Your activity: 30 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Hydroxyurea use in sickle cell disease

Hydroxyurea use in sickle cell disease
Authors:
Griffin P Rodgers, MD
Alex George, MD, PhD
John Strouse, MD, PhD
Section Editor:
Michael R DeBaun, MD, MPH
Deputy Editor:
Jennifer S Tirnauer, MD
Literature review current through: Dec 2022. | This topic last updated: Mar 02, 2022.

INTRODUCTION — The major causes of morbidity and mortality in sickle cell disease (SCD) are the acute and long-term consequences of vaso-occlusion and hemolysis, many of which cannot be reversed (eg, tissue infarction, vasculopathy).

The approaches that are available for reducing these pathophysiologic processes are regular red blood cell (RBC) transfusions, medications (hydroxyurea, L-glutamine, voxelotor, and crizanlizumab) and stem cell therapies (hematopoietic cell transplantation [HCT] and gene therapy).

This topic review discusses hydroxyurea therapy in SCD, including the mechanism of action, administration, dosing, and adverse effects.

Separate topic reviews discuss the following subjects:

Evaluation of pain – (See "Evaluation of acute pain in sickle cell disease".)

Prevention of pain – (See "Disease-modifying therapies to prevent pain and other complications of sickle cell disease".)

Transplant – (See "Hematopoietic stem cell transplantation in sickle cell disease".)

Investigational therapies – (See "Investigational therapies for sickle cell disease".)

Overview of management – (See "Overview of the management and prognosis of sickle cell disease".)

Routine care for children – (See "Sickle cell disease in infancy and childhood: Routine health care maintenance and anticipatory guidance".)

Transition from pediatric to adult care – (See "Sickle cell disease (SCD) in adolescents and young adults (AYA): Transition from pediatric to adult care".)

RBC transfusions – (See "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques".)

The choice of preventive therapy in patients with specific complications such as acute chest syndrome (ACS) or stroke is also presented separately. (See "Acute chest syndrome (ACS) in sickle cell disease (adults and children)", section on 'Prevention' and "Prevention of stroke (initial or recurrent) in sickle cell disease".)

MECHANISM OF ACTION — Hydroxyurea has been in use for decades, first approved by the US Food and Drug Administration (FDA) in 1967 as an antineoplastic drug for the treatment of multiple cancers (melanoma, ovarian cancer, myeloproliferative neoplasms) and then for SCD in 1998. Despite this long experience, our understanding of the mechanism of action of hydroxyurea in SCD remains incomplete [1].

Inhibition of ribonucleotide reductase — Hydroxyurea can bind metals. The primary cellular target of hydroxyurea is the ribonucleotide reductase enzymes, iron-containing enzymes that convert ribonucleoside diphosphates to deoxyribonucleotide triphosphates (dNTPs), which are used in DNA synthesis and repair [2,3]. Ribonucleotide reductases use a free radical mechanism to catalyze the conversion to dNTPs, and hydroxyurea appears to block their function by binding the iron molecules and scavenging the free radicals [4]. The lack of available dNTPs causes cells to arrest in S-phase of the cell cycle. Eventually, cells with stalled DNA replication forks will either delay S-phase until DNA synthesis can proceed, or they will undergo cell death.

Hydroxyurea has been shown to suppress other enzymes such as iron-sulfur cluster-containing enzymes (see "Causes and pathophysiology of the sideroblastic anemias", section on 'Iron-sulfur (Fe-S) cluster biogenesis'), but it is not clear whether there is any physiologic effect in vivo.

Because hydroxyurea affects cell division, it causes global myelosuppression, including neutropenia, anemia, and thrombocytopenia. (See 'Myelosuppression' below.)

Increased Hb F production — Hydroxyurea causes a shift in gene expression at the beta globin locus, such that expression from the gamma globin locus is increased relative to that from the beta globin locus. This results in increased production of fetal hemoglobin (Hb F; α2γ2) and decreased production of adult hemoglobin (Hb A; α2β2), the reverse of the normal fetal switch. Since the gamma globin chain is not affected by the sickle mutation, the overall effect in patients with SCD is to reduce the relative concentration of hemoglobin S (Hb S; α2βS2).

Hb F is the form of hemoglobin normally produced during fetal development and early infancy. Hb F has other properties that differ slightly from normal adult hemoglobin (eg, slightly higher oxygen affinity), but it is well tolerated and causes no clinical problems. Hydroxyurea causes Hb F production to be increased at least twofold above baseline, often much more. The concentration of Hb F per cell, the proportion of Hb F-containing cells, and the overall percentage of Hb F are all increased. The resultant decrease in the relative intracellular concentration of Hb S leads to less hemoglobin polymerization and precipitation (see "Pathophysiology of sickle cell disease", section on 'Effects on the RBC'). These effects reduce the formation of sickled red blood cells (RBCs). RBC lifespan is increased; RBC hydration is improved, hemolysis is reduced, and adhesion of cells to the vascular endothelium is lessened [5-9]. In turn, blood flow through the microcirculation improves and vaso-occlusive events are less likely to occur. This is thought to be the major mechanism by which hydroxyurea reduces vaso-occlusive events. In a prospective observational study of 230 children with SCD who were treated with hydroxyurea, a Hb F percentage >20 percent was associated with much lower rates of hospitalization, vaso-occlusive pain, acute chest syndrome, and fever [10].

Additional evidence to support the role of increased Hb F in ameliorating SCD complications comes from observational studies of populations with naturally higher Hb F levels and mouse models [11-15]. The role of Hb F in these populations is discussed in more detail separately. (See "Fetal hemoglobin (hemoglobin F) in health and disease".)

The mechanism(s) by which hydroxyurea increases Hb F production are incompletely understood and may include effects on epigenetic modification, gene transcription, and cell signaling [16]. Increased nitric oxide (NO) levels and cyclic nucleotides such as guanylyl cyclase may induce Hb F transcription via genes that regulate fetal hemoglobin transcription and translation, including BCL11A, a key regulator of baseline Hb F levels [17-19]. Hydroxyurea may induce expression of the small GTP-binding protein SAR1 (secretion-associated and ras-related signaling protein), which activates gamma globin expression through c-Jun N-terminal kinase (JNK/Jun) [20,21]. Expression is increased from the G-gamma rather than the A-gamma gene [22]. These effects require a degree of active erythropoiesis, which may be suppressed by transfusion and possibly increased by erythropoietin. Alternative approaches to increasing Hb F production via these pathways are under investigation. (See "Investigational therapies for sickle cell disease".)

The changes in Hb F and Hb S production are first seen in reticulocytes. It may take up to three or more weeks to be seen in mature RBCs, and up to six months to affect clinical symptoms. The effects are completely reversible upon drug discontinuation, necessitating lifelong therapy in most cases. (See 'Can therapy be discontinued?' below.)

Other mechanisms — While reduced sickling due to increased Hb F levels is a major beneficial effect of hydroxyurea in SCD, other mechanisms may also play a role:

Nitric oxideHydroxyurea may increase nitric oxide (NO) levels by two mechanisms. Free hemoglobin, released during hemolysis, is a natural NO scavenger. Hydroxyurea reduces hemolysis and in turn may reduce free hemoglobin, allowing NO levels to be higher. Intracellularly, hydroxyurea leads to NO production in RBCs and endothelial cells via multiple mechanisms including direct NO generation by reacting with heme proteins [23-27]. NO is a potent vasodilator, and depletion in certain vascular beds could contribute to vaso-occlusion; increases in NO may improve blood flow in certain circulatory beds such as the pulmonary vasculature [17,23-26,28]. (See "Pulmonary hypertension associated with sickle cell disease", section on 'Pathogenesis'.)

RBC rheology – The lower reticulocyte count and corresponding shift to more mature RBCs may also improve RBC volume, density, adhesivity, and passage through the microcirculation independent of Hb F levels [9,29-32]. This may be due in part to a reduction in the proportion of reticulocytes and young, low-density RBCs, which are particularly likely to adhere to vascular endothelium [29,33]. Effects of hydroxyurea on cellular signaling pathways may also reduce adhesion [31]. (See "Pathophysiology of sickle cell disease", section on 'Effects on the RBC'.)

White blood cells – Reduced white blood cell (WBC) counts and/or neutrophil adhesivity to the vascular endothelium may also contribute to reduced vaso-occlusion [34]. In vitro studies have shown that neutrophils from individuals with SCD have enhanced binding to fibronectin and increased activation [35,36]. In a trial that randomly assigned individuals with SCD to hydroxyurea or placebo, there was a strong correlation between neutrophil count and pain episodes over two years of observation (lower neutrophil counts were associated with fewer pain episodes) [37]. In contrast, an inverse relationship between Hb F-containing cells and pain episodes was only seen during the first three months of the trial. Interpretation is difficult because the hydroxyurea dose was titrated to the neutrophil count. (See "Pathophysiology of sickle cell disease", section on 'Inflammation'.)

Predictors of response — The Hb F response to hydroxyurea is variable and complex; there is no specific feature that can be used to determine whether an individual treated with hydroxyurea will obtain a clinical benefit or the magnitude of the benefit. Even children with low baseline percent Hb F levels can develop substantial increases in Hb F when treated at maximally tolerated doses [38]. Thus, hydroxyurea is offered to all individuals who could potentially benefit from it. (See 'Indications and appropriate age to start therapy' below.)

Predictors of response to hydroxyurea were explored in a subset of children who participated in the HUG-KIDS study, in which hydroxyurea was administered to children between 5 and 15 years of age and titrated to a maximum tolerated dose [38] (see 'Monitoring and dose titration' below). For the 53 children who were treated at maximum tolerated dose for at least a year, the following characteristics predicted the greatest increase in percentage of Hb F, which is typically used as a surrogate for clinical efficacy:

Higher baseline percent Hb F

Higher baseline reticulocyte count

Higher baseline WBC count

Higher baseline hemoglobin concentration

Higher maximally tolerated dose of hydroxyurea

Better compliance (fewer unused pills returned at follow-up)

Greater treatment-related increases in hemoglobin concentration and mean corpuscular volume (MCV)

Greater treatment-related decreases in WBC count and reticulocyte count

Baseline Hb F percentages were in the range of 4 to 8 percent, and those in the top quartile of responses had Hb F percentages of approximately 21 percent.

At least some of this variability in response to hydroxyurea appears to be under genetic control, as discussed in more detail separately. (See "Fetal hemoglobin (hemoglobin F) in health and disease", section on 'Hemoglobin switching: genetic basis of HbF expression'.)

INDICATIONS AND EVIDENCE FOR EFFICACY

Indications and appropriate age to start therapy — Hydroxyurea (hydroxycarbamide, Droxia, Hydrea, Siklos) has been demonstrated to reduce complications and may increase life expectancy in SCD, especially in individuals with the most clinically severe genotypes (eg, homozygous SCD [Hb SS], sickle beta0 thalassemia). These benefits are summarized in the table (table 1). Hydroxyurea takes weeks to months to be effective and thus is used to prevent complications, not to treat them in the acute setting.

Our approach to deciding when hydroxyurea is appropriate in patients with homozygous Hb SS or sickle beta0 thalassemia is as follows (algorithm 1):

Infants 6 to 9 months – For infants between six and nine months with symptomatic disease (eg, severe anemia, dactylitis, acute pain episodes), we consider hydroxyurea. This is based on indirect evidence from older children and the potential value of preserving high levels of fetal hemoglobin. Limited evidence in this age group demonstrates higher levels of total hemoglobin and lower neutrophil counts at 24 months with early initiation of hydroxyurea.

Infants ≥9 months, children, and adolescents – For all infants nine months of age or older, children, and adolescents, we recommend hydroxyurea, regardless of disease severity. This is based on secondary endpoints from a randomized trial, as discussed below, and is consistent with the 2014 National Heart, Lung, and Blood Institute guidelines (NHLBI; in the National Institutes of Health in the United States) for the management of SCD.

Adults – For all adults with homozygous Hb SS or sickle beta0 thalassemia who are taking hydroxyurea, we recommend continuing hydroxyurea therapy unless they are attempting to conceive a child in the next three months or are pregnant.

For adults not taking hydroxyurea, we recommend starting hydroxyurea if they have at least one episode of moderate to severe sickle cell acute pain or acute chest syndrome in the last 12 months, symptomatic anemia, pulmonary hypertension, chronic hypoxemia, or chronic pain impacting their quality of life. We suggest starting hydroxyurea for men with episodes of priapism, adults with chronic kidney disease or proteinuria, or individuals with prior stroke if chronic transfusion therapy is not feasible.

The use of hydroxyurea in those with other SCD genotypes such as Hb SC disease or sickle beta+ thalassemia is individualized based on disease severity, which is more variable with these genotypes. We consider hydroxyurea for individuals with clinical manifestations similar to Hb SS or sickle beta0 thalassemia, and we do not recommend it for those with milder disease. This is based on limited case series that suggest hydroxyurea may be beneficial for patients with Hb SC and significant symptomatology [39,40]; there has been only one small, randomized trial evaluating hydroxyurea for Hb SC.

This approach is consistent with a 2008 National Institutes of Health consensus statement on the use of hydroxyurea in SCD, 2014 guidelines from the NHLBI, an updated systematic review, and 2020 guidelines from the American Society of Hematology (ASH), and the practices of other SCD experts [4,34,41-44]. A patient education booklet is available from ASH [45].

Hydroxyurea was approved for adults with SCD by the US Food and Drug Administration (FDA) in 1998 and by the European Medicines Agency in 2007 [16]. In 2017, the FDA extended its approval of hydroxyurea to include pediatric patients with SCD [46].

Hydroxyurea may be given concurrently with L-glutamine, voxelotor, or crizanlizumab, and the benefits of the combination appear to be greater than either agent alone. (See "Disease-modifying therapies to prevent pain and other complications of sickle cell disease", section on 'Importance of prevention'.)

Evidence for efficacy — Initial evidence for the efficacy of hydroxyurea in SCD came from studies in adults; these were followed soon after by studies in children and then in infants [34]. Subsequent prospective observational studies over many years have demonstrated a reduction in mortality with long-term hydroxyurea use. Despite the available evidence to support the use of hydroxyurea, many individuals with SCD who would benefit from this therapy do not receive it, or are treated in a suboptimal manner. Barriers to appropriate therapy should be addressed, as discussed below. (See 'Eliminating barriers to appropriate therapy' below.)

Improved survival — Evidence that hydroxyurea improves survival in SCD comes from the following observational studies:

A 2010 observational study reported the long-term outcomes of 299 individuals with SCD who were originally enrolled in a randomized trial (the Multicenter Study of Hydroxyurea in Sickle Cell Anemia [MSH] trial, described in greater detail below) comparing hydroxyurea with placebo and were subsequently followed for over 17 years [47,48]. Many assigned to placebo had subsequently switched to hydroxyurea after the short-term benefits of hydroxyurea had become apparent. Most of the participants were between 20 and 30 years of age at the time of initial study entry, and only individuals with Hb SS and at least three painful episodes per year were included. The majority of deaths occurred in individuals who were never exposed to hydroxyurea or who had <5 years of exposure. When evaluated according to length of hydroxyurea exposure in five-year increments, death rates per 100 person-years were as follows:

Never exposed – 5 deaths per 100 person-years

<5 years exposure – 6.8 deaths

5 to 10 years – 4.4 deaths

10 to 15 years – 1.8 deaths

≥15 years – 0 deaths

Additional benefits documented from the MSH trial are listed below. (See 'Reduced complications' below.)

A 2010 prospective nonrandomized study involving 131 Greek adults with SCD (all genotypes) followed for five to eight years found that mortality was lower in hydroxyurea-treated individuals compared with controls (10 versus 25 percent) [49]. The survival benefit was preserved across multiple SCD genotypes (homozygous Hb SS, Hb S beta0 thalassemia, and Hb S beta+ thalassemia).

A 2013 retrospective study involving 267 Brazilian children with SCD (ages 3 to 18 years, all genotypes) treated with hydroxyurea for a median of two years found that the hydroxyurea-treated children had lower mortality compared with controls (0.5 versus 5.5 percent [95% CI 0.92-0.99]) [50].

General information about survival and prognosis in SCD is presented separately. (See "Overview of the management and prognosis of sickle cell disease", section on 'Survival and prognosis'.)

Reduced complications — Evidence from several randomized trials and observational studies has demonstrated a reduction in SCD complications with hydroxyurea therapy in various age groups and SCD genotypes (table 1). These effects are most pronounced in individuals with clinically severe disease (frequent pain episodes or acute chest syndrome) and high-risk genotypes (eg, Hb SS or Hb S/beta0 thalassemia). Examples include the following, many of which have been summarized in systematic literature summaries and review articles [42,51,52]:

Infants – The 2011 BABY HUG trial randomly assigned 193 infants 9 to 18 months with Hb SS or sickle beta0 thalassemia irrespective of clinical severity to receive placebo or hydroxyurea (20 mg/kg daily without dose escalation) for two years [53,54]. The median age at entry was 13.6 months. Primary endpoints included splenic function and glomerular filtration rate (GFR), both of which were chosen because they become abnormal in SCD early in life. Compared with controls, hydroxyurea-treated infants had a trend towards improved splenic function (assessed by cell morphology after qualitative radionucleotide scans were discontinued) and no difference in GFR. However, there were significant reductions in clinical endpoints including pain episodes (hazard ratio [HR] 0.59; 95% CI 0.42-0.83), acute chest syndrome (HR 0.36; 95% CI 0.15-0.87), dactylitis (HR 0.27; 95% CI 0.15-0.50), and constipation (HR 0.33), as well as in need for transfusions (HR 0.55). Therapy was well tolerated with no severe adverse events.

Children – Although there are no randomized trials in children older than 18 months, observational studies have demonstrated similar benefits as those seen in infants and adults including reduction in painful episodes and hospitalizations [55-57]. In a cohort of 110 children treated with hydroxyurea, none had an abnormal measurement by transcranial Doppler (a surrogate marker for stroke risk) [58].

Adults – The 1995 MSH (Multicenter Study of Hydroxyurea in Sickle Cell Anemia) trial randomly assigned 299 adults with Hb SS and at least three painful episodes in a year to receive placebo or hydroxyurea, titrated to maximum tolerated dose and treated for approximately two years [59]. The primary outcome was the number of painful crises, defined as visits to a medical facility lasting more than four hours and resulting in treatment with an opioid analgesic; acute chest syndrome, priapism, or hepatic sequestration. Compared with controls, the hydroxyurea-treated individuals had a decrease in painful events (median annualized rate, 4.5 versus 2.5 events). The benefit persisted when crises severe enough to cause hospitalization were compared (median annualized rate, 2.4 versus 1.0). There were no differences in the rates of death, stroke, or hepatic sequestration.

As described above, extended observation of the original MSH participants suggested a survival benefit after 9 and 17.5 years. (See 'Improved survival' above.)

A 2010 observational study in which hydroxyurea was administered to 131 Greek adults for eight years found similar reductions in painful episodes, acute chest syndrome, hospitalizations, and mortality [49].

Hb SC disease – The CHAMPS trial randomly assigned 44 individuals with Hb SC disease who were five years old or older and at least one vaso-occlusive event (painful episode or acute chest syndrome) in the previous year to receive placebo or hydroxyurea for 2 to 10 months [60]. There were no differences in clinical events, but the sample size and study duration may have been insufficient to detect a benefit.

Retrospective series of children and adults with Hb SC disease who were treated with hydroxyurea have documented increases in Hb F levels and decreases in the frequency of acute pain events and hospitalization for pain [39,40].

The benefits of SCD extend to individuals in resource-limited settings such as sub-Saharan Africa, where SCD is common. Two studies published in 2017 and 2019 demonstrated reduced complications of SCD when hydroxyurea was administered in regions of Africa in which malaria, other infectious diseases, and malnutrition are common, and these found significantly lower rates of SCD complications and infections, as well as improved survival [61,62]. A third study from 2020 demonstrated the superiority of dose-escalated hydroxyurea over fixed-dose hydroxyurea in children in sub-Saharan Africa (median dose in the dose-escalated group, 29.5±3.6 mg/kg per day, versus 19.2±1.8 mg/kg per day in the fixed dose group) [63]. Children treated with dose-escalated hydroxyurea had fewer sickle cell-related complications (incidence rate ratio [IRR] 0.43 95% CI 0.34 to 0.56). The use of hydroxyurea in sub-Saharan Africa, including indications, dosing, and monitoring, are discussed in detail separately. (See "Sickle cell disease in sub-Saharan Africa", section on 'Hydroxyurea'.)

Stroke prevention trials have generally shown that chronic transfusion of RBCs is effective for primary and secondary prevention, although there is evidence of hydroxyurea noninferiority in a subset of individuals at high risk for a first stroke who have received chronic transfusions for two years or more. There is also limited evidence that hydroxyurea alone can prevent progression from conditional to abnormal TCD screening results and may reduce stroke risk in patients with abnormal TCD results who do not have access to transfusion therapy. This subject is discussed in detail separately. (See "Prevention of stroke (initial or recurrent) in sickle cell disease".)

Adverse effects of hydroxyurea are discussed below. (See 'Adverse effects' below.)

Other benefits — Other potential benefits of hydroxyurea include improved general quality of life and daily functioning. These in turn may translate to better school attendance and fewer days lost from work. A retrospective cohort study compared quality of life issues in 191 children with SCD. Those in the group treated with hydroxyurea reported better overall health-related quality of life (HRQL) and better physical HRQL than children who did not receive this therapy [64].

Cost savings due to reduced number of clinical encounters and hospitalizations may also be seen [65,66]. As an example, in the BABY HUG trial, which randomized young children to hydroxyurea versus placebo, use of hydroxyurea was associated with a 21 percent annual cost savings due to reduction in the number of hospitalizations [66]. A similar cost reduction was not demonstrated in the Multicenter Study of Hydroxyurea (MSH) in sickle cell anemia [47,48].

ELIMINATING BARRIERS TO APPROPRIATE THERAPY — Despite the clear clinical benefits of hydroxyurea (see 'Evidence for efficacy' above), many individuals are treated with hydroxyurea in a suboptimal manner or do not receive this therapy at all [4,34,41,67-70]. This lack of hydroxyurea use has been demonstrated in large reviews such as the following:

Children – A database review identified 7963 children with SCD in six states in the United States during the period from 2005 to 2012, representing 22,424 person-years [71]. Among all person-years, 78 percent had 0 days of hydroxyurea in a year; 3 percent had 1 to 30 days, and 20 percent had >30 days. Of those who received hydroxyurea, the average number of days' supply in a year was 189. Factors predictive of receiving hydroxyurea included younger age of the child and more frequent outpatient visits. The odds of receiving hydroxyurea did not increase over the course of the study, and in fact appears to have decreased in the final years.

A subsequent study of over 5000 children with SCD enrolled in Medicaid reported an increase in hydroxyurea use from 14 percent in 2009 to 28 percent in 2015 [72].

Adults – A database review identified 2086 adults with SCD based on insurance codes identified 677 (approximately one-third) who had at least three pain-related hospitalizations or emergency department visits within a one-year period [73]. However, only 86 of these individuals with frequent painful episodes (15 percent) filled a prescription for hydroxyurea within three months of the third encounter; this percentage increased to 23 percent at one year.

These data suggest that significant barriers to appropriate therapy persist for patients with SCD. However, the first pediatric and adult studies were conducted before the 2014 practice guideline from the National Heart, Lung, and Blood Institute (NHLBI), which recommended hydroxyurea for all infants, children, and adolescents with SCD regardless of symptoms, as well as for adults with three or more vaso-occlusive episodes in a year [43]. Evidence-based strategies are required to address best practices to improve adherence to hydroxyurea in children and adults with SCD.

Several studies have defined specific barriers to appropriate administration of hydroxyurea that may be addressed. These seem to be extensive and span patient-, parent/family/caregiver-, provider-, and system-level obstacles to use. Examples include hesitancy among providers about the safety and efficacy of hydroxyurea; patient concerns about carcinogenicity, teratogenicity, and other side effects; and difficulty in complying with daily dosing, obtaining refills in a timely fashion, and adhering to frequent clinical and laboratory monitoring. In the pediatric setting, limited access to hydroxyurea solution, which can be prepared only by specialized compounding pharmacies, is also a factor. Finally, inadequate health insurance coverage is a significant barrier among those who live in poverty, especially with the high cost of the hydroxyurea formulation approved for children (Siklos) [74,75].

To ensure good adherence to, and maximal benefit from, hydroxyurea therapy, it is important for providers to clearly present the risks and benefits of hydroxyurea, as well as the practical aspects of administration.

BASELINE TESTING — We obtain the following studies before starting hydroxyurea:

Complete blood count (CBC) with red blood cell indices, white blood cell differential, platelet count

Reticulocyte count

Hemoglobin F (Hb F) percentage (quantitative measure)

Pregnancy test for females of childbearing potential

The baseline mean corpuscular volume (MCV) should be noted because hydroxyurea causes macrocytosis and might mask iron, B12, or folate deficiency. If the baseline MCV is high, we check vitamin B12 and folate levels if the history is suggestive of vitamin deficiency associated with increased MCV and thyroid studies if a family history or symptoms are suggestive of thyroid disease. (See "Macrocytosis/Macrocytic anemia", section on 'Causes of macrocytosis/macrocytic anemia'.)

If the baseline MCV is low, we take a dietary history for possible iron deficiency and consider whether the patient may have alpha thalassemia trait, an established risk factor for low MCV values. Approximately 40 percent of unselected children with Hb SS or sickle beta0 thalassemia will have single alpha globin gene deletion. Thus, consistently low MCV levels are most likely related to alpha globin gene deletion. Those with acquired microcytosis should be evaluated for iron deficiency (see "Microcytosis/Microcytic anemia", section on 'Causes of microcytosis'). Individuals with renal insufficiency are treated with a lower initial dose of hydroxyurea. (See 'Dosing in chronic kidney disease' below.)

ADMINISTRATION AND DOSING

Initial dosing — Hydroxyurea is initiated at a low dose and gradually increased to a dose that does not cause severe hematologic toxicity (see 'Monitoring and dose titration' below).

The dose is administered as a single once-daily dose, although it can be given in divided doses if this is preferable to the patient. To improve adherence, a single hydroxyurea dose per day is recommended.

Patients who experience gastrointestinal upset with therapy may have improvement if the dose is given at bedtime. (See 'GI side effects' below.)

A 2019 pharmacokinetic model for hydroxyurea therapy in children provided evidence that the patient could reach maximum tolerated dose (MTD) faster using pharmacokinetic-guided dosing than with the standard increment of 5 mg/kg dose every eight weeks [76]. Although MTD was reached more rapidly, there was no evidence of decreased morbidity from SCD.

Individuals with impaired renal function are given a lower initial dose. (See 'Dosing in chronic kidney disease' below.)

Concomitant use of L-glutamine does not affect hydroxyurea dosing.

Infants and young children — For infants younger than one year of age treated with hydroxyurea who have good kidney function (creatinine clearance >60 mL/minute), we use an initial dose of 20 mg/kg per day. This starting dose was well tolerated in the BABY HUG and NOHARM trials [53,61,77].

Hydroxyurea is not commercially available as a liquid; compounding pharmacy support is required to prepare an oral solution from the commercially available capsules. Such oral solutions (typical concentration, 100 mg/mL) have been shown to be chemically stable at room temperature for up to six months [78].

Cost and lack of access to a reliable compounding pharmacy for preparation of the oral solution are potential barriers to therapy in very young children. The following may be useful in such cases:

One formulation (brand name, Siklos) is available as 100 mg and scored 1000 mg tablets that can be dissolved in water [79].

Another alternative is to round off the dose to the nearest available capsule size and instruct parents on how to open the capsules, mix the contents into a small quantity of food, and administer this mixture to the child.

Another option, if the desired dose is not available with the capsule, is to give four days of one dose and three days of a second dose. For example, if the desired hydroxyurea dose is 1250 mg daily, the child can receive 1000 mg on Monday, Tuesday, Wednesday, and Thursday, and they can receive 1500 mg on Friday, Saturday, and Sunday. This averages to a weekly dose of 1250 mg/day.

For ease of dosing of either the compounded liquid, dissolvable tablets, or capsules, the dose can be rounded off to the nearest 2.5 mg/kg (eg, give 200 mg instead of 180 mg for a 9 kg infant, or 400 mg instead of 450 mg for a 22 kg child).

Monitoring and dose titration is described below. (See 'Monitoring and dose titration' below.)

Older children, adolescents, and adults — Individuals who are able to take hydroxyurea in pill form are given a dose in increments rounded to the nearest capsule size. The recommended initial oral dose of hydroxyurea for children with a creatinine clearance >60 mL/min is 20 mg/kg per day, rounded to the nearest 2.5 mg/kg per day. Adults may be started on 15 to 20 mg/kg per day with lower doses for older adults and those with normal to mildly reduced glomerular filtration rates. Capsules are available in 200 mg, 300 mg, 400 mg, and 500 mg doses.

Subsequent dose increments are determined by the patient's response. Monitoring and dose titration is described below. (See 'Monitoring and dose titration' below.)

Dosing in chronic kidney disease — Hydroxyurea is excreted by the kidney, and individuals with end-stage kidney disease or creatinine clearance <60 mL/minute may have higher exposure for a given dose.

In these individuals, we typically start at half the starting dose that would be used for normal kidney function (7.5 mg/kg daily rather than 15 mg/kg daily). Smaller increments of dose titration may also be used in these individuals.

It is important to continue to monitor renal function during dose titration. (See 'Monitoring and dose titration' below.)

Monitoring and dose titration — Reduction in vaso-occlusive pain events and other vaso-occlusive complications is the major, patient-important clinical endpoint in hydroxyurea therapy. Bone marrow suppression is the major dose-limiting toxicity.

Although clinical benefit is the goal of therapy, we use hematologic parameters as a surrogate for clinical effects and an endpoint for dose titration, referred to as the maximum tolerated dose (MTD), as outlined in the table (table 2). Monitoring is more frequent while the dose is being adjusted and continues at less frequent intervals once the individual is on a stable dose. In resource-limited settings where frequent complete blood counts (CBCs) are not an option, less intensive dosing with less frequent monitoring may be feasible and may reduce vaso-occlusive complications, including stroke. (See "Sickle cell disease in sub-Saharan Africa", section on 'Hydroxyurea'.)

There is some nuance to determining the ideal set of parameters to monitor, as there is a need to balance the ease of use (especially by non-SCD experts) and cost (especially in low-resource settings) with the added benefit of including more parameters. As an example, some experts advocate use of the hemoglobin/hematocrit and absolute reticulocyte count (ARC), as long as the additional monitoring is not a disincentive to prescribing hydroxyurea.

General principles include the following:

The initial dose is calculated based on weight, as indicated above. (See 'Initial dosing' above.)

The dose is increased approximately every eight weeks (range, 6 to 12 weeks) by 5 mg/kg daily, to a maximum dose of 35 mg/kg daily or 2500 mg daily, or until one or more of the MTD parameters are reached. The complete blood count (CBC) and reticulocyte count should be obtained every four weeks while the dose is being increased as the hematologic response may evolve over this period.

Parameters to determine MTD include any of the following:

An absolute neutrophil count (ANC) (calculator 1) of 1500 to 3000/microL (infants with lower baseline ANC may tolerate an on-therapy ANC as low as 1000/microL)

ARC of 80,000 to 100,000/microL

A platelet count of 80,000 to 150,000/microL

In practice, the most common parameter determining MTD is the ANC, but some patients will reach the target ARC before lowering of the ANC to the target range. Reduction of the platelet count is rarely the criterion for determining MTD except in patients that have splenomegaly.

Reticulocytopenia and worsening anemia occurs more frequently in patients with chronic kidney disease, iron deficiency, viral bone marrow suppression, and delayed hemolytic transfusion reactions [80-83]. A low ARC with anemia may be a more common reason to hold hydroxyurea in adults who are hospitalized or who have significant comorbidities; it may be less commonly used in children.

In patients with a strong response in hemoglobin level (eg, hemoglobin >9 g/dL), an ARC below 80,000/microL is acceptable as long as the hemoglobin remains stable over several months.

If significant cytopenias develop (eg, ANC <1000/microL, platelet count of <80,000/microL), the dose is temporarily held and the CBC is rechecked once per week until the value is greater than that listed above. The drug is then restarted at the same dose if recovery occurs within a week, or at a lower, previously tolerated dose (eg, 2.5 to 5 mg/kg daily less than the dose that caused hematologic toxicity that exceeded the MTD range) if the myelosuppression was prolonged or recurs.

Detailed criteria for determining eligibility for dose escalation, MTD, and toxicity are presented in the table (table 2).

Routine monitoring of Hb F percentage is not necessary as it is not used to titrate the dose, although it may be checked periodically to evaluate efficacy (eg, once per year). Most hydroxyurea responses are associated with at least a twofold increase in Hb F percentage over baseline, often much greater. Hb F that is >20 percent of total hemoglobin is ideal, and there is no upper limit [10]. Greater increases in total hemoglobin (greater than 1 g/dL) or Hb F percentage generally correlate with greater efficacy. (See 'Increased Hb F production' above.)

A pregnancy test should be obtained if menses are delayed more than two weeks for females of childbearing potential. Formal discussion about the teratogenic impact of hydroxyurea and contraception options should be discussed.

Once the MTD is reached, monitoring is continued at less-frequent intervals. Ongoing monitoring is needed because dose effects and kidney function can change over time. The CBC with platelet count and reticulocyte count can be measured once every three months; and liver and kidney function are evaluated as needed for ongoing monitoring of sickle cell disease (at least every 12 months, more frequently if there is known or suspected organ dysfunction). Additional monitoring may be needed if there is an intercurrent illness that might affect renal or hematologic function.

Can therapy be discontinued? — Hydroxyurea therapy should be continued for as long as it is tolerated and effective. Exceptions include the following:

Persistent cytopenias despite dose reductions, which can be a sign of reduced bone marrow capacity (see 'Monitoring and dose titration' above). Other causes of cytopenias (kidney or liver disease, nutritional deficiencies, hypothyroidism) should be excluded.

Worsening kidney function, which may increase the risk of excessive exposure due to delayed clearance of hydroxyurea, as well as of decreased erythropoiesis due to decreased erythropoietin production. This can usually be managed with a reduction in dose and sometimes the addition of an erythroid stimulating agent if the primary toxicity is reticulocytopenia. (See 'Myelosuppression' below.)

Severe cutaneous toxicity, pancreatitis, or other adverse effects that interfere with quality of life. (See 'Adverse effects' below.)

Both women and men attempting conception, and during pregnancy and breastfeeding; therapy is restarted after the infant is no longer breastfeeding. (See 'Pregnancy and breastfeeding' below.)

Individuals who have a complication necessitating chronic (regular) transfusion therapy while taking hydroxyurea may discontinue hydroxyurea if the complication occurred despite hydroxyurea therapy. (See 'Hospitalization' below.)

ADVERSE EFFECTS — Adverse effects of hydroxyurea in individuals with SCD have been assessed in several studies, as summarized in the table (table 3). Evidence from over 30 years of use in individuals with SCD has demonstrated that long-term use of hydroxyurea is not associated with clinically significant adverse effects on growth or development. This includes very young children enrolled in the BABY HUG trial (ages 9 to 18 months), who had no adverse effects on growth or other anthropomorphic measures during two years of hydroxyurea therapy [84].

The BABY HUG trial is one of the few trials that compared fixed moderate dose hydroxyurea at 20 mg/kg/day with placebo in children with sickle cell anemia (SCA) [53]. Importantly, infants treated with hydroxyurea did not have significant differences from those treated with placebo in rates of severe neutropenia (absolute neutrophil count [ANC] <500/microL), thrombocytopenia (platelet count <80,000/microL), anemia (hemoglobin <7 g/dL), reticulocytopenia (absolute reticulocyte count [ARC] <80,000/microL), or abnormal tests of liver function (alanine aminotransferase [ALT] >150 units/L or bilirubin >10 mg/dL).

These data are among the most compelling evidence that moderate-dose hydroxyurea may not require the same degree of close laboratory monitoring as maximum-tolerated-dose hydroxyurea.

Impact on vaccination — The package insert for hydroxyurea states that live virus vaccines should be avoided. We have not encountered any problems related to live vaccines, and we provide all routine vaccinations to individuals receiving hydroxyurea, including live virus vaccines. The benefits of reducing infections in this population of functionally asplenic individuals is likely to outweigh any risks associated with live vaccines.

The BABY HUG trial measured immunological effects of hydroxyurea including T-cell subsets, naïve and memory T-cells and antibody response to inactivated and live vaccines [85]. Hydroxyurea treatment resulted in lower total lymphocytes, CD4-positive T-cells, and memory T-cells; however, counts remained within the normal range for healthy children. Antibody responses to pneumococcus, mumps, and rubella were unchanged, but there was a delay of 14 days in developing protective levels of measles antibody.

Myelosuppression — Hydroxyurea is relatively nontoxic, with myelosuppression as the major dose-limiting toxicity.

For most individuals, myelosuppression is predictable, dose-dependent, and reversible [39,40,47,48]. Myelosuppression is used to adjust hydroxyurea dosing and can be easily controlled as long as there is regular hematologic monitoring and dose reduction for severe neutropenia, anemia, or thrombocytopenia. (See 'Monitoring and dose titration' above.)

Some individuals have severe myelosuppression such that they are unable to tolerate even low doses of the drug. (See 'Severe myelosuppression' below.)

Falsely elevated serum creatinine and other laboratory tests — A point-of-care device for measuring serum creatinine (the i-STAT system) that is used for routine testing in some clinical laboratories can give a falsely elevated creatinine measurement in patients receiving hydroxyurea [86].

Individuals receiving hydroxyurea who have a high serum creatinine should be retested using a different method to distinguish this artifact from true renal disease. (See "Sickle cell disease effects on the kidney".)

Hydroxyurea has also been reported to cause falsely elevated results for urea, uric acid, and lactic acid, due to analytical interference of the drug with the enzymes used to determine these values.

GI side effects — Some individuals receiving hydroxyurea may have gastrointestinal toxicity with nausea or anorexia. In the HUG-KIDS study, the two children with gastrointestinal upset attributed to hydroxyurea had improvement in symptoms when the dose was given at bedtime.

Dermatologic — Skin and nail changes and leg ulcers have been described, although it is not clear whether hydroxyurea was in fact responsible [42].

Individuals receiving doses several times the therapeutic dose (eg, in overdose) have developed mucocutaneous toxicity, with erythema and soreness on the palms and soles of the hands and feet; scaling, generalized hyperpigmentation; and/or stomatitis. In such cases the dose should be stopped and supportive treatments used.

Pregnancy outcomes — Data on adverse pregnancy outcomes are extremely limited. A review of nine pregnancies in individuals who were receiving hydroxyurea either during the first trimester or throughout the pregnancy and 10 pregnancies in which the male partner was receiving hydroxyurea at the time of conception did not reveal evidence of mutagenicity or teratogenicity [87].

We discontinue hydroxyurea prior to pregnancy (or as soon as pregnancy is documented), as discussed below. (See 'Pregnancy and breastfeeding' below.)

Fertility — One of the consistently noted barriers to hydroxyurea therapy has been a concern about possible effects on fertility, especially since SCD may already confer a reduction in sexual functioning, sperm counts, or other reproductive factors [88,89].

Males – There has been concern based on data from animal models that suggest a further reduction in spermatogenesis with hydroxyurea. However, preliminary data from two pilot studies provide some reassurance that fertility is unlikely to be permanently impaired by the drug.

In one study, sperm parameters were compared in 15 adolescents and young adults with SCD who were treated with hydroxyurea during childhood (median duration, 4 years) and 23 controls who had not received hydroxyurea [90]. Most had more than one sample analyzed. There were abnormalities of sperm number and quality, but sperm parameters were not adversely affected in the individuals treated with hydroxyurea.

In another study, 30 boys with SCD who were undergoing hematopoietic stem cell transplant had harvesting of testicular tissue for fertility preservation; 17 had been treated with hydroxyurea (for a median of 36 months) and 13 had not, allowing a comparison between groups [91]. On histological examination of resected testicular tissue, spermatogonial tissue was similar between groups. However, compared with controls without SCD, all of the children with SCD had a reduced spermatogonial stem cell pool, indicating that SCD contributes to spermatogonial depletion.

Larger studies are awaited.

It is possible that earlier cohort studies that observed reduced sperm counts and low sperm quality may have reflected changes due to SCD rather than hydroxyurea, although they do not eliminate a possible role for hydroxyurea in reduced male fertility [92,93]. If an effect on fertility exists, it is not known whether the effect is reversible with cessation of hydroxyurea.

Females – There are few data on the impact of hydroxyurea therapy on fertility in women with SCD [89].

A study from 2020 found an association between hydroxyurea and decreased ovarian reserve in a cohort of women with SCD, using anti-müllerian hormone (AMH) as a marker [94]. In a multivariable analysis, hydroxyurea use was associated with decreased AMH levels (see "Evaluation of female infertility", section on 'Anti-müllerian hormone'). Further research in this area is needed so that individuals can be accurately counseled about this possible risk and be offered measures for fertility preservation when appropriate.

No evidence for carcinogenicity — A theoretical concern with long-term hydroxyurea administration is whether the drug is a carcinogen in individuals with SCD. This concern is mostly based on reports of increased incidence of malignancy in individuals receiving hydroxyurea for myeloproliferative neoplasms (MPNs), such as essential thrombocythemia (ET) or polycythemia vera (PV), for whom the baseline risk of hematologic malignancy is increased. Even for these MPNs, it is not clear whether hydroxyurea is a mild leukemogen or whether it merely unmasks the risk of leukemia by allowing patients to live longer.

There is no evidence from the published literature or our clinical experience that suggests individuals with SCD receiving hydroxyurea have an increased risk of malignancy compared with individuals with SCD who are not receiving hydroxyurea. Examples of the available evidence include the following:

Reports from 2003 and 2010 described long-term outcomes in 233 adults who participated in the Multicenter Study of Hydroxyurea in Sickle Cell Anemia (MSH) trial, which evaluated the role of hydroxyurea in reducing painful episodes [47,48]. Nine years after the trial started, three patients had developed a malignancy, including one each of breast, cervix, and uterus cancer. At least one of the three patients had a pre-existing condition placing her at increased risk for cancer. There were no additional cancers described after an additional 17.5 years of follow-up, with an overall rate of malignancy of 0.1 per 100 patient-years.

A 2004 report described outcomes in 122 children treated with hydroxyurea for up to eight years [95]. There were no malignancies or cases of myelodysplasia, and in vitro testing for increased DNA mutations using VDJ rearrangements in 26 of the children showed no evidence of mutagenesis.

A 2011 report tested in vitro parameters of chromosome stability in 50 children with SCD who had received hydroxyurea for up to 12 years compared with 28 children who had not received hydroxyurea [96]. Compared with controls, the children who had received hydroxyurea had less chromosome damage and similar chromosome repair.

Additional long-term studies to address this concern are needed, as people with SCD have an increased risk of hematologic malignancies without exposure to hydroxyurea [97].

While it is possible that these studies failed to detect a very small increase in carcinogenicity, this risk, if present, must be weighed against the clinically significant benefits of hydroxyurea in reducing complications of SCD and possibly prolonging survival.

SPECIAL SCENARIOS

Lack of hematologic response — Lack of hematologic response (eg, failure of the hemoglobin F [Hb F] to increase by twofold or more or failure of the mean corpuscular volume [MCV] to increase to >100 fL or by at least 10 fL) is sometimes seen and is usually attributed to a failure to take the medication as directed. Often, medication nonadherence is the reason for lack of a hematologic response; however, lack of response should not automatically be used as a surrogate for lack of compliance with therapy. This is because a significant proportion of individuals who take hydroxyurea as directed do not have a hematologic response. When efficacy is carefully tracked and compliance is assumed to be 100 percent, it appears that approximately 5 to 10 percent of children and 25 to 30 percent of adults have true lack of efficacy from hydroxyurea [9].

The reason for this lack of response in some individuals is not completely understood but may be related to one of a number of factors that control Hb F production. (See 'Mechanism of action' above.)

For an individual who does not have the expected response, the following is appropriate:

Proper daily use of hydroxyurea should be reviewed with the patient (or parents), and barriers to proper use addressed (see 'Eliminating barriers to appropriate therapy' above). In particular, patient adherence should be reviewed and optimized as best as possible before significant dose escalation to reduce the risk of excessive myelosuppression at higher doses.

If the individual has only been taking the medication for a short time, we continue therapy for six months after the maximum tolerated dose has been achieved, to assess clinical improvement. (See 'Monitoring and dose titration' above.)

If the patient has clinical benefit from treatment despite lack of the expected hematologic changes, we continue the medication as long as there are no apparent adverse effects.

An erythroid-stimulating agent can be added to hydroxyurea, typically at doses similar to those used for cancer or inflammatory causes of anemia. This approach was suggested in the American Society of Hematology 2019 guidelines for sickle cell disease: cardiopulmonary and kidney disease [98]. The endogenous amount of erythropoietin is usually elevated but decreased from the greatly elevated levels typically seen in young people with SCD secondary to the development of renal disease and aging.

In some cases, iron, vitamin B12, or folate deficiency may limit response to hydroxyurea. If the patient's clinical or laboratory picture suggests these possibilities, assess and supplement as needed. (See "Iron deficiency in infants and children <12 years: Screening, prevention, clinical manifestations, and diagnosis" and "Iron requirements and iron deficiency in adolescents" and "Causes and diagnosis of iron deficiency and iron deficiency anemia in adults" and "Clinical manifestations and diagnosis of vitamin B12 and folate deficiency".)

If the patient is taking the drug as directed and not experiencing any clinical benefit (eg, reduction in pain episodes or other vaso-occlusive complications) and/or is not having the expected hematologic changes, we may discontinue the medication, as the risks of continuing therapy, albeit small, may no longer be justified.

For these patients, other options include other disease-modifying therapies, chronic transfusions, investigative agents, and hematopoietic cell transplantation. These approaches are not mutually exclusive. The choice among them is individualized according to the needs of the individual. The risks and benefits of these therapies are discussed in separate topic reviews. (See "Disease-modifying therapies to prevent pain and other complications of sickle cell disease" and "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques" and "Investigational therapies for sickle cell disease" and "Hematopoietic stem cell transplantation in sickle cell disease".)

Severe myelosuppression — Some individuals treated with hydroxyurea may have more severe myelosuppression than expected. Typically, the drug is held and restarted at a lower dose, and a maximum tolerated dose can eventually be determined. (See 'Monitoring and dose titration' above.)

Rarely, an individual may be unable to tolerate hydroxyurea even at reduced doses (eg, severe renal insufficiency). For these patients, options for treatment include chronic transfusion therapy, investigational agents, hematopoietic cell transplantation, and supportive/symptomatic care. The choice among these is individualized according to the needs of the individual. The risks and benefits of these therapies are discussed in detail separately. (See "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques" and "Investigational therapies for sickle cell disease" and "Hematopoietic stem cell transplantation in sickle cell disease".)

Transition from chronic transfusions to hydroxyurea — Selected individuals can be transitioned from chronic transfusions to hydroxyurea therapy, as shown in the table (table 2). This subject is discussed in detail separately. (See "Prevention of stroke (initial or recurrent) in sickle cell disease", section on 'Chronic transfusion followed by transition to hydroxyurea'.)

Pregnancy and breastfeeding — Hydroxyurea is considered to be an embryonic and fetal toxin based on animal studies, and it should not be used during pregnancy. However, evidence of human teratogenicity is lacking. Small amounts of hydroxyurea are excreted in breast milk (mean of 2.2 mg with a 1000 mg daily dose; 1.2 mg in the first three hours) [99]. Our approach to preconception planning, pregnancy, and postpartum use is as follows:

Females of reproductive potential who are receiving hydroxyurea should be counseled to use effective contraception. For those who wish to become pregnant, it is prudent to discontinue hydroxyurea three months before conception.

For women with severe disease and no other treatment options besides hydroxyurea, we discuss the balance between maternal and fetal risks with stopping versus continuing hydroxyurea. In rare cases, we suggest continuing hydroxyurea until pregnancy is documented, stopping it for the first trimester, and restarting it for the second and third trimesters. The third trimester and early postpartum are the times in which 90 percent of pain and acute chest syndrome events occur during pregnancy [100].This approach is controversial and should be discussed in detail with the person with SCD. Consideration should be focused on abating vaso-occlusive events in the third trimester and the first six weeks postpartum.

Other aspects of pre-pregnancy planning are discussed in detail separately. (See "Sickle cell disease: Pregnancy considerations", section on 'Management of medications and immunizations'.)

Males of reproductive potential who are receiving hydroxyurea should be advised to use effective contraception. For those who wish to have a child, it is prudent to discontinue hydroxyurea three to six months before conception because spermatogenesis takes approximately two to three months. (See "Male reproductive physiology", section on 'Spermatogenesis'.)

If a patient becomes pregnant while taking hydroxyurea, we discontinue the drug immediately. Most outcomes of pregnancies conceived while taking hydroxyurea are good, although there are few data that address this issue. Clinical and hematologic monitoring upon discontinuation of the drug are important, since the patient may experience a worsening of symptoms (eg, pain crises) once the drug is stopped. We restart hydroxyurea once the infant is no longer breastfeeding. Transfusion during pregnancy is discussed separately (see "Sickle cell disease: Pregnancy considerations", section on 'Transfusion therapy').

The duration of breastfeeding is based on balancing the benefits to the infant and mother against the risks associated with not taking hydroxyurea for that individual. Breastfeeding is considered to be contraindicated in women with SCD receiving hydroxyurea, but the evidence to support the recommendation is limited, and the small amount of hydroxyurea secreted in breast milk is unlikely to cause myelosuppression. In a small study of 16 women with SCD, hydroxyurea transferred into breastmilk with a relative infant dose of 3.4 percent [99]. The threshold safety for hydroxyurea breastmilk is 5 percent and could be reduced to approximately 1 mg per 24 hours by pumping and dumping breast milk for three hours after taking the dose. These preliminary data suggest that breastfeeding should be discussed in women with SCD on hydroxyurea therapy. (See "Infant benefits of breastfeeding" and "Maternal and economic benefits of breastfeeding".)

Hospitalization — Patients receiving hydroxyurea should continue the drug during an acute hospitalization unless there is significant myelosuppression or severe acute kidney injury, both of which would warrant discontinuation and possibly restarting at a lower dose. (See 'Initial dosing' above.)

Hydroxyurea generally can be continued during hospitalization for a febrile illness or an uncomplicated infection. It is prudent to temporarily hold hydroxyurea during severe infectious illnesses with worsening cytopenias or in the setting of worsening clinical status in order to avoid severe myelosuppression. We do not hold hydroxyurea therapy before surgery, because the major concern postoperatively is acute chest syndrome, and hydroxyurea decreases the incidence of acute chest syndrome. There is a risk of poor wound healing with myelosuppression, but this risk is far less than the risk of acute chest syndrome, a potentially life-threatening postoperative complication.

Hydroxyurea is also continued for individuals who are admitted for complications that require treatment with acute transfusions because the effect of transfusions is likely to be transient. For those who are transitioned to chronic (regular) transfusion therapy, hydroxyurea may be discontinued, since the complication that necessitated transfusion occurred despite hydroxyurea therapy. Restarting the hydroxyurea at a future time may be appropriate if the individual had some degree of response; this decision is individualized.

For those who are unable to take anything by mouth, hydroxyurea must be held, as there is no parenteral formulation. The medication should be restarted as soon as reasonably possible, using the nasogastric route when necessary.

If a patient with SCD who is not receiving hydroxyurea is hospitalized and a decision is made to start the medication, this is typically done on an outpatient basis to allow for full discussion of the risks and benefits and establishment of a mutually agreed upon surveillance schedule.

Resource-limited settings — (See "Sickle cell disease in sub-Saharan Africa", section on 'Hydroxyurea'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Sickle cell disease and thalassemias".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Sickle cell disease (The Basics)")

SUMMARY AND RECOMMENDATIONS

Mechanism of actionHydroxyurea reduces vaso-occlusive events such as pain episodes and acute chest syndrome (ACS) in people with sickle cell disease (SCD). A number of mechanisms may be involved, the most important of which is an increase in the level of fetal hemoglobin (Hb F), which reduces sickle hemoglobin (Hb S) polymerization, sickling, and vaso-occlusion. (See 'Mechanism of action' above.)

Indications – Our approach to deciding whether to use hydroxyurea is shown in the algorithm (algorithm 1) and discussed above. (See 'Indications and evidence for efficacy' above.)

For infants ≥9 months, children, and adolescents who have Hb SS or sickle beta0 thalassemia and any clinical severity, we recommend hydroxyurea (Grade 1B). This is based on a randomized trial and several observational studies that have demonstrated reductions in vaso-occlusive complications (eg, dactylitis, painful episodes, acute chest syndrome) with hydroxyurea therapy. Hydroxyurea may be considered in selected infants between six and nine months. (See 'Infants and young children' above and 'Older children, adolescents, and adults' above.)

For all adults with homozygous Hb SS and sickle beta0 thalassemia, we continue (or offer) hydroxyurea therapy unless they are trying to conceive in the next three months or a woman is pregnant.

Hydroxyurea is effective in reducing complications of SCD, as summarized in the table (table 1), and the rate of infectious diseases in African countries with high rates of malaria, other infectious diseases, and malnutrition. The use of hydroxyurea in sub-Saharan Africa is discussed in detail separately. (See "Sickle cell disease in sub-Saharan Africa", section on 'Hydroxyurea'.)

The use of hydroxyurea in those with other SCD genotypes such as Hb SC disease or sickle beta+ thalassemia is individualized based on disease severity, which is more variable with these genotypes. We consider hydroxyurea for those with clinical manifestations similar to Hb SS or sickle beta0 thalassemia and do not recommend it for those with more mild disease. (See "Disease-modifying therapies to prevent pain and other complications of sickle cell disease", section on 'Importance of prevention'.)

Baseline testing – Baseline testing prior to initiating hydroxyurea includes a complete blood count (CBC) with differential, platelet count, reticulocyte count, Hb F percentage, tests of renal and hepatic function, and a pregnancy test for females of childbearing potential. (See 'Baseline testing' above.)

Dosing – We generally use an initial hydroxyurea dose of 20 mg/kg daily in infants and children, and 15 to 20 mg/kg daily in adults; the daily dose may be rounded to the nearest 2.5 mg/kg. Those with creatinine clearance <60 mL/minute have the dose reduced by half. The dose is titrated typically by 5 mg/kg per day every eight weeks to maximum tolerated dose using hematologic parameters (table 2). Compounding support is needed to create a liquid formulation. Treatment is continued indefinitely in those for whom it is effective. (See 'Administration and dosing' above.)

Adverse effectsHydroxyurea is relatively nontoxic, with myelosuppression as the major, predictable, dose-limiting toxicity (table 3). Other adverse effects include gastrointestinal upset and skin, hair, and nail changes. Some measures of serum creatinine may be abnormally altered. Preliminary data suggest individuals with SCD may have reduced sperm number and quality, but hydroxyurea does not appear to worsen these parameters. Concerns have been raised possible carcinogenesis. Further studies are needed. (See 'Adverse effects' above.)

Approach to lack of response – Our approach to managing individuals with no apparent response and other clinical scenarios are presented above and in separate topic reviews. (See 'Special scenarios' above and "Disease-modifying therapies to prevent pain and other complications of sickle cell disease", section on 'Importance of prevention' and "Sickle cell disease: Pregnancy considerations" and "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques".)

Alternatives to hydroxyurea – General overviews of SCD management and discussions of other therapies to reduce vaso-occlusive events, include red blood cell (RBC) transfusion and hematopoietic cell transplantation (HCT) are presented separately. (See "Overview of the management and prognosis of sickle cell disease" and "Sickle cell disease in infancy and childhood: Routine health care maintenance and anticipatory guidance" and "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques" and "Hematopoietic stem cell transplantation in sickle cell disease".)

ACKNOWLEDGMENTS — We are saddened by the death of Stanley L Schrier, MD, who passed away in August 2019. The editors at UpToDate gratefully acknowledge Dr. Schrier's role as Section Editor on this topic, his tenure as the founding Editor-in-Chief for UpToDate in Hematology, and his dedicated and longstanding involvement with the UpToDate program.

The UpToDate editorial staff also acknowledges extensive contributions of Donald H Mahoney, Jr, MD to earlier versions of this topic review.

  1. McGann PT, Ware RE. Hydroxyurea therapy for sickle cell anemia. Expert Opin Drug Saf 2015; 14:1749.
  2. Singh A, Xu YJ. The Cell Killing Mechanisms of Hydroxyurea. Genes (Basel) 2016; 7.
  3. Ware RE, de Montalembert M, Tshilolo L, Abboud MR. Sickle cell disease. Lancet 2017.
  4. Platt OS. Hydroxyurea for the treatment of sickle cell anemia. N Engl J Med 2008; 358:1362.
  5. Goldberg MA, Brugnara C, Dover GJ, et al. Treatment of sickle cell anemia with hydroxyurea and erythropoietin. N Engl J Med 1990; 323:366.
  6. Franco RS, Yasin Z, Palascak MB, et al. The effect of fetal hemoglobin on the survival characteristics of sickle cells. Blood 2006; 108:1073.
  7. Setty BN, Kulkarni S, Dampier CD, Stuart MJ. Fetal hemoglobin in sickle cell anemia: relationship to erythrocyte adhesion markers and adhesion. Blood 2001; 97:2568.
  8. Ballas SK, Dover GJ, Charache S. Effect of hydroxyurea on the rheological properties of sickle erythrocytes in vivo. Am J Hematol 1989; 32:104.
  9. Rodgers GP, Dover GJ, Noguchi CT, et al. Hematologic responses of patients with sickle cell disease to treatment with hydroxyurea. N Engl J Med 1990; 322:1037.
  10. Estepp JH, Smeltzer MP, Kang G, et al. A clinically meaningful fetal hemoglobin threshold for children with sickle cell anemia during hydroxyurea therapy. Am J Hematol 2017; 92:1333.
  11. Perrine RP, Pembrey ME, John P, et al. Natural history of sickle cell anemia in Saudi Arabs. A study of 270 subjects. Ann Intern Med 1978; 88:1.
  12. Wood WG, Pembrey ME, Serjeant GR, et al. Hb F synthesis in sickle cell anaemia: a comparison of Saudi Arab cases with those of African origin. Br J Haematol 1980; 45:431.
  13. Brittenham G, Lozoff B, Harris JW, et al. Sickle cell anemia and trait in southern India: further studies. Am J Hematol 1979; 6:107.
  14. Diop S, Thiam D, Cisse M, et al. New results in clinical severity of homozygous sickle cell anemia, in Dakar, Senegal. Hematol Cell Ther 1999; 41:217.
  15. Fabry ME, Suzuka SM, Weinberg RS, et al. Second generation knockout sickle mice: the effect of HbF. Blood 2001; 97:410.
  16. Pule GD, Mowla S, Novitzky N, et al. A systematic review of known mechanisms of hydroxyurea-induced fetal hemoglobin for treatment of sickle cell disease. Expert Rev Hematol 2015; 8:669.
  17. Cokic VP, Smith RD, Beleslin-Cokic BB, et al. Hydroxyurea induces fetal hemoglobin by the nitric oxide-dependent activation of soluble guanylyl cyclase. J Clin Invest 2003; 111:231.
  18. Costa FC, da Cunha AF, Fattori A, et al. Gene expression profiles of erythroid precursors characterise several mechanisms of the action of hydroxycarbamide in sickle cell anaemia. Br J Haematol 2007; 136:333.
  19. Flanagan JM, Steward S, Howard TA, et al. Hydroxycarbamide alters erythroid gene expression in children with sickle cell anaemia. Br J Haematol 2012; 157:240.
  20. Zhu J, Chin K, Aerbajinai W, et al. Hydroxyurea-inducible SAR1 gene acts through the Giα/JNK/Jun pathway to regulate γ-globin expression. Blood 2014; 124:1146.
  21. Green NS. A step forward back to (induced) fetal. Blood 2014; 124:993.
  22. Xu J, Zimmer DB. Differential regulation of A gamma and G gamma fetal hemoglobin mRNA levels by hydroxyurea and butyrate. Exp Hematol 1998; 26:265.
  23. Gladwin MT, Shelhamer JH, Ognibene FP, et al. Nitric oxide donor properties of hydroxyurea in patients with sickle cell disease. Br J Haematol 2002; 116:436.
  24. Nahavandi M, Tavakkoli F, Wyche MQ, et al. Nitric oxide and cyclic GMP levels in sickle cell patients receiving hydroxyurea. Br J Haematol 2002; 119:855.
  25. Iyamu EW, Cecil R, Parkin L, et al. Modulation of erythrocyte arginase activity in sickle cell disease patients during hydroxyurea therapy. Br J Haematol 2005; 131:389.
  26. Cokic VP, Beleslin-Cokic BB, Tomic M, et al. Hydroxyurea induces the eNOS-cGMP pathway in endothelial cells. Blood 2006; 108:184.
  27. Pacelli R, Taira J, Cook JA, et al. Hydroxyurea reacts with heme proteins to generate nitric oxide. Lancet 1996; 347:900.
  28. Westerman M, Pizzey A, Hirschman J, et al. Microvesicles in haemoglobinopathies offer insights into mechanisms of hypercoagulability, haemolysis and the effects of therapy. Br J Haematol 2008; 142:126.
  29. Bridges KR, Barabino GD, Brugnara C, et al. A multiparameter analysis of sickle erythrocytes in patients undergoing hydroxyurea therapy. Blood 1996; 88:4701.
  30. Hillery CA, Du MC, Wang WC, Scott JP. Hydroxyurea therapy decreases the in vitro adhesion of sickle erythrocytes to thrombospondin and laminin. Br J Haematol 2000; 109:322.
  31. Bartolucci P, Chaar V, Picot J, et al. Decreased sickle red blood cell adhesion to laminin by hydroxyurea is associated with inhibition of Lu/BCAM protein phosphorylation. Blood 2010; 116:2152.
  32. Orringer EP, Blythe DS, Johnson AE, et al. Effects of hydroxyurea on hemoglobin F and water content in the red blood cells of dogs and of patients with sickle cell anemia. Blood 1991; 78:212.
  33. Steinberg MH, Nagel RL, Brugnara C. Cellular effects of hydroxyurea in Hb SC disease. Br J Haematol 1997; 98:838.
  34. Ware RE. How I use hydroxyurea to treat young patients with sickle cell anemia. Blood 2010; 115:5300.
  35. Kasschau MR, Barabino GA, Bridges KR, Golan DE. Adhesion of sickle neutrophils and erythrocytes to fibronectin. Blood 1996; 87:771.
  36. Benkerrou M, Delarche C, Brahimi L, et al. Hydroxyurea corrects the dysregulated L-selectin expression and increased H(2)O(2) production of polymorphonuclear neutrophils from patients with sickle cell anemia. Blood 2002; 99:2297.
  37. Charache S, Barton FB, Moore RD, et al. Hydroxyurea and sickle cell anemia. Clinical utility of a myelosuppressive "switching" agent. The Multicenter Study of Hydroxyurea in Sickle Cell Anemia. Medicine (Baltimore) 1996; 75:300.
  38. Ware RE, Eggleston B, Redding-Lallinger R, et al. Predictors of fetal hemoglobin response in children with sickle cell anemia receiving hydroxyurea therapy. Blood 2002; 99:10.
  39. Yates AM, Dedeken L, Smeltzer MP, et al. Hydroxyurea treatment of children with hemoglobin SC disease. Pediatr Blood Cancer 2013; 60:323.
  40. Luchtman-Jones L, Pressel S, Hilliard L, et al. Effects of hydroxyurea treatment for patients with hemoglobin SC disease. Am J Hematol 2016; 91:238.
  41. 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.
  42. Wong TE, Brandow AM, Lim W, Lottenberg R. Update on the use of hydroxyurea therapy in sickle cell disease. Blood 2014; 124:3850.
  43. 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; 312:1033.
  44. DeBaun MR, Jordan LC, King AA, et al. American Society of Hematology 2020 guidelines for sickle cell disease: prevention, diagnosis, and treatment of cerebrovascular disease in children and adults. Blood Adv 2020; 4:1554.
  45. http://portal.criticalimpact.com/user/25984/files/ASH_Design_2018-10-09_WEB.pdf (Accessed on January 02, 2019).
  46. https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm590096.htm (Accessed on January 04, 2019).
  47. Steinberg MH, Barton F, Castro O, et al. Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment. JAMA 2003; 289:1645.
  48. 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.
  49. 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.
  50. Lobo CL, Pinto JF, Nascimento EM, et al. The effect of hydroxcarbamide therapy on survival of children with sickle cell disease. Br J Haematol 2013; 161:852.
  51. Lanzkron S, Strouse JJ, Wilson R, et al. Systematic review: Hydroxyurea for the treatment of adults with sickle cell disease. Ann Intern Med 2008; 148:939.
  52. Strouse JJ, Lanzkron S, Beach MC, et al. Hydroxyurea for sickle cell disease: a systematic review for efficacy and toxicity in children. Pediatrics 2008; 122:1332.
  53. Wang WC, Ware RE, Miller ST, et al. Hydroxycarbamide in very young children with sickle-cell anaemia: a multicentre, randomised, controlled trial (BABY HUG). Lancet 2011; 377:1663.
  54. Thornburg CD, Files BA, Luo Z, et al. Impact of hydroxyurea on clinical events in the BABY HUG trial. Blood 2012; 120:4304.
  55. 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.
  56. 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.
  57. 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.
  58. Ghafuri DL, Chaturvedi S, Rodeghier M, et al. Secondary benefit of maintaining normal transcranial Doppler velocities when using hydroxyurea for prevention of severe sickle cell anemia. Pediatr Blood Cancer 2017; 64.
  59. Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med 1995; 332:1317.
  60. Wang W, Brugnara C, Snyder C, et al. The effects of hydroxycarbamide and magnesium on haemoglobin SC disease: results of the multi-centre CHAMPS trial. Br J Haematol 2011; 152:771.
  61. Opoka RO, Ndugwa CM, Latham TS, et al. Novel use Of Hydroxyurea in an African Region with Malaria (NOHARM): a trial for children with sickle cell anemia. Blood 2017; 130:2585.
  62. Tshilolo L, Tomlinson G, Williams TN, et al. Hydroxyurea for Children with Sickle Cell Anemia in Sub-Saharan Africa. N Engl J Med 2019; 380:121.
  63. John CC, Opoka RO, Latham TS, et al. Hydroxyurea Dose Escalation for Sickle Cell Anemia in Sub-Saharan Africa. N Engl J Med 2020; 382:2524.
  64. Thornburg CD, Calatroni A, Panepinto JA. Differences in health-related quality of life in children with sickle cell disease receiving hydroxyurea. J Pediatr Hematol Oncol 2011; 33:251.
  65. Moore RD, Charache S, Terrin ML, et al. Cost-effectiveness of hydroxyurea in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. Am J Hematol 2000; 64:26.
  66. Wang WC, Oyeku SO, Luo Z, et al. Hydroxyurea is associated with lower costs of care of young children with sickle cell anemia. Pediatrics 2013; 132:677.
  67. Steinberg MH. Sickle cell disease and hydroxyurea: the good, the bad, and the future (editorial). Blood 2005; 105:441.
  68. Zumberg MS, Reddy S, Boyette RL, et al. Hydroxyurea therapy for sickle cell disease in community-based practices: a survey of Florida and North Carolina hematologists/oncologists. Am J Hematol 2005; 79:107.
  69. Candrilli SD, O'Brien SH, Ware RE, et al. Hydroxyurea adherence and associated outcomes among Medicaid enrollees with sickle cell disease. Am J Hematol 2011; 86:273.
  70. Haywood C Jr, Beach MC, Bediako S, et al. Examining the characteristics and beliefs of hydroxyurea users and nonusers among adults with sickle cell disease. Am J Hematol 2011; 86:85.
  71. Reeves SL, Jary HK, Gondhi JP, et al. Hydroxyurea use among children with sickle cell anemia. Pediatr Blood Cancer 2019; 66:e27721.
  72. Brousseau DC, Richardson T, Hall M, et al. Hydroxyurea Use for Sickle Cell Disease Among Medicaid-Enrolled Children. Pediatrics 2019; 144.
  73. Stettler N, McKiernan CM, Melin CQ, et al. Proportion of adults with sickle cell anemia and pain crises receiving hydroxyurea. JAMA 2015; 313:1671.
  74. Brandow AM, Panepinto JA. Hydroxyurea use in sickle cell disease: the battle with low prescription rates, poor patient compliance and fears of toxicities. Expert Rev Hematol 2010; 3:255.
  75. https://www.drugs.com/price-guide/siklos (Accessed on September 21, 2020).
  76. McGann PT, Niss O, Dong M, et al. Robust clinical and laboratory response to hydroxyurea using pharmacokinetically guided dosing for young children with sickle cell anemia. Am J Hematol 2019; 94:871.
  77. Kinney TR, Helms RW, O'Branski EE, et al. Safety of hydroxyurea in children with sickle cell anemia: results of the HUG-KIDS study, a phase I/II trial. Pediatric Hydroxyurea Group. Blood 1999; 94:1550.
  78. Heeney MM, Whorton MR, Howard TA, et al. Chemical and functional analysis of hydroxyurea oral solutions. J Pediatr Hematol Oncol 2004; 26:179.
  79. https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm590096.htm (Accessed on April 25, 2019).
  80. Haddy TB, Castro O. Overt iron deficiency in sickle cell disease. Arch Intern Med 1982; 142:1621.
  81. Saraf S, Farooqui M, Infusino G, et al. Standard clinical practice underestimates the role and significance of erythropoietin deficiency in sickle cell disease. Br J Haematol 2011; 153:386.
  82. Vidler JB, Gardner K, Amenyah K, et al. Delayed haemolytic transfusion reaction in adults with sickle cell disease: a 5-year experience. Br J Haematol 2015; 169:746.
  83. Hankins JS, Penkert RR, Lavoie P, et al. Original Research: Parvovirus B19 infection in children with sickle cell disease in the hydroxyurea era. Exp Biol Med (Maywood) 2016; 241:749.
  84. Rana S, Houston PE, Wang WC, et al. Hydroxyurea and growth in young children with sickle cell disease. Pediatrics 2014; 134:465.
  85. Lederman HM, Connolly MA, Kalpatthi R, et al. Immunologic effects of hydroxyurea in sickle cell anemia. Pediatrics 2014; 134:686.
  86. https://www.abbottpointofcare.com/download?docUri=/technical-library/static-assets/technical-documentation/714183-00Y.pdf (Accessed on November 04, 2016).
  87. 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.
  88. DeBaun MR. Hydroxyurea therapy contributes to infertility in adult men with sickle cell disease: a review. Expert Rev Hematol 2014; 7:767.
  89. Smith-Whitley K. Reproductive issues in sickle cell disease. Blood 2014; 124:3538.
  90. Joseph L, Jean C, Manceau S, et al. Effect of hydroxyurea exposure before puberty on sperm parameters in males with sickle cell disease. Blood 2021; 137:826.
  91. Gille AS, Pondarré C, Dalle JH, et al. Hydroxyurea does not affect the spermatogonial pool in prepubertal patients with sickle cell disease. Blood 2021; 137:856.
  92. Berthaut I, Bachir D, Kotti S, et al. Adverse effect of hydroxyurea on spermatogenesis in patients with sickle cell anemia after 6 months of treatment. Blood 2017; 130:2354.
  93. Stukenborg JB, Alves-Lopes JP, Kurek M, et al. Spermatogonial quantity in human prepubertal testicular tissue collected for fertility preservation prior to potentially sterilizing therapy. Hum Reprod 2018; 33:1677.
  94. Pecker LH, Hussain S, Christianson MS, Lanzkron S. Hydroxycarbamide exposure and ovarian reserve in women with sickle cell disease in the Multicenter Study of Hydroxycarbamide. Br J Haematol 2020; 191:880.
  95. Zimmerman SA, Schultz WH, Davis JS, et al. Sustained long-term hematologic efficacy of hydroxyurea at maximum tolerated dose in children with sickle cell disease. Blood 2004; 103:2039.
  96. McGann PT, Howard TA, Flanagan JM, et al. Chromosome damage and repair in children with sickle cell anaemia and long-term hydroxycarbamide exposure. Br J Haematol 2011; 154:134.
  97. Brunson A, Keegan THM, Bang H, et al. Increased risk of leukemia among sickle cell disease patients in California. Blood 2017; 130:1597.
  98. Liem RI, Lanzkron S, D Coates T, et al. American Society of Hematology 2019 guidelines for sickle cell disease: cardiopulmonary and kidney disease. Blood Adv 2019; 3:3867.
  99. Ware RE, Marahatta A, Ware JL, et al. Hydroxyurea Exposure in Lactation: a Pharmacokinetics Study (HELPS). J Pediatr 2020; 222:236.
  100. Asare EV, Olayemi E, Boafor T, et al. Third trimester and early postpartum period of pregnancy have the greatest risk for ACS in women with SCD. Am J Hematol 2019; 94:E328.
Topic 7071 Version 75.0

References