalassemia: incidence and correlation with serum N-terminal pro-brain natriuretic peptide concentrations. Haematologica 2007; 92:738-43. https://doi.org/10.3324/haematol.11136 PMid:17550845    
  69. Fonseca GH, Souza R, Salemi VM, et al. Pulmonary hypertension diagnosed by right heart catheterization in sickle cell disease. EurRespir J 2012; 39:112-8. https://doi.org/10.1183/09031936.00134410 PMid:21778170    
  70. Parent F, Bachir D, Inamo J, et al. A hemodynamic study of pulmonary hypertension in sickle cell disease. N Engl J Med 2011; 365:44-53. https://doi.org/10.1056/NEJMoa1005565 PMid:21732836    
  71. Dahoui HA, Hayek MN, Nietert PJ, et al. Pulmonary hypertension in children and young adults with sickle cell disease: evidence for familial clustering. Pediatr Blood Cancer 2010; 54:398-402. https://doi.org/10.1002/pbc.22306 PMid:19827138    
  72. Ataga KI, Moore CG, Jones S, et al. Pulmonary hypertension in patients with sickle cell disease: a longitudinal study. Br J Haematol 2006; 134:109-15. https://doi.org/10.1111/j.1365-2141.2006.06110.x PMid:16803576    
  73. De Castro LM, Jonassaint JC, Graham FL, et al. Pulmonary hypertension associated with sickle cell disease: clinical and laboratory endpoints and disease outcomes. Am J Hematol 2008; 83:19-25. https://doi.org/10.1002/ajh.21058 PMid:17724699    
  74. Chou ST, Jackson T, Vege S, et al. High prevalence of red blood cell alloimmunization in sickle cell disease despite transfusion from Rh-matched minority donors. Blood 2013; 122:1062-71. https://doi.org/10.1182/blood-2013-03-490623 PMid:23723452     
  75. Aygun B, Mortier NA, Kesler K, et al. Therapeutic phlebotomy is safe in children with sickle cell anaemia and can be effective treatment for transfusional iron overload. Br J Haematol 2015; 169:262-6. https://doi.org/10.1111/bjh.13280 PMid:25612463 PMCid:PMC4631316  
  76. Ware RE, Despotovic JM, Mortier NA, et al. Pharmacokinetics, pharmacodynamics, and pharmacogenetics of hydroxyurea treatment for children with sickle cell anemia. Blood 2011; 118:4985-91. https://doi.org/10.1182/blood-2011-07-364190 PMid:21876119 PMCid:PMC3208303  
  77. Charache S, Dover GJ, Moore RD, et al. Hydroxyurea. Effects on hemoglobin F production in patients with sickle cell anemia.Blood 1992; 79:2555-65. PMid:1375104    
  78. Ware RE. Optimizing hydroxyurea therapy for sickle cell anemia. Hematology (Educational Sessions of the American Society of Hematology) 2015; 436-43. https://doi.org/10.1182/asheducation-2015.1.436 PMid:26637755   
  79. Savage WJ, Buchanan GR, Yawn BP, et al. Evidence gaps in the management of sickle cell disease: a summary of needed research. Am J Hematol 2015; 90:273-5. https://doi.org/10.1002/ajh.23945 PMid:25639238    
  80. Flanagan JM, Howard TA, Mortier N, et al. Assessment of genotoxicity associated with hydroxyurea therapy in children with sickle cell anemia. Mutat Res 2010; 698:38-42. https://doi.org/10.1016/j.mrgentox.2010.03.001 PMid:20230905    
  81. Ballas SK, McCarthy WF, Guo N, et al. Exposure to hydroxyurea and pregnancy outcomes in patients with sickle cell anemia. J Natl Med Assoc 2009; 101:1046-51. https://doi.org/10.1016/S0027-9684(15)31072-5  
  82. Alvarez O, Miller ST, Wang WC, et al. Effect of hydroxyurea treatment on renal function parameters: results from the multicenter placebo-controlled BABY HUG clinical trial for infants with sickle cell anemia. Pediatr Blood Cancer 2012; 59:668-74. https://doi.org/10.1002/pbc.24100 PMid:22294512 PMCid:PMC3396762  
  83. Hankins JS, Helton KJ, McCarville MB, et al. Preservation of spleen and brain function in children with sickle cell anemia treated with hydroxyurea. Pediatr Blood Cancer 2008; 50:293-7. https://doi.org/10.1002/pbc.21271 PMid:17554794    
  84. Aygun B, Mortier NA, Smeltzer MP, et al. Hydroxyurea treatment decreases glomerular hyperfiltration in children with sickle cell anemia. Am J Hematol 2013; 88:116-9. https://doi.org/10.1002/ajh.23365 PMid:23255310 PMCid:PMC4673980  
  85. Estepp JH, Smeltzer MP, Wang WC, et al. Protection from sickle cell retinopathy is associated with elevated HbF levels and hydroxycarbamine use in children. Br J Haematol 2013; 161:402-5. https://doi.org/10.1111/bjh.12238 PMid:23384083    
  86. McKie KT, Hanevold CD, Hernandez C, et al. Prevalence, prevention, and treatment of microalbuminuria and proteinuria in children with sickle cell disease. J Pediatr Hematol Oncol 2007; 29:140-4. https://doi.org/10.1097/mph.0b013e3180335081  
  87. Jayabose S, Tugal O, Sandoval C, et al. Clinical and hematologic effects of hydroxyurea in children with sickle cell anemia. J Pediatr 1996; 129:559-65. https://doi.org/10.1016/S0022-3476(96)70121-X  
  88. Ferster A, Vermylen C, Cornu G, et al. Hydroxyurea for treatment of severe sickle cell anemia : a pediatric clinical trial. Blood 1996; 88:1960-4. PMid:8822914    
  89. Ferster A, Tahriri P, Vermylen C, et al. Five years of experience with hydroxyurea in children and young adults with sickle cell disease. Blood 2001; 97:3628-32. https://doi.org/10.1182/blood.V97.11.3628 PMid:11369660    
  90. Mulaku M, Opiyo N, Karumbi J, et al. Evidence review of hydroxyurea for the prevention of sickle cell complications in low-income countries. Arch Dis Child 2013; 98:908-14. https://doi.org/10.1136/archdischild-2012-302387 PMid:23995076 PMCid:PMC3812872  
  91. Ware RE, Zimmerman SA, Schultz WH. Hydroxyurea as an alternative to blood to blood transfusions for the prevention of recurrent stroke in children with sickle cell disease. Blood 1999; 94:3022-6. PMid:10556185    
  92. Ware RE, Zimmerman SA, Sylvestre PB, et al. Prevention of secondary stroke and resolution of transfusional iron overload in children with sickle cell anemia using hydroxyurea and phlebotomy. J Pediatr 2004; 145:346-52. https://doi.org/10.1016/j.jpeds.2004.04.058 PMid:15343189    
  93. Zimmerman SA, Schultz WH, Burgett S, et al. Hydroxyurea therapy lowers transcranial Doppler flow velocities in children with sickle cell anemia. Blood 2007; 110:1043-7. https://doi.org/10.1182/blood-2006-11-057893 PMid:17429008    
  94. Hankins JS, McCarville MB, Rankine-Mullings A, et al. Prevention of conversion to abnormal TCD with hydroxyurea in sickle cell anemia: A phase III international randomized clinical trial. Am J Hematol 2015; 90:1099-105. https://doi.org/10.1002/ajh.24198 PMid:26414435 PMCid:PMC4715740  
  95. Wang WC, Wynn LW, Rogers ZR, et al. A two-year pilot trial of hydroxyurea in very young children with sickle-cell anemia J Pediatr 2001; 139:790-6. https://doi.org/10.1067/mpd.2001.119590 PMid:11743503    
  96. Wang WC, Helms RW, Lynn HS, et al. Effects of hydroxyurea on growth in children with sickle cell anemia: Results of the HUG-KIDS study. J Pediatr 2002; 140:225-9. https://doi.org/10.1067/mpd.2002.121383 PMid:11865275    
  97. Mundee Y, Bigelow NC, Davis BH, Porter JB. Flow cytometric method for simultaneous assay of foetal haemoglobin containing red cells, reticulocytes and foetal haemoglobin containing reticulocytes. Clin Lab Haematol 2001; 23:149-54. https://doi.org/10.1046/j.1365-2257.2001.00344.x PMid:11553054    
  98. Dover GJ, Boyer SH. Quantitation of hemoglobins within individual red cells: asynchronous biosynthesis of fetal and adult hemoglobin during erythroid maturation in normal subjects. Blood 1980; 56:1082-91. PMid:6159933    
  99. Steinberg MH, Chui DHK, Dover GJ, et al. Fetal hemoglobin in sickle cell anemia: a glass half full? Blood 2014; 123:481-5. https://doi.org/10.1182/blood-2013-09-528067 PMid:24222332    
  100. Ngo DA, Aygun B, Akinsheye I, et al. Fetal haemoglobin levels and haematological characteristics of compound heterozygotes for haemoglobin S and deletional hereditary persistence of fetal haemoglobin. Br J Haematol 2012; 156:259-64. https://doi.org/10.1111/j.1365-2141.2011.08916.x PMid:22017641    
  101. Horiuchi K, Osterhout ML, Kamma H, et al. Estimation of fetal hemoglobin levels in individual red cells via fluorescence image cytometry. Cytometry 1995; 20:261-7. https://doi.org/10.1002/cyto.990200310 PMid:7587712    
  102. Steinberg MH, Lu ZH, Barton FB, et al. Multicenter study of hydroxyurea. Fetal hemoglobin in sickle cell anemia: determinants of response to hydroxyurea. Blood 1997; 89:1078-88. PMid:9028341    
  103. Steinberg MH, Sebastiani P. Genetic modifiers of sickle cell disease. Am J Hematol 2012; 87:795-803. https://doi.org/10.1002/ajh.23232 PMid:22641398 PMCid:PMC4562292  
  104. Elion J, Berg PE, Lapoumeroulie C, et al. DNA sequencevariation in a negative control region 5' to the beta-globin gene correlates with the phenotypic expression of the beta S mutation. Blood 1992; 79:787-92. PMid:1346253    
  105. Lorey FW, Arnopp J, Cunningham GC. Distribution of hemoglobinopathy variants by ethnicity in a multiethnic state.Genet Epidemiol 1996; 13:501-12. https://doi.org/10.1002/(SICI)1098-2272(1996)13:5<501::AID-GEPI6>3.0.CO;2-4  
  106. Treadwell MJ, McClough L, Vichinsky E. Using qualitative and quantitative strategies to evaluate knowledge and perceptions about sickle cell disease and sickle cell trait. J Natl Med Assoc 2006; 98:704-10. PMid:16749645 PMCid:PMC2569269  
  107. Kavanagh PL, Wang CJ, Therrell BL, et al. Communication of positive newborn screening results for sickle cell disease and sickle cell trait: variation across states. Am J Med Genet C Semin Med Genet 2008; 148C:15-22. https://doi.org/10.1002/ajmg.c.30160 PMid:18200513    
  108. Weatherall DJ. The inherited diseases of hemoglobin are an emerging global health burden. Blood 2010; 115:4331-6. https://doi.org/10.1182/blood-2010-01-251348 PMid:20233970 PMCid:PMC2881491  
  109. Piel FB, Hay SI, Gupta S, et al. Global burden of sickle cell anaemia in children under five, 2010-2050: modeling based on demographics, excess mortality, and interventions. PLoS Med 2013; 10:e1001484. https://doi.org/10.1371/journal.pmed.1001484 PMid:23874164 PMCid:PMC3712914  
  110. World Health Organization. Sickle-cell disease: a strategy for the WHO African Region. Report AFR/RC60/8. Geneva, Switzerland: World Health Organization; 2010.  
  111. Yawn BP, Buchanan GR, Afenyi-Annan AN, et al. Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. JAMA 2014; 12:1033-48. https://doi.org/10.1001/jama.2014.10517 PMid:25203083   
  112. Cunningham-Myrie C, Abdulkadri A, Waugh A, et al. Hydroxyurea use in prevention of stroke recurrence in children with sickle cell disease in a developing country: a cost effectiveness analysis. Pediatr Blood Cancer 2015; 62:1862-4. https://doi.org/10.1002/pbc.25563 PMid:25929458    
  113. Steiner CA, Miller JL. Sickle cell disease patients in U.S. hospitals, 2004.Statistical brief. No.21. Rockville, Md: Agency for Healthcare Research and Quality, December 2006.  
  114. Moore RD, Charache S, Terrin ML, et al. Cost-effectiveness of hydroxyurea in sickle cell anemia. Am J Hematol 2000; 64:26-31. https://doi.org/10.1002/(SICI)1096-8652(200005)64:1<26::AID-AJH5>3.0.CO;2-F  
  115. Telen MJ. Beyond hydroxyurea: new and old drugs in the pipeline for sickle cell disease. Blood 2016; 127:810-9. https://doi.org/10.1182/blood-2015-09-618553 PMid:26758919    
  116. Atweh GF, Sutton M, Nassif I, et al. Sustained induction of fetal hemoglobin by pulse butyrate therapy in sickle cell disease. Blood 1999; 93:1790-7. PMid:10068649 PMCid:PMC4269326  
  117. Saunthararajah Y, Molokie R, Saraf S, et al. Clinical effectiveness of decitabine in severe sickle cell disease. Br J Haematol 2008; 141:126-9. https://doi.org/10.1111/j.1365-2141.2008.07027.x PMid:18324975    
  118. Dos Santos JL, Lanaro C, Lima LM, et al. Design, synthesis, and pharmacological evaluation of novel hydrid compounds to treat sickle cell disease symptoms. J Med Chem 2011; 54:5811-9. https://doi.org/10.1021/jm200531f PMid:21766854    
  119. Meiler SE, Wade M, Kutlar F, et al. Pomalidomide augments fetal hemoglobin production without the myelosuppressive effects of hydroxyurea in transgenic sickle cell mice. Blood 2011; 118:248-58. https://doi.org/10.1182/blood-2010-11-319137 PMid:21536862 PMCid:PMC3148160  
  120. Fard AD, Hosseini SA, Shahjahani M, et al. Evaluation of novel fetal hemoglobin inducer drugs in treatment of ß-hemoglobinopathy disorders. Int J Hematol Oncol Stem Cell Res 2013; 7:47-54. PMid:24505535 PMCid:PMC3913144  
  121. Ataga KI, Stocker J. Senicapoc (ICA-17043): a potential therapy for the prevention and treatment of hemolysis-associated complications in sickle cell anemia. Expert Opin Investig Drugs 2009; 18:231-9. https://doi.org/10.1517/13543780802708011 PMid:19236269    
  122. Misra H, Lickliter J, Kazo F, Abuchowski A. PEGylated carboxyhemoglobin bovine (SANGUINATE): result of a phase I clinical trial. Artif Organs 2014; 38:702-7. https://doi.org/10.1111/aor.12341 PMid:25113835    
  123. Chang J, Patton JT, Sarkar A, et al. GMI-1070, a novel pan-selectin antagonist, reverses acute vascular occlusions in sickle cell mice. Blood 2010; 116:1779-86. https://doi.org/10.1182/blood-2009-12-260513 PMid:20508165 PMCid:PMC2947397  
  124. Mandarino D, Kawar Z, Alvarez R, et al. Placebo-controlled, double-blind, first-in-human, ascending single dose, healthy subject study of intravenous-administered SelG1, a humanized anti-P-selectin antibody in development for sickle cell disease. Blood 2013; 122:abstract 970.  
  125. Batchvarova M, Shan S, Zennadi R, et al. Sevuparin reduces adhesion of both sickle red cells and leukocytes to endothelial cells in vitro and inhibits vaso-occlusion in vivo. Blood 2013;122:abstract 182.  
  126. Qari MH, Aljaouni SK, Alardawi MS, et al. Reduction of painful vaso-occlusive crisis of sickle cell anaemia by tinzaparin in a double-blind randomized trial. Thromb Haemost 2007; 98:392-6. https://doi.org/10.1160/th06-12-0718  
  127. De Castro LM, Zennadi R, Jonassaint JC, et al. Effect of propranolol as anti-adhesive therapy in sickle cell disease. Clin Transl Sci 2012; 5:437-44. https://doi.org/10.1111/cts.12005 PMid:23253664 PMCid:PMC3762678  
  128. Zennadi R. MEK inhibitors, novel anti-adhesive molecules, reduce sickle red blood cell adhesion in vitro and in vivo, and vasoocclusion in vivo. PLoS One 2014; 9:e110306. .  
  129. Cheung AT, Chan MS, Ramanujam S, et al. Effects of poloxamer 188 treatment on sickle cell vaso-occlusive crisis: computer-assisted intravital microscopy study. J Investig Med 2004; 52:402-6. https://doi.org/10.1136/jim-52-06-35 PMid:15612454    
  130. Field JJ, Lin G, Okam MM, et al. Sickle cell vaso-occlusion causes activation of iNKT cells that is decreased by the adenosine A2A receptor agonist ragadenoson. Blood 2013; 121:3329-34. https://doi.org/10.1182/blood-2012-11-465963 PMid:23377438 PMCid:PMC3637009  
  131. Field JJ, Ataga KI, Majerus E, et al. A phase I single ascending dose study of NKTT120 in stable adult sickle cell patients. Blood 2013; 122:abstract 977.  
  132. Knight-Perry J, DeBaun MR, Strunk RC, Field JJ. Leukotriene pathway in sickle cell disease: a potential target for directed therapy. Expert Rev Hematol 2009; 2:57-68. https://doi.org/10.1586/17474086.2.1.57 PMid:21082995    
  133. Chang J, Shi PA, Chiang EY, Frenette PS. Intravenous immunoglobulins reverse acute vaso-occlusive crises in sickle cell mice through rapid inhibition of neutrophil adhesion. Blood 2008; 111:915-23. https://doi.org/10.1182/blood-2007-04-084061 PMid:17932253 PMCid:PMC2200843  
  134. Hoppe C, Kuypers F, Larkin S, et al. A pilot study of the short-term use of simvastatin in sickle cell disease: effects on markers of vascular dysfunction. Br J Haematol 2011; 153:655-63. https://doi.org/10.1111/j.1365-2141.2010.08480.x PMid:21477202 PMCid:PMC3601917  
  135. Bhatia M, Walters MC. Hematopoietic cell transplantation for thalassemia and sickle cell disease: past, present and future. Bone Marrow Transplant 2008; 41:109-17. https://doi.org/10.1038/sj.bmt.1705943 PMid:18059330    

[TOP]