Abstract
Sickle cell disease (SCD) afflicts millions of people worldwide but is referred to as an orphan disease in the United States. Over the past several decades, there has been an increasing understanding of the pathophysiology of SCD and its complications. While most individuals with SCD in resource-rich countries survive into adulthood, the life expectancy of patients with SCD remains substantially shorter than for the general African-American population. SCD can be cured using hematopoietic stem cell transplantation and possibly gene therapy, but these treatment approaches are not available to most patients, the majority of whom reside in low- and middle-income countries. Until relatively recently, only one drug, hydroxyurea, was approved by the US Food and Drug Administration to ameliorate disease severity. Multiple other drugs (L-glutamine, crizanlizumab, and voxelotor) have recently been approved for the treatment of SCD, with several others at various stages of clinical testing. The availability of multiple agents to treat SCD raises questions related to the choice of appropriate drug therapy, combination of multiple agents, and affordability of recently approved products. The enthusiasm for new drug development provides opportunities to involve patients in low- and middle-income nations in the testing of potentially disease-modifying therapies and has the potential to contribute to capacity building in these environments. Demonstration that these agents, alone or in combination, can prevent or decrease end-organ damage would provide additional evidence for the role of drug therapies in improving outcomes in SCD.
Keywords: Sickle cell disease, Treatment, Novel Drugs, Drug Development, Clinical Trials
Introduction
Although referred to as an orphan disease in the United States (US), sickle cell disease (SCD) affects millions of individuals worldwide, with the vast majority residing in sub-Saharan Africa and India 1. SCD is characterized by the presence of sickle hemoglobin (HbS), hemolytic anemia, vaso-occlusive complications, and cumulative end-organ damage. The mortality rate associated with SCD in sub-Saharan Africa remains high, with an estimated 50 to 90% of children dying before the age of 5 2. However, the majority of children with SCD in resource-rich countries live to adulthood 3– 5. Despite increased survival to adulthood, individuals with SCD in resource-rich nations continue to have a shorter life expectancy than the general population 6– 9. There has been substantial progress with the use of allogeneic bone marrow transplantation as a curative therapy in SCD, and increasing evidence supports the curative potential of gene therapy and gene editing 10, 11. However, as these modalities are not available to the vast majority of patients, most of whom reside in resource-limited countries, the availability of drug therapies that are safe, effective, and affordable remains highly desirable.
This review will focus on approaches to develop drug therapies in SCD, ongoing and recently completed trials, and our perspective on the use of approved drugs.
Pathophysiology
The development of effective therapies for SCD depends on an adequate understanding of its pathophysiology. Although the pathophysiology of SCD is complex and involves multiple pathways, the primary event is due to the polymerization of HbS following deoxygenation 12. The rate and extent of polymer formation depends on the degree and duration of HbS deoxygenation, presence of fetal hemoglobin (HbF), and the intracellular concentration of HbS. Clinical manifestations of SCD appear to be driven by two major pathophysiological processes: vaso-occlusion with ischemia-reperfusion injury and hemolytic anemia 13. Vaso-occlusion occurs because of adhesive interactions of leukocytes and sickle RBCs with the endothelium causing microvascular obstruction and subsequent tissue ischemia 13. These episodes of vascular obstruction are followed by the restoration of blood flow, which promotes further tissue injury by reperfusion. The inflammatory cascade resulting from ischemia-reperfusion is amplified by the activation of CD1d-restricted invariant natural killer T (iNKT) cells 14. The release of free plasma hemoglobin following intravascular hemolysis results in direct scavenging of nitric oxide (NO), as well as the generation of reactive oxygen species, powerful scavengers of NO 15, 16. NO is usually produced by the endothelium and regulates basal vasodilator tone as well as inhibits the activation of platelets and the coagulation system and the transcriptional expression of nuclear factor κB (NFκB)-dependent adhesion molecules, such as vascular cell-adhesion molecule-1, intercellular cell-adhesion molecule-1, and selectins 17, 18. HbS polymerization as well as its multiple downstream consequences, including endothelial cell injury, endothelial dysfunction, increased oxidant stress, inflammation, and coagulation and platelet activation, are therapeutic targets in SCD ( Figure 1). SCD has been dichotomized into two overlapping sub-phenotypes: viscosity-vaso-occlusion (higher hemoglobin levels, possibly increased blood viscosity, and complications such as osteonecrosis, acute chest syndrome, and acute pain crisis) and hemolysis-endothelial dysfunction (increased hemolysis with lower hemoglobin levels and higher levels of hemolytic markers, including reticulocyte count and serum lactate dehydrogenase, and complications such as leg ulcers, priapism, stroke, and possibly pulmonary hypertension) 19. While somewhat simplistic, this classification is useful to understand the pathobiology of disease complications and may provide guidance on the effects of therapies on disease-related complications. The pathophysiology of SCD has been reviewed more extensively elsewhere 20– 22.
Drug therapies for sickle cell disease
Despite an improved understanding of the pathophysiology of SCD, available drug treatments remain limited. For many years, hydroxyurea was the only drug available to modify the severity of SCD 23. However, there has been progress in the development of drug therapies with recent approvals of L-glutamine, crizanlizumab, and voxelotor by the US Food and Drug Administration (FDA) 24– 26. Based on our current understanding of SCD pathophysiology, there are multiple possible approaches to treating the disease and its complications, including inhibition of HbS polymerization and amelioration of multiple downstream sequelae of HbS polymerization.
Inhibition of sickle hemoglobin polymerization
Therapeutic approaches targeting HbS polymerization, including 1) blocking intermolecular contacts in the sickle fiber, 2) induction of HbF synthesis, 3) reduction of intracellular HbS concentration, 4) increase in oxygen affinity, and 5) reduction of the concentration of 2,3-diphosphoglycerate, have been described 27. Table 1 lists ongoing studies of anti-sickling agents in SCD.
Table 1. Ongoing clinical trials of anti-sickling agents in sickle cell disease.
Mechanism | Drug | Sponsor | NCT number
(study acronym) |
Clinical phase/status | Intervention | Number/age | Objective |
---|---|---|---|---|---|---|---|
HbF induction | Nicotinamide vs. THU
and decitabine |
EpiDestiny, Inc;
NIH; NHLBI |
NCT04055818 | Phase I
Recruiting |
Oral nicotinamide vs. THU
+ decitabine for 12 weeks followed by combination for a further 12 weeks |
20/≥18 years | Compare effect of oral nicotinamide
vs. THU-decitabine and in combination on hemoglobin level at week 12 |
Panobinostat
(LBH589) |
Abdullah Kutlar | NCT01245179 | Phase I
Active, not recruiting |
Oral, for 12 weeks | 18/≥18 years | Evaluate safety, HbF, F cells,
total hemoglobin, markers of inflammation, QOL measures |
|
Metformin | Baylor College of
Medicine |
NCT02981329
(FITMet) |
Phase I
Recruiting |
Hydroxyurea + metformin
vs. metformin |
56/10–60 years | Change in HbF or total hemoglobin,
QOL, RNA sequencing |
|
Allosteric
modifier (to the R-state) |
Voxelotor (formerly
GBT440) |
Global blood
therapeutics |
NCT03943615
(expanded access) |
Approved for
marketing |
Oral | ≥12 years | To provide early access to patients
without alternative treatment options |
NCT04247594 | Phase II
Recruiting |
Oral, open-label | 45/18–60 years | Dose escalation study to evaluate
safety and tolerability of doses, 1,500 mg to 3,000 mg daily |
|||
NCT02850406 | Phase IIa
Recruiting |
Part A: single dose Part B:
24 weeks Part C: 48 weeks |
125/4–17 years | Pharmacokinetics, change in
hemoglobin, effect on hemolysis, TCD velocity, safety |
|||
NCT03573882 | Phase III
Active, not recruiting |
Oral, daily | 179/≥12 years | Open-label extension study,
adverse events, frequency of SCD- related complications |
|||
NCT04218084
(HOPE kids 2) |
Phase III
Not yet recruiting |
Oral voxelotor vs. placebo | 224/2–14 years | Evaluate effect on TCD in children | |||
NCT04188509 | Phase III
Enrolling by invitation |
Oral, open-label | 50/4–18 years | Evaluate safety and tolerability,
SCD-related complications |
|||
Allosteric
activator of RBC pyruvate kinase-R |
AG-348 (Mitapivat
sulfate) |
NHLBI | NCT04000165 | Phase I
Recruiting |
Oral, with two dose
escalations after 2 weeks |
25/≥18 years | Safety and tolerability,
pharmacokinetics, and pharmacodynamics |
FT-4202 | Forma
therapeutics |
NCT03815695 | Phase I
Recruiting |
Single and multiple
ascending oral doses of FT-4202 vs. placebo |
130/12–60
years |
Safety, pharmacokinetics,
pharmacodynamics |
|
RBC ion
transport channels |
SCD-101 | Invenux | NCT02380079 | Phase Ib
Recruiting |
Part A: open-label, dose-
escalation study Part B: randomized, placebo-controlled, cross- over study |
60/18–55 years | Safety, change in hemoglobin,
markers of hemolysis, QOL measures, functional capacity |
Memantine
(NMDAR antagonist) |
HaEmek
Medical Center, Israel |
NCT03247218 | Phase IIa/b
Recruiting |
Oral, once daily for 1 year | 40/≥10years | Safety, frequency of
hospitalizations, duration of hospitalizations, analgesic use, transfusion requirement, QOL measures |
HbF, fetal hemoglobin; QOL, quality of life; NHLBI, National Heart, Lung, and Blood Institute; NMDAR, N-methyl-D-aspartate receptor; RBC, red blood cell; SCD, sickle cell disease; TCD, transcranial doppler; THU, tetrahydrouridine; VOC, vaso-occlusive crisis.
Induction of fetal hemoglobin. High levels of HbF ameliorate the severity of SCD 8, 28, 29. Hydroxyurea strongly induces HbF 30, although the mechanisms by which it does so are still unclear 30– 33. Hydroxyurea improves erythrocyte deformability, lowers hemolysis, decreases circulating leukocytes and reticulocytes 34, reduces adhesion receptor expression 35– 38, and is an NO donor 39, 40. In two placebo-controlled, phase III trials, hydroxyurea significantly reduced the frequency of vaso-occlusive crises, acute chest syndrome, blood transfusion requirement, and hospitalizations in adults and children with SCD 23, 41. Treatment with low, fixed weight-based dosing of hydroxyurea (10 mg/kg daily) also decreased the frequency of SCD-related complications with low toxicity 42, 43. Hydroxyurea further ameliorates SCD morbidity by decreasing the risk of stroke and chronic kidney disease 44, 45 and may reduce mortality in SCD 46– 49. Recent studies to optimize hydroxyurea therapy are exploring dose maximization via pharmacokinetics-based dosing 50– 54. Despite its multiple benefits, a substantial number of patients on hydroxyurea may not obtain an adequate clinical response.
Inhibitors of epigenetic enzymes which repress γ-globin genes, including DNA methyltransferase (DNMT1), histone deacetylases, and lysine-specific demethylase (LSD-1), and γ-globin gene co-activator agonists are being evaluated in SCD. Decitabine (5-aza-2’-deoxycytidine), a DNA hypomethylating agent which depletes DNMT1, increased HbF and total hemoglobin levels at non-cytotoxic doses in patients who had no responses to hydroxyurea 55. However, it has a very short plasma half-life and negligible oral bioavailability due to rapid inactivation by cytidine deaminase (CDA) 55. The combination of escalating doses of oral decitabine and an oral CDA inhibitor, tetrahydrouridine (THU), was well tolerated, and the highest tested dose increased HbF, doubled F-cells by approximately 80% of total RBCs, increased total hemoglobin, and decreased biomarkers of hemolysis, coagulation activation, and inflammation 56.
Histone deacetylase (HDAC) inhibitors increase HbF levels 57– 63. Although treatment with intermittent doses of sodium butyrate produced sustained increases in HbF, F-cells, and total hemoglobin 57, the orally available sodium dimethyl butyrate (HQK-1001) did not significantly increase HbF and appeared to result in more pain crisis compared with placebo 63. A phase II study of the global HDAC inhibitor vorinostat was terminated early owing to poor accrual. Selective HDAC1/2 inhibition with ACY-957 increased HbF in vitro and represents a promising therapeutic approach with a better safety profile 64. Based on the success of the LSD-1 inhibitors RN-1 and ORY-3001 in increasing HbF and F-cells in preclinical studies 65– 67, INCB059872 was investigated (NCT03132324). However, this phase I study was terminated early for business reasons. Pomalidomide, a third-generation immunomodulatory drug, produced modest increases of HbF, augmented erythropoiesis, and preserved bone marrow function following 8 weeks of treatment in transgenic sickle mice 68. Pomalidomide appeared to increase HbF and total hemoglobin only at the highest dose or with greater than 6 months of exposure 69. The safety and efficacy of increasing the expression of γ-globin gene co-activators—FOXO3 agonist 70 (metformin [NCT02983129]), nuclear factor-like 2 (Nrf2) agonist 71 (dimethyl fumarate), and SIRT1 72—to augment HbF levels are being explored.
Allosteric modification of sickle hemoglobin to its high-oxygen affinity R-state. Voxelotor, recently approved under the FDA’s accelerated approval program, is an orally bioavailable small molecule which binds to α-globin chains of hemoglobin, increases hemoglobin oxygen affinity, and stabilizes the oxyhemoglobin state 73. Early studies showed a reduction in markers of hemolysis, with a rapid rise in hemoglobin by day 15 74. This response was not accompanied by worsening tissue hypoxia, based on serum erythropoietin levels and change in oxygen consumption (VO 2 max), even in participants receiving daily doses of 1,000 mg for 28 days and 900 mg for at least 90 days. Furthermore, voxelotor has been shown to reduce blood viscosity in vitro 75. In a multicenter, phase III study of patients 12 to 65 years of age, randomly assigned to once-daily oral voxelotor doses of 1,500 mg, 900 mg, or placebo, a significantly higher percentage of subjects on 1,500 mg had a hemoglobin response of 1.0 g/dL at 24 weeks compared with placebo 76. In addition, subjects on the 1,500 mg dose saw significant reductions in indirect bilirubin and reticulocyte counts from baseline. A dose-dependent increase in hemoglobin following treatment with voxelotor was not accompanied by an increase in pain crisis rate. As this trial was not enriched for subjects with frequent pain crises, appropriately designed studies of patients with frequent pain episodes are needed to determine the effect of voxelotor on reducing such crises. Open-label extension studies to assess the long-term effects of voxelotor are ongoing.
5-hydroxymethyl-2-furfural (5HMF, Aes-103) interacts allosterically with HbS, increasing oxygen affinity and decreasing HbS polymerization and RBC sickling during hypoxia. A phase I trial, with oral doses of up to 4,000 mg, showed no significant side effects 77, but a phase II trial was terminated early because of unblinding of drug groups (NCT01987908). FT-4202, a novel selective activator of RBC pyruvate kinase, decreases intracellular 2,3-diphosphoglycerate levels, with a resulting increase in hemoglobin–oxygen affinity. It demonstrated a favorable safety profile in healthy individuals 78 and is being investigated in a phase I study in SCD (NCT03815695).
The oxygen delivery therapeutic agents sanguinate (pegylated bovine carboxyhemoglobin) and MP4CO (pegylated human carboxyhemoglobin) are dual transfer agents which release carbon monoxide when delivering oxygen to hypoxic tissues. They stabilize HbS in its R-state and appear to exhibit anti-inflammatory and anti-apoptotic activity by induction of Nrf2 and heme oxygenase-1 79, 80. Sanguinate had an acceptable safety profile 81, 82. A phase II study evaluating its efficacy during acute pain crisis was recently completed (NCT02411708).
Increase red blood cell hydration. The inverse relationship between HbS concentration and delay time suggests that even small decreases in intracellular HbS might be beneficial 83. Senicapoc selectively blocks the calcium-activated, potassium efflux (Gardos) channel and improves anemia and hemolysis in SCD 84, 85. Despite improvements in anemia and hemolysis, a phase III trial showed no significant decrease in the rate of pain crises compared to placebo 84. Similarly, studies of agents that block the potassium-chloride co-transport channel showed no clinical benefits 86– 88. SCD101, a botanical drug with an unclear anti-sickling mechanism which might involve dilution of HbS by affecting the RBC membrane, was well tolerated in a phase Ib study, with a decrease in chronic pain and fatigue and improvement in leg ulcers 89.
Targeting downstream sequelae of sickle hemoglobin polymerization
Antioxidant therapy. Agents that upregulate antioxidant and/or reactive oxygen species scavenging processes have been evaluated in SCD. Glutamine, a conditionally essential amino acid, is a precursor for nicotinamide adenine dinucleotide (NAD) and improves NAD redox potential. In a randomized, multicenter trial of 230 patients with HbSS or HbSβ 0-thalassemia, L-glutamine significantly reduced the number of pain crises, hospitalizations, cumulative hospital days, and frequency of acute chest syndrome compared with placebo 90. However, the effect size of L-glutamine in reducing pain crises was relatively small, with only a 25% reduction in the median number of pain crises. L-glutamine was well tolerated, although low-grade nausea, non-cardiac chest pain, fatigue, and musculoskeletal pain occurred more frequently than with placebo. Despite some concerns related to the high dropout rate in the trial (36% L-glutamine arm; 24% placebo arm), L-glutamine was approved by the FDA to reduce the acute complications of SCD in patients 5 years and older 24.
In an open-label pilot trial of oral N-acetylcysteine (NAC), treatment with either 1,200 mg or 2,400 mg daily for 6 weeks increased whole blood glutathione levels and decreased erythrocyte outer membrane phosphatidylserine exposure, plasma levels of advanced glycation products, and cell-free hemoglobin in both groups 91. A randomized, placebo-controlled, double-blind trial of NAC at 600 mg twice daily for 6 months did not decrease the rate of SCD-related pain days per patient year, vaso-occlusive crises, hospital admission days, number of admissions, or days with home analgesic use compared with placebo 92. The safety and efficacy of NAC, administered at a higher dose during pain crisis, is being explored in a phase I/II study (NCT01800526) ( Table 2).
Table 2. Ongoing clinical trials of anti-adhesion agents and antioxidants in sickle cell disease.
Mechanism | Drug | Sponsor | NCT number
(study acronym) |
Clinical phase/status | Intervention | Number/age | Objective |
---|---|---|---|---|---|---|---|
P-selectin
antagonist |
Crizanlizumab | Novartis
Pharmaceuticals |
NCT03264989
(SOLACE- adults) |
Phase II
Active, not recruiting |
IV infusion, open-label | 57/16–70 years | Pharmacokinetics,
pharmacodynamics, safety, and efficacy |
NCT03814746
(STAND) |
Phase III
Recruiting |
IV infusion every 2 weeks for
1 st month and then monthly for 1 year |
240/≥12 years | Compare efficacy and safety of 5
mg/kg and 7.5 mg/kg doses with placebo |
|||
NCT04053764
(STEADFAST) |
Phase II
Recruiting |
IV infusion every 2 weeks for
1 st month and then every 4 weeks for 51 weeks + SoC vs. SoC alone |
170/≥16 years | Evaluating effect on kidney
function (albumin-creatinine ratio, protein-creatinine ratio, estimated glomerular filtration rate) |
|||
NCT03938454
(SPARTAN) |
Phase II
Recruiting |
IV infusion every 2 weeks for
1 st month and then every 4 weeks x 51 weeks |
56/≥16 years | Evaluate efficacy in priapism,
uncomplicated VOC events |
|||
NCT03474965 | Phase II
Recruiting |
IV infusion every 2 weeks for
1 st month and then every 4 weeks |
100/6 months–<18
years |
Evaluate pharmacokinetics,
pharmacodynamics, safety, and effect on VOC events |
|||
Blockade of fcγrIII
receptors |
IVIG | Albert Einstein
College of Medicine |
NCT01757418 | Phase I–II
Recruiting |
Single dose of IVIG vs.
placebo given within 24 hours of hospitalization |
94/12–65 years | Length of VOC, total opioid
use, time to end of VOC, in vitro adhesion studies |
Antioxidant
(increased glutathione) |
NAC | Bloodworks | NCT01800526 | Phase I/II
Enrolling by invitation |
IV or oral, NAC
Part 1: two doses of IV infusion over 8 hours 4 weeks apart or oral NAC for 4 weeks Part 2 (during VOC): IV infusion every 6 hours for 5 days |
20/≥18 years | Evaluate effect on vWF activity,
measures of hemolysis and oxidation In Part 2, evaluate efficacy during VOC |
IV, intravenous; IVIG, intravenous gammaglobulin; NAC, N-acetylcysteine; SoC, standard of care; VOC, vaso-occlusive crisis; vWF activity, von Willebrand factor activity.
Anti-adhesive therapy. Agents targeting adhesion of blood cells to the endothelium have been investigated in SCD. Crizanlizumab, a humanized monoclonal anti-P-selectin antibody, was recently approved for use in patients 16 years and older for the prevention of vaso-occlusive crises 25 based on results of a randomized, double-blind, phase II study which evaluated the benefit of 2.5 mg/kg or 5 mg/kg doses versus placebo. Significantly lower median crisis rate, longer median times to first and second crises, and lower median rate of uncomplicated crises per year were observed with high-dose crizanlizumab compared with placebo following a 52-week treatment period 93. Multiple other studies of crizanlizumab are ongoing ( Table 2).
Purified poloxamer 188, a nonionic block copolymer surfactant with hemorheologic and antithrombotic properties, was previously shown to significantly decrease the duration of pain episodes, especially in children and patients on hydroxyurea 94. However, a more recent phase III study in children and adults reported no significant effect of purified poloxamer 188 (vepoloxamer or MST-188) on duration of vaso-occlusive crises compared with placebo 95. Rivipansel sodium (formerly GMI-1070) is a small-molecule pan-selectin inhibitor that binds to E-, P-, and L-selectin 96. In a randomized, double-blind, adaptive, phase II trial, treatment with rivipansel during pain crisis produced a significant reduction in the mean cumulative intravenous opioid analgesic use compared to placebo 97. However, despite the promising phase II trial results, the recently completed multicenter phase III RESET trial failed to meet its primary (time to readiness for discharge) and key secondary (time to discharge, cumulative intravenous opioid utilization, and time to discontinuation of intravenous opioids) efficacy endpoints 98.
Heparins inhibit adhesive interactions via P-selectin 99, 100. Tinzaparin, a low-molecular-weight (LMW) heparin, at therapeutic dose reportedly decreased the number of hospital days, the number of days with pain crisis, and the number of days with the most severe pain scores compared with placebo 101. Sevuparin, a derivative of LMW heparin, which retains the P-selectin-binding domain of heparin but largely lacks anticoagulant properties, binds to P- and L-selectins, thrombospondin, fibronectin, and von Willebrand factor, inhibits the adhesion of sickle RBCs to stimulated cultured endothelial cells in vitro, and prevents vaso-occlusion with normalization of blood flow in a mouse model of vaso-occlusion 102. A recently completed randomized, double-blind, placebo-controlled trial found that, although safe, sevuparin did not significantly reduce the time to resolution of vaso-occlusive crisis 103. A phase II feasibility study of therapeutic-dose unfractionated heparin (NCT02098993) was recently terminated for poor enrollment, but a phase III, randomized, placebo-controlled study of tinzaparin in acute chest syndrome is ongoing (NCT02580773).
Intravenous immunoglobulin (IVIG) binds to FcγRIII receptors, inhibits neutrophil adhesion to the endothelium, reduces RBC capture by leukocytes, and reduces Mac-1 activity due to recruitment of Src homology 2-containing tyrosine phosphatase-1 104. IVIG was tolerated at doses of up to 800 mg/kg during acute vaso-occlusive crises and decreased Mac-1 function from baseline 105. A phase II clinical trial of IVIG during acute pain episodes is ongoing (NCT01757418).
Anti-inflammatory agents. Multiple approaches to downregulate inflammatory pathways have been evaluated. Initial clinical responses with the use of steroids for pain crisis and acute chest syndrome were followed by “rebound” pain episodes and re-admissions after treatment discontinuation 106, 107. Inhaled corticosteroids did not significantly reduce the morbidity of acute chest syndrome in a retrospective cohort study 108, but inhaled mometasone significantly reduced daily pain diary scores and levels of soluble vascular adhesion molecule-1 over the 16-week treatment period in another single-center, placebo-controlled trial 109.
The activation of iNKT cells is downregulated by the activation of adenosine A2A receptors (A 2AR) 110. Regadenoson, a partially selective A 2AR agonist, was reportedly safe but did not decrease length of hospital stay, total opioid use, or pain scores compared to placebo during pain crises 111, 112. In addition, there were no significant differences between regadenoson and placebo regarding the number of patients exhibiting a greater than 30% reduction in activated iNKT cells. The anti-iNKT cell monoclonal antibody NKTT120 resulted in rapid, specific, and continuous iNKT cell depletion in a single-ascending-dose study 113, but further studies are required to determine its long-term safety and efficacy.
Statins have benefits independent of their cholesterol-lowering effect. In a pilot study, simvastatin was well tolerated, increased levels of NO metabolites, and decreased C-reactive protein (CRP) and interleukin-6 levels 114. Daily treatment with simvastatin for 3 months significantly reduced pain crises, oral analgesic use, and levels of high-sensitivity CRP, soluble E-selectin, soluble ICAM-1, soluble ICAM-3, and VEGF 115, providing supporting data for conduct of a placebo-controlled trial. Atorvastatin was well tolerated in a pilot study but did not improve endothelial function or decrease albuminuria 116.
Long-chain omega-3 polyunsaturated fatty acids such as eicosapentaenoic acid (EPA [n-3]) and docosahexaenoic acid (DHA [n-6]) provide benefit to individuals with chronic inflammatory disorders. In a randomized, single-center study, daily omega-3 administered in capsule form significantly lowered the occurrence of vaso-occlusive events, severe anemia, blood transfusion requirements, and school absence compared with placebo 117. More recently, a randomized, placebo-controlled study of different doses of SC411, a novel DHA ethyl ester formulation with high DHA bioavailability, found it was well tolerated, with significant reductions in d-dimer and E-selectin and an increase in hemoglobin 118. SC411 also significantly reduced electronic diary-recorded pain episodes, analgesic use at home, and days absent from school but did not significantly lower pain crises in the pooled active groups compared to placebo.
Preliminary results suggest that the monoclonal anti-IL1β antibody canakinumab (ACZ885) is well tolerated and not associated with major side effects in SCD 119.
Anticoagulant and antiplatelet therapies. With abundant evidence that SCD is a hypercoagulable state 21, combined with data that coagulation and platelet activation may play roles in disease pathophysiology 120– 124, multiple studies have evaluated the effects of anticoagulants and antiplatelet agents in SCD. Although treatment with the LMW heparin tinzaparin produced more rapid resolution of pain crisis and shorter duration of hospitalization than placebo 101, it is uncertain whether the reported beneficial effects were a result of its anticoagulant or anti-adhesive effects. The results of a pilot study of the direct oral anticoagulant rivaroxaban in SCD are awaited (NCT02072668).
Multiple relatively small studies of aspirin have reported modest benefit at best 125– 127, although one study reported an increase in oxygen affinity, hemoglobin level, and RBC lifespan 126. A pilot study of the glycoprotein IIb/IIIa inhibitor eptifibatide showed that it was safe during acute pain episodes but did not reduce the time to resolution of such episodes 128. Ticlopidine, a P2Y 12 ADP-receptor antagonist, reportedly decreased the number of pain episodes, mean duration of pain episodes, and severity of such episodes 129. However, a more recent phase III trial of prasugrel, a newer generation P2Y 12 receptor blocker, showed no significant reduction of the frequency of pain episodes in children with SCD 130. Ticagrelor, a reversible P2Y 12 receptor blocker, was well tolerated in children 131 and young adults 132 but had no beneficial effects on pain. A phase III study to determine the efficacy of ticagrelor in reducing vaso-occlusive crises in children is ongoing 133 ( Table 3).
Table 3. Ongoing clinical trials of anticoagulants, antiplatelet agents, anti-inflammatory agents, and nitric oxide-related drugs in sickle cell disease.
Mechanism | Drug | Sponsor | NCT number
(study acronym) |
Clinical phase/
status |
Intervention | Number/age | Objective |
---|---|---|---|---|---|---|---|
Anticoagulant,
anti-adhesive (P- selectin blocker) |
Tinzaparin
(low-molecular-weight heparin) |
Assistance Publique
–Hopitaux de Paris, LEO pharma |
NCT02580773
(TASC) |
Phase III
Unknown status |
Subcutaneous therapeutic vs.
prophylactic dose for 7 days |
200/≥18 years
|
Time to ACS resolution, bleeding
events, hospital mortality |
Antiplatelet | Ticagrelor | AstraZeneca |
NCT03615924
(HESTIA3) |
Phase III
Active, not recruiting |
Oral ticagrelor vs. placebo for
at least 12 months |
193/2–17 years | Evaluate efficacy in decreasing VOC,
acute chest syndrome, duration of VOC, QOL measures |
Antithrombotic,
anti-inflammatory |
Defibrotide | New York Medical
College |
NCT03805581 | Phase II Recruiting | Intravenous, every 6 hours
during ACS event for maximum of 7 days |
20/2–40 years | Safety (allergic reaction, bleeding),
improvement in clinical signs of ACS |
Anti-inflammatory | Mometasone | Jeffery Glassberg |
NCT03758950
(IMPROVE2) |
Phase II
Recruiting |
Inhaled, one puff daily vs.
placebo for 48 weeks |
80/≥18 years | Change in soluble VCAM-1,
hemoglobin, markers of hemolysis, leukocyte count, QOL measures |
Canakinumab
(anti-IL1β antibody) |
Novartis | NCT02961218 | Phase II
Active, not recruiting |
Subcutaneous, monthly for 6
months vs. placebo, followed by 6-month open-label extension |
49/8–20 years | Reduction of average daily pain, days
missed from school, blood counts, pharmacokinetics, hs-CRP, measures of hemolysis |
|
Docosahexaenoic acid
and eicosapentaenoic acid |
Thomas Jefferson
University |
NCT01202812 | Phase II
Not yet recruiting |
Oral, daily for 6 months vs.
placebo |
48/10–19 years | Decrease inflammatory biomarkers,
QOL |
|
Docosahexaenoic acid
(SC411) |
Micelle BioPharma |
NCT02973360
(SCOT study) |
Phase II
Active, not recruiting |
Oral for 8 weeks vs. placebo
followed by 49-month open- label extension |
68/5–17 years | Safety, pharmacokinetics, effect on
VOC |
|
Omega-3-fatty acid | Sultan Qaboos
University |
NCT02525107 | Phase III
Recruitment status unknown |
Oral, daily for 52 weeks vs.
placebo |
280/13–70 years | Effect on VOC, duration of
hospitalization, RBC membrane fatty acid profile |
|
Rifaximin (antibiotic,
decrease aged neutrophils) |
New York Medical
College |
NCT03719729 | Phase II
Recruiting |
Oral, twice daily for 1 year | 20/18–70 years | Safety, frequency of hospitalizations for
VOCs, duration of hospitalizations, RBC transfusions, QOL measures |
|
Increased NO production |
Arginine |
Emory University |
NCT02447874 | Phase I/II
Enrolling by invitation |
Intravenous, three times a day
for maximum of 7 days |
21/7–21 years | Pharmacokinetics, NO metabolites |
NCT02536170 | Phase II
Recruiting |
Intravenous, three times
a day through duration of hospitalization |
114/3–21 years | Effect on VOC, including total
parenteral opioid use, length of hospital stay, resolution of VOC |
|||
Vasodilator | Olinciguat (IW-1701)
(soluble guanylate cyclase stimulator) |
Cyclerion
therapeutics |
NCT03285178 | Phase II
Recruiting |
Oral, daily 12 weeks vs.
placebo |
88/16–70 years | Safety and tolerability,
pharmacokinetics, pharmacodynamics |
Riociguat (soluble
guanylate cyclase stimulator) |
Mark Gladwin | NCT02633397 | Phase II
Recruiting |
Oral, three times daily for 12
weeks |
100/≥18 years | Safety, changes in pain intensity,
SCD-related complications, functional capacity, blood pressure, plasma NT-proBNP, changes in laboratory measures, TRV |
|
IMR-687 (selective
phosphodiesterase-9 inhibitor) |
Imara | NCT03401112 | Phase IIa
Recruiting |
Oral, daily for 24 weeks vs.
placebo |
70/18–55 years | Safety, pharmacokinetics | |
NCT04053803
|
Phase II
Enrolling by invitation |
Oral, daily for 49 months,
open-label extension |
70/≥18 years
|
Safety, change in hemoglobin, HbF,
soluble adhesion markers, renal and cardiac function, QOL |
|||
Ambrisentan
(endothelin receptor A antagonist) |
Augusta University | NCT02712346 | Phase I
Active, not recruiting |
Oral, once daily for 12 weeks | 26/18–65 years | Safety and tolerability,
microalbuminuria/proteinuria, TRV, inflammatory markers, pain questionnaire |
ACS, acute chest syndrome; HbF, fetal hemoglobin; hs-CRP, high-sensitivity C-reactive protein; NO, nitric oxide; NT-proBNP, N-terminal probrain natriuretic peptide; QOL, quality of life; TRV, echocardiography-derived tricuspid regurgitant jet velocity; VCAM-1, vascular cell adhesion molecule-1; VOC, vaso-occlusive crisis; vWF, von Willebrand factor.
Nitric oxide and related agents. With the role of hemolysis in NO scavenging and subsequent endothelial dysfunction, NO and related agents may be beneficial in SCD 134. Inhaled NO did not improve time to resolution of pain crisis, length of hospitalization, opioid usage, or rate of acute chest syndrome compared with placebo 135. In another study, inhaled NO did not reduce the rate of treatment failure in adult patients with mild to moderate acute chest syndrome 136. However, L-arginine, an NO precursor, significantly decreased total parenteral opioid use and pain scores at discharge compared to placebo in children, although there was no difference in length of hospital stay 137. Furthermore, supplementation of adult patients on hydroxyurea with oral arginine significantly increased the level of NO metabolites and reduced the frequency of pain crises versus placebo following 4 months of treatment 138. Sildenafil, a phosphodiesterase-5 inhibitor which increases NO-mediated effects by inhibiting the degradation of cyclic guanosine monophosphate (cGMP), was associated with more frequent serious adverse events, predominantly hospitalization for pain, compared with placebo 139. No evidence of a treatment effect of sildenafil was seen on evaluated study outcomes. Studies of soluble guanylate cycle stimulators, riociguat (NCT02633397) and olinciguat/IW-1701 (NCT03285178), and the phosphodiesterase-9 inhibitor IMR-687 (NCT03401112; NCT04053803) are ongoing ( Table 3).
Summary
Recent advances in drug development for SCD raise important issues related to the use and availability of these agents. With limited availability of allogeneic bone marrow transplantation and gene therapy, especially in low- and middle-income countries, the number of recently approved drugs offers hope for further improved outcomes in SCD. Choice of initial drug therapy may be guided by a patient’s clinical sub-phenotype as well as the cost of the drug ( Table 4). Patients with frequent vaso-occlusive complications (acute pain episodes, acute chest syndrome) may benefit from hydroxyurea, L-glutamine, and crizanlizumab, while those with increased hemolytic anemia (or quite possibly complications related to hemolytic anemia) may benefit from hydroxyurea and voxelotor. However, more studies are required before definite recommendations can be made regarding the effect of voxelotor on SCD-related complications. As with other chronic disease conditions, it is important to consider the cost of the drug when deciding on initial therapy. Hydroxyurea is much cheaper than L-glutamine, crizanlizumab, and voxelotor and is likely more cost effective as initial therapy for vaso-occlusive complications and hemolytic anemia. However, with the complex pathophysiology of SCD and the limited clinical efficacy of available pharmacological therapies, it is unlikely that a single drug will ameliorate all SCD-related complications. As such, the availability of multiple drugs offers an opportunity for combination therapy based on different mechanisms of action and non-overlapping side effect profiles. Although more studies are required to evaluate the effect of combinations of these drugs, all of the recently approved agents were tested in combination with hydroxyurea. These studies showed benefit whether or not participants received concomitant hydroxyurea, with no increased toxicity. Despite potential benefits, combination therapy involving the use of multiple drugs increases the likelihood of decreased medication adherence owing to polypharmacy. Like in other chronic diseases, however, this problem may be somewhat ameliorated by the development of pills that contain more than one active drug.
Table 4. Summary characteristics of FDA-approved drugs for sickle cell disease.
Hydroxyurea | L-Glutamine | Crizanlizumab | Voxelotor | |
---|---|---|---|---|
Age (years) | ≥2 | ≥5 | ≥16 | ≥12 |
Genotypes | HbSS, HbSβ 0 thalassemia | All genotypes (only
studied in HbSS, HbSβ 0 thalassemia) |
All genotypes | All genotypes |
Mechanism of action | Multiple, but primarily by
increasing HbF production |
Uncertain, but thought
to reduce NAD redox potential, possible decrease in cell adhesion |
Anti P-selectin inhibitor
(decreases adhesion of WBCs and RBCs to endothelium) |
Decreases HbS
polymerization by increasing Hb–oxygen affinity |
Route of administration | Oral (capsules/tablets) | Oral (powder) | Intravenous | Oral (tablets) |
Clinical effects
of therapy |
Decreased frequency of VOC,
decreased frequency of ACS, decreased hospitalization, decreased RBC transfusion requirement, decreased stroke risk |
Decreased frequency
of VOC, decreased frequency of ACS, decreased hospitalization |
Decreased frequency
of VOC |
Increased hemoglobin |
Effect size for primary
endpoint (% change and IRR) |
44% decrease in VOC per year
(median from 4.5 to 2.5), IRR = 0.56 |
25% decrease in VOC in
48 weeks (median from 4 to 3), IRR = 0.75 |
45% decrease in crisis
rate per year (median from 3 to 1.6), IRR = 0.55 |
5.5-fold increase in the
hemoglobin responders (9% to 59%) at 24 weeks, incidence proportion ratio = 6.6 * |
Common toxicities | Myelosuppression, skin
hyperpigmentation, nail discoloration, teratogenicity, decreased sperm counts, nausea and vomiting |
Constipation, nausea,
headaches, abdominal pain |
Nausea, arthralgia | Headache, diarrhea,
nausea |
Pharmacokinetics | Excreted via kidneys Adjust dose
for eGFR <60 mL/min/1.73 m 2 |
Use with caution with
hepatic and renal impairment, but no recommended dose adjustment |
No dosage
adjustments in manufacturer labeling for renal and hepatic impairment (not tested in ESRD) |
No dosage adjustment
for renal impairment, but not yet studied in ESRD requiring dialysis Dose reduction for severe liver disease (Child Pugh class C) |
Cost | $ | $$$ | $$$$$ | $$$$$ |
ACS, acute chest syndrome; eGFR, estimated glomerular filtration rate; ESRD, end-stage renal disease; FDA, US Food and Drug Administration; Hb, hemoglobin; HbF, fetal hemoglobin; HbS, sickle hemoglobin; IRR, incidence rate ratio; NAD, nicotinamide adenine dinucleotide; RBC, red blood cell; VOC, vaso-occlusive crisis; WBC, white blood cell.
Patients treated with 1,500 mg of voxelotor had 6.6 times increased proportion of hemoglobin responders (>1 g/dL increase from baseline at 24 weeks)
It is particularly important that ongoing trials of novel drugs for SCD involve individuals in resource-limited countries where the burden of disease is high. This offers patients access to potentially disease-modifying therapies but, perhaps equally as important, contributes substantially to capacity building by improving local infrastructure, strengthening health systems, involving local leaders as collaborators, and training local providers. Furthermore, trials of novel agents in sub-Saharan Africa provide data on the effects of these therapies in a setting with fewer resources and unique challenges, including a high prevalence of malnutrition and infections such as malaria. With the high cost of recently approved agents, mechanisms to ensure affordable access to these drugs should be established. A situation where novel agents are tested in resource-limited countries but patients in these countries are unable to afford such drugs following approval is unacceptable. Finally, studies evaluating the effects of recently approved drugs on organ damage are needed. Demonstration that these agents prevent or decrease end-organ damage would provide additional evidence for the role of drug therapies in improving outcomes in SCD.
Acknowledgements
The authors thank Dr. Joacy Gerard Matthias and Dr. Matthew P. Smeltzer (Both from the Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, Tennessee, USA) for helping to ascertain the drug effect sizes.
Editorial Note on the Review Process
F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).
The referees who approved this article are:
Yogen Saunthararajah, Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
Daniel E. Bauer, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
Funding Statement
This work is supported by an award from the US Food and Drug Administration - R01FD006030 (KIA).
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[version 1; peer review: 2 approved]
References
- 1. Piel FB, Hay SI, Gupta S, et al. : Global burden of sickle cell anaemia in children under five, 2010-2050: Modelling based on demographics, excess mortality, and interventions. PLoS Med. 2013;10(7):e1001484. 10.1371/journal.pmed.1001484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Grosse SD, Odame I, Atrash HK, et al. : Sickle cell disease in Africa: A neglected cause of early childhood mortality. Am J Prev Med. 2011;41(6 Suppl 4):S398–405. 10.1016/j.amepre.2011.09.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Telfer P, Coen P, Chakravorty S, et al. : Clinical outcomes in children with sickle cell disease living in England: A neonatal cohort in East London. Haematologica. 2007;92(7):905–12. 10.3324/haematol.10937 [DOI] [PubMed] [Google Scholar]
- 4. Quinn CT, Rogers ZR, McCavit TL, et al. : Improved survival of children and adolescents with sickle cell disease. Blood. 2010;115(17):3447–52. 10.1182/blood-2009-07-233700 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 5. Couque N, Girard D, Ducrocq R, et al. : Improvement of medical care in a cohort of newborns with sickle-cell disease in North Paris: Impact of national guidelines. Br J Haematol. 2016;173(6):927–37. 10.1111/bjh.14015 [DOI] [PubMed] [Google Scholar]
- 6. Elmariah H, Garrett ME, de Castro LM, et al. : Factors associated with survival in a contemporary adult sickle cell disease cohort. Am J Hematol. 2014;89(5):530–5. 10.1002/ajh.23683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Gardner K, Douiri A, Drasar E, et al. : Survival in adults with sickle cell disease in a high-income setting. Blood. 2016;128(10):1436–8. 10.1182/blood-2016-05-716910 [DOI] [PubMed] [Google Scholar]
- 8. Maitra P, Caughey M, Robinson L, et al. : Risk factors for mortality in adult patients with sickle cell disease: A meta-analysis of studies in North America and Europe. Haematologica. 2017;102(4):626–36. 10.3324/haematol.2016.153791 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. DeBaun MR, Ghafuri DL, Rodeghier M, et al. : Decreased median survival of adults with sickle cell disease after adjusting for left truncation bias: A pooled analysis. Blood. 2019;133(6):615–7. 10.1182/blood-2018-10-880575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Fitzhugh CD, Abraham AA, Tisdale JF, et al. : Hematopoietic stem cell transplantation for patients with sickle cell disease: Progress and future directions. Hematol Oncol Clin of North Am. 2014;28(6):1171–85. 10.1016/j.hoc.2014.08.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ribeil JA, Hacein-Bey-Abina S, Payen E, et al. : Gene Therapy in a Patient with Sickle Cell Disease. N Engl J Med. 2017;376(9):848–55. 10.1056/NEJMoa1609677 [DOI] [PubMed] [Google Scholar]
- 12. Epstein FH, Bunn HF: Pathogenesis and Treatment of Sickle Cell Disease. N Engl J Med. 1997;337(11):762–9. 10.1056/NEJM199709113371107 [DOI] [PubMed] [Google Scholar]
- 13. Rees DC, Williams TN, Gladwin MT: Sickle-cell disease. Lancet. 2010;376(9757):2018–31. 10.1016/S0140-6736(10)61029-X [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 14. Lawson V: Turned on by danger: Activation of CD1d-restricted invariant natural killer T cells. Immunology. 2012;137(1):20–7. 10.1111/j.1365-2567.2012.03612.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Reiter CD, Wang X, Tanus-Santos JE, et al. : Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat Med. 2002;8(12):1383–9. 10.1038/nm1202-799 [DOI] [PubMed] [Google Scholar]
- 16. Repka T, Hebbel RP: Hydroxyl radical formation by sickle erythrocyte membranes: Role of pathologic iron deposits and cytoplasmic reducing agents. Blood. 1991;78(10):2753–8. 10.1182/blood.V78.10.2753.bloodjournal78102753 [DOI] [PubMed] [Google Scholar]
- 17. de Caterina R, Libby P, Peng HB, et al. : Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J Clin Invest. 1995;96(1):60–8. 10.1172/JCI118074 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Palmer RMJ, Ashton DS, Moncada S: Vascular endothelial cells synthesize nitric oxide from L-arginine. Nature. 1988;333(6174):664–6. 10.1038/333664a0 [DOI] [PubMed] [Google Scholar]
- 19. Kato GJ, Gladwin MT, Steinberg MH: Deconstructing sickle cell disease: Reappraisal of the role of hemolysis in the development of clinical subphenotypes. Blood Rev. 2007;21(1):37–47. 10.1016/j.blre.2006.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Manwani D, Frenette PS: Vaso-occlusion in sickle cell disease: Pathophysiology and novel targeted therapies. Hematology Am Soc Hematol Educ Program. 2013;2013:362–9. 10.1182/asheducation-2013.1.362 [DOI] [PubMed] [Google Scholar]
- 21. Noubouossie D, Key NS, Ataga KI: Coagulation Abnormalities of Sickle Cell Disease: Relationship With Clinical Outcomes and the Effect of Disease Modifying Therapies. Blood Rev. 2016;30(4):245–56. 10.1016/j.blre.2015.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Kato GJ, Piel FB, Reid CD, et al. : Sickle cell disease. Nat Rev Dis Primers. 2018;4:18010. 10.1038/nrdp.2018.10 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 23. 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(20):1317–22. 10.1056/NEJM199505183322001 [DOI] [PubMed] [Google Scholar]
- 24. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-l-glutamine-sickle-cell-disease [Google Scholar]
- 25. https://www.novartis.com/news/media-releases/new-novartis-medicine-adakveo-crizanlizumab-approved-fda-reduce-frequency-pain-crises-individuals-living-sickle-cell-disease [Google Scholar]
- 26. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-voxelotor-sickle-cell-disease [Google Scholar]
- 27. Eaton WA, Bunn HF: Treating sickle cell disease by targeting HbS polymerization. Blood. 2017;129(20):2719–26. 10.1182/blood-2017-02-765891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. 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(23):1639–44. 10.1056/NEJM199406093302303 [DOI] [PubMed] [Google Scholar]
- 29. Castro O, Brambilla DJ, Thorington B, et al. : The acute chest syndrome in sickle cell disease: Incidence and risk factors. The Cooperative Study of Sickle Cell Disease. Blood. 1994;84(2):643–9. [PubMed] [Google Scholar]
- 30. Mabaera R, West RJ, Conine SJ, et al. : A Cell Stress Signaling Model of Fetal Hemoglobin Induction: What Doesn't Kill Red Blood Cells May Make Them Stronger. Exp Hematol. 2008;36(9):1057–72. 10.1016/j.exphem.2008.06.014 [DOI] [PubMed] [Google Scholar]
- 31. Alcaín FJ, Löw H, Crane FL, et al. : Iron chelators hydroxyurea and bathophenanthroline disulfonate inhibit DNA synthesis by different pathways. Biochem Mol Biol Int. 1994;34(2):273–9. [PubMed] [Google Scholar]
- 32. 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(2):231–9. 10.1172/JCI16672 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Huang J, Yakubu M, Kim-Shapiro DB, et al. : Rat liver-mediated metabolism of hydroxyurea to nitric oxide. Free Radic Biol Med. 2006;40(9):1675–81. 10.1016/j.freeradbiomed.2006.01.002 [DOI] [PubMed] [Google Scholar]
- 34. McGann PT, Ware RE: Hydroxyurea for sickle cell anemia: What have we learned and what questions still remain? Curr Opin Hematol. 2011;18(3):158–65. 10.1097/MOH.0b013e32834521dd [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Bridges KR, Barabino GD, Brugnara C, et al. : A multiparameter analysis of sickle erythrocytes in patients undergoing hydroxyurea therapy. Blood. 1996;88(12):4701–10. [PubMed] [Google Scholar]
- 36. Hillery CA, Du MC, Wang WC, et al. : Hydroxyurea therapy decreases the in vitro adhesion of sickle erythrocytes to thrombospondin and laminin. Br J Haematol. 2000;109(2):322–7. 10.1046/j.1365-2141.2000.02040.x [DOI] [PubMed] [Google Scholar]
- 37. Odievre MH, Bony V, Benkerrou M, et al. : Modulation of erythroid adhesion receptor expression by hydroxyurea in children with sickle cell disease. Haematologica. 2008;93(4):502–10. 10.3324/haematol.12070 [DOI] [PubMed] [Google Scholar]
- 38. Laurance S, Lansiaux P, Pellay FX, et al. : Differential modulation of adhesion molecule expression by hydroxycarbamide in human endothelial cells from the micro- and macrocirculation: Potential implications in sickle cell disease vasoocclusive events. Haematologica. 2011;96(4):534–42. 10.3324/haematol.2010.026740 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. 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(2):436–44. 10.1046/j.1365-2141.2002.03274.x [DOI] [PubMed] [Google Scholar]
- 40. Glover RE, Ivy ED, Orringer EP, et al. : Detection of nitrosyl hemoglobin in venous blood in the treatment of sickle cell anemia with hydroxyurea. Mol Pharmacol. 1999;55(6):1006–10. 10.1124/mol.55.6.1006 [DOI] [PubMed] [Google Scholar]
- 41. 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(9778):1663–72. 10.1016/S0140-6736(11)60355-3 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 42. Dehury S, Purohit P, Patel S, et al. : Low and Fixed Dose of Hydroxyurea Is Effective and Safe in Patients With HbSβ(+) Thalassemia With IVS1-5(G→C) Mutation. Pediatr Blood Cancer. 2015;62(6):1017–23. 10.1002/pbc.25391 [DOI] [PubMed] [Google Scholar]
- 43. 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(10):929–33. 10.1007/s13312-013-0264-0 [DOI] [PubMed] [Google Scholar]
- 44. Ware RE, Davis BR, Schultz WH, et al. : Hydroxycarbamide Versus Chronic Transfusion for Maintenance of Transcranial Doppler Flow Velocities in Children With Sickle Cell anaemia-TCD With Transfusions Changing to Hydroxyurea (TWiTCH): A Multicentre, Open-Label, Phase 3, Non-Inferiority Trial. Lancet. 2016;387(10019):661–70. 10.1016/S0140-6736(15)01041-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 45. Bartolucci P, Habibi A, Stehlé T, et al. : Six Months of Hydroxyurea Reduces Albuminuria in Patients With Sickle Cell Disease. J Am Soc Nephrol. 2016;27(6):1847–53. 10.1681/ASN.2014111126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Lobo CLdC, Pinto JFC, Nascimento EM, et al. : The Effect of Hydroxcarbamide Therapy on Survival of Children With Sickle Cell Disease. Br J Haematol. 2013;161(6):852–60. 10.1111/bjh.12323 [DOI] [PubMed] [Google Scholar]
- 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(13):1645–51. 10.1001/jama.289.13.1645 [DOI] [PubMed] [Google Scholar]
- 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(6):403–8. 10.1002/ajh.21699 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 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(12):2354–63. 10.1182/blood-2009-05-221333 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 50. Ware RE: Optimizing Hydroxyurea Therapy for Sickle Cell Anemia. Hematology Am Soc Hematol Educ Program. 2015;2015:436–43. 10.1182/asheducation-2015.1.436 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 51. 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(8):871–9. 10.1002/ajh.25510 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 52. 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(12):1333–9. 10.1002/ajh.24906 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Hydroxyurea Optimization Through Precision Study (HOPS). Clinical Trials. 2020 doi: 10.1186/s13063-020-04912-z. Reference Source [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Nazon C, Sabo AN, Becker G, et al. : Optimizing Hydroxyurea Treatment for Sickle Cell Disease Patients: The Pharmacokinetic Approach. J Clin Med. 2019;8(10):1701. 10.3390/jcm8101701 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 55. Saunthararajah Y, Molokie R, Saraf S, et al. : Clinical effectiveness of decitabine in severe sickle cell disease. Br J Haematol. 2008;141(1):126–9. 10.1111/j.1365-2141.2008.07027.x [DOI] [PubMed] [Google Scholar]
- 56. Molokie R, Lavelle D, Gowhari M, et al. : Oral tetrahydrouridine and decitabine for non-cytotoxic epigenetic gene regulation in sickle cell disease: A randomized phase 1 study. PLoS Med. 2017;14(9):e1002382. 10.1371/journal.pmed.1002382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Atweh GF, Sutton M, Nassif I, et al. : Sustained induction of fetal hemoglobin by pulse butyrate therapy in sickle cell disease. Blood. 1999;93(6):1790–7. 10.1182/blood.V93.6.1790.406k27_1790_1797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Hebbel RP, Vercellotti GM, Pace BS, et al. : The HDAC inhibitors trichostatin A and suberoylanilide hydroxamic acid exhibit multiple modalities of benefit for the vascular pathobiology of sickle transgenic mice. Blood. 2010;115(12):2483–90. 10.1182/blood-2009-02-204990 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Ikuta T, Atweh G, Boosalis V, et al. : Cellular and Molecular Effects of a Pulse Butyrate Regimen and New Inducers of Globin Gene Expression and Hematopoiesis. Ann N Y Acad Sci. 1998;850:87–99. 10.1111/j.1749-6632.1998.tb10466.x [DOI] [PubMed] [Google Scholar]
- 60. Kutlar A, Ataga K, Reid M, et al. : A phase 1/2 trial of HQK-1001, an oral fetal globin inducer, in sickle cell disease. Am J Hematol. 2012;87(11):1017–21. 10.1002/ajh.23306 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Kutlar A, Reid ME, Inati A, et al. : A dose-escalation phase IIa study of 2,2-dimethylbutyrate (HQK-1001), an oral fetal globin inducer, in sickle cell disease. Am J Hematol. 2013;88(11):E255–E260. 10.1002/ajh.23533 [DOI] [PubMed] [Google Scholar]
- 62. Okam MM, Esrick EB, Mandell E, et al. : Phase 1/2 trial of vorinostat in patients with sickle cell disease who have not benefitted from hydroxyurea. Blood. 2015;125(33):3668–9. 10.1182/blood-2015-03-635391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Reid ME, El Beshlawy A, Inati A, et al. : A double-blind, placebo-controlled phase II study of the efficacy and safety of 2,2-dimethylbutyrate (HQK-1001), an oral fetal globin inducer, in sickle cell disease. Am J Hematol. 2014;89(7):709–13. 10.1002/ajh.23725 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 64. Shearstone JR, Golonzhka O, Chonkar A, et al. : Chemical Inhibition of Histone Deacetylases 1 and 2 Induces Fetal Hemoglobin through Activation of GATA2. PLoS One. 2016;11:e0153767. 10.1371/journal.pone.0153767 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Rivers A, Vaitkus K, Ruiz MA, et al. : RN-1, a potent and selective lysine-specific demethylase 1 inhibitor, increases γ-globin expression, F reticulocytes, and F cells in a sickle cell disease mouse model. Exp Hematol. 2015;43(7):546–553.e3. 10.1016/j.exphem.2015.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Cui S, Lim KC, Shi L, et al. : The LSD1 inhibitor RN-1 induces fetal hemoglobin synthesis and reduces disease pathology in sickle cell mice. Blood. 2015;126(3):386–96. 10.1182/blood-2015-02-626259 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Rivers A, Vaitkus K, Jagadeeswaran R, et al. : Oral administration of the LSD1 inhibitor ORY-3001 increases fetal hemoglobin in sickle cell mice and baboons. Exp Hematol. 2018;67:60–64.e2. 10.1016/j.exphem.2018.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 68. 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(4):1109–12. 10.1182/blood-2010-11-319137 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 69. Kutlar A, Swerdlow PS, Meiler SE, et al. : Pomalidomide In Sickle Cell Disease: Phase I Study Of a Novel Anti-Switching Agent. Blood. 2013;122(21):777 10.1182/blood.V122.21.777.777 [DOI] [Google Scholar]
- 70. Zhang Y, Paikari A, Sumazin P, et al. : Metformin induces FOXO3-dependent fetal hemoglobin production in human primary erythroid cells. Blood. 2018;132(3):321–33. 10.1182/blood-2017-11-814335 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 71. Krishnamoorthy S, Pace B, Gupta D, et al. : Dimethyl fumarate increases fetal hemoglobin, provides heme detoxification, and corrects anemia in sickle cell disease. JCI Insight. 2017;2(20):e96409. 10.1172/jci.insight.96409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Dai Y, Chen T, Ijaz H, et al. : SIRT1 activates the expression of fetal hemoglobin genes. Am J Hematol. 2017;92(11):1177–1186. 10.1002/ajh.24879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Oksenberg D, Dufu K, Patel MP, et al. : GBT440 increases haemoglobin oxygen affinity, reduces sickling and prolongs RBC half-life in a murine model of sickle cell disease. Br J Haematol. 2016;175(1):141–53. 10.1111/bjh.14214 [DOI] [PubMed] [Google Scholar]
- 74. Howard J, Hemmaway CJ, Telfer P, et al. : A phase 1/2 ascending dose study and open-label extension study of voxelotor in patients with sickle cell disease. Blood. 2019;133(17):1865–1875. 10.1182/blood-2018-08-868893 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 75. Dufu K, Patel M, Oksenberg D, et al. : GBT440 improves red blood cell deformability and reduces viscosity of sickle cell blood under deoxygenated conditions. Clin Hemorheol Microcirc. 2018;70(1):95–105. 10.3233/CH-170340 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 76. Vichinsky E, Hoppe CC, Ataga KI, et al. : A Phase 3 Randomized Trial of Voxelotor in Sickle Cell Disease. N Engl J Med. 2019;381(6):509–519. 10.1056/NEJMoa1903212 [DOI] [PubMed] [Google Scholar]
- 77. Kato GJ, Lawrence MP, Mendelsohn LG, et al. : Phase 1 Clinical Trial Of The Candidate Anti-Sickling Agent Aes-103 In Adults With Sickle Cell Anemia. Blood. 2013;122:1009 10.1182/blood.V122.21.1009.1009 [DOI] [Google Scholar]
- 78. Kalfa TA, Kuypers FA, Telen MJ, et al. : Phase 1 Single (SAD) and Multiple Ascending Dose (MAD) Studies of the Safety, Tolerability, Pharmacokinetics (PK) and Pharmacodynamics (PD) of FT-4202, an Allosteric Activator of Pyruvate Kinase-R, in Healthy and Sickle Cell Disease Subjects. Blood. 2019;134(Supplement_1):616 10.1182/blood-2019-121889 [DOI] [Google Scholar]
- 79. Belcher JD, Young M, Chen C, et al. : MP4CO, a pegylated hemoglobin saturated with carbon monoxide, is a modulator of HO-1, inflammation, and vaso-occlusion in transgenic sickle mice. Blood. 2013;122(15):2757–64. 10.1182/blood-2013-02-486282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Abuchowski A: SANGUINATE (PEGylated Carboxyhemoglobin Bovine): Mechanism of Action and Clinical Update. Artif Organs. 2017;41(4):346–350. 10.1111/aor.12934 [DOI] [PubMed] [Google Scholar]
- 81. Keipert PE: Clinical Evaluation of MP4CO: A Phase 1b Escalating-Dose, Safety and Tolerability Study in Stable Adult Patients with Sickle Cell Disease. Adv Exp Med Biol. 2016;923:23–29. 10.1007/978-3-319-38810-6_3 [DOI] [PubMed] [Google Scholar]
- 82. Misra H, Bainbridge J, Berryman J, et al. : A Phase Ib open label, randomized, safety study of SANGUINATE™ in patients with sickle cell anemia. Rev Bras Hematol Hemoter. 2017;39(1):20–27. 10.1016/j.bjhh.2016.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Sunshine HR, Hofrichter J, Eaton WA: Requirement for therapeutic inhibition of sickle haemoglobin gelation. Nature. 1978;275(5677):238–40. 10.1038/275238a0 [DOI] [PubMed] [Google Scholar]
- 84. Ataga KI, Reid M, Ballas SK, et al. : Improvements in haemolysis and indicators of erythrocyte survival do not correlate with acute vaso-occlusive crises in patients with sickle cell disease: A phase III randomized, placebo-controlled, double-blind study of the Gardos channel blocker senicapoc (ICA-17043). Br J Haematol. 2011;153(1):92–104. 10.1111/j.1365-2141.2010.08520.x [DOI] [PubMed] [Google Scholar]
- 85. Ataga KI, Smith WR, de Castro LM, et al. : Efficacy and safety of the Gardos channel blocker, senicapoc (ICA-17043), in patients with sickle cell anemia. Blood. 2008;111(8):3991–7. 10.1182/blood-2007-08-110098 [DOI] [PubMed] [Google Scholar]
- 86. Brousseau DC, Scott JP, Badaki-Makun O, et al. : A multicenter randomized controlled trial of intravenous magnesium for sickle cell pain crisis in children. Blood. 2015;126(14):1651–7. 10.1182/blood-2015-05-647107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Goldman RD, Mounstephen W, Kirby-Allen M, et al. : Intravenous magnesium sulfate for vaso-occlusive episodes in sickle cell disease. Pediatrics. 2013;132(6):e1634–41. 10.1542/peds.2013-2065 [DOI] [PubMed] [Google Scholar]
- 88. de Franceschi L, Bachir D, Galacteros F, et al. : Oral magnesium pidolate: Effects of long-term administration in patients with sickle cell disease. Br J Haematol. 2000;108(2):284–9. 10.1046/j.1365-2141.2000.01861.x [DOI] [PubMed] [Google Scholar]
- 89. Swift R, Abdulmalik O, Chen Q, et al. : SCD-101: A New Anti-Sickling Drug Reduces Pain and Fatigue and Improves Red Blood Cell Shape in Peripheral Blood of Patients with Sickle Cell Disease. Blood. 2016;128(22):121 10.1182/blood.V128.22.121.121 [DOI] [Google Scholar]
- 90. Niihara Y, Smith WR, Stark CW: A Phase 3 Trial of l-Glutamine in Sickle Cell Disease. N Engl J Med. 2018;379(19):1880. 10.1056/NEJMc1811050 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 91. Nur E, Brandjes DP, Teerlink T, et al. : N-acetylcysteine reduces oxidative stress in sickle cell patients. Ann Hematol. 2012;91(7):1097–105. 10.1007/s00277-011-1404-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Sins JWR, Fijnvandraat K, Rijneveld AW, et al. : Effect of N-acetylcysteine on pain in daily life in patients with sickle cell disease: A randomised clinical trial. Br J Haematol. 2018;182(3):444–8. 10.1111/bjh.14809 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 93. Ataga KI, Kutlar A, Kanter J, et al. : Crizanlizumab for the Prevention of Pain Crises in Sickle Cell Disease. N Engl J Med. 2017;376(5):429–39. 10.1056/NEJMoa1611770 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Orringer EP, Casella JF, Ataga KI, et al. : Purified poloxamer 188 for treatment of acute vaso-occlusive crisis of sickle cell disease: A randomized controlled trial. JAMA. 2001;286(17):2099–106. 10.1001/jama.286.17.2099 [DOI] [PubMed] [Google Scholar]
- 95. https://www.prnewswire.com/news-releases/mast-therapeutics-reports-top-line-results-from-phase-3-study-in-sickle-cell-disease-300331289.html [Google Scholar]
- 96. 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(10):1779–86. 10.1182/blood-2009-12-260513 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 97. Telen MJ, Wun T, McCavit TL, et al. : Randomized phase 2 study of GMI-1070 in SCD: Reduction in time to resolution of vaso-occlusive events and decreased opioid use. Blood. 2015;125(17):2656–64. 10.1182/blood-2014-06-583351 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 98. https://www.pfizer.com/news/press-release/press-release-detail/pfizer_announces_phase_3_top_line_results_for_rivipansel_in_patients_with_sickle_cell_disease_experiencing_a_vaso_occlusive_crisis [Google Scholar]
- 99. Matsui NM, Varki A, Embury SH: Heparin inhibits the flow adhesion of sickle red blood cells to P-selectin. Blood. 2002;100(10):3790–6. 10.1182/blood-2002-02-0626 [DOI] [PubMed] [Google Scholar]
- 100. Embury SH, Matsui NM, Ramanujam S, et al. : The contribution of endothelial cell P-selectin to the microvascular flow of mouse sickle erythrocytes in vivo. Blood. 2004;104(10):3378–85. 10.1182/blood-2004-02-0713 [DOI] [PubMed] [Google Scholar]
- 101. 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. 2017;98(02):392–6. 10.1160/Th06-12-0718 [DOI] [PubMed] [Google Scholar]
- 102. Telen MJ, Batchvarova M, Shan S, et al. : Sevuparin Binds to Multiple Adhesive Ligands and Reduces Sickle Red Blood Cell-induced Vaso-occlusion. Br J Haematol. 2016;175:935–48. 10.1111/bjh.14303 [DOI] [PubMed] [Google Scholar]
- 103. Biemond BJ, Tombak A, Kilinc Y, et al. : Efficacy and Safety of Sevuparin, a Novel Non-Anti-Coagulant Heparinoid, in Patients with Acute Painful Vaso-Occlusive Crisis; A Global, Multicenter Double-Blind, Randomized, Placebo-Controlled Phase 2 Trial (TVOC01). Blood. 2019;134(supp 1):614 10.1182/blood-2019-124653 31270104 [DOI] [Google Scholar]
- 104. Jang JE, Hidalgo A, Frenette PS: Intravenous immunoglobulins modulate neutrophil activation and vascular injury through FcγRIII and SHP-1. Circ Res. 2012;110(8):1057–66. 10.1161/CIRCRESAHA.112.266411 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Manwani D, Chen G, Carullo V, et al. : Single-dose Intravenous Gammaglobulin Can Stabilize Neutrophil Mac-1 Activation in Sickle Cell Pain Crisis. Am J Hematol. 2015;90(5):381–5. 10.1002/ajh.23956 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Griffin TC, McIntire D, Buchanan GR: High-dose intravenous methylprednisolone therapy for pain in children and adolescents with sickle cell disease. N Engl J Med. 1994;330(11):733–7. 10.1056/NEJM199403173301101 [DOI] [PubMed] [Google Scholar]
- 107. Bernini JC, Rogers ZR, Sandler ES, et al. : Beneficial Effect of Intravenous Dexamethasone in Children With Mild to Moderately Severe Acute Chest Syndrome Complicating Sickle Cell Disease. Blood. 1998;92(9):3082–9. 10.1182/blood.V92.9.3082 [DOI] [PubMed] [Google Scholar]
- 108. Leonard A, Godiwala N, Herrera N, et al. : Early Initiation of Inhaled Corticosteroids does not Decrease Acute Chest Syndrome Morbidity in Pediatric Patients with Sickle Cell Disease. Blood Cells Mol Dis. 2018;71:55–62. 10.1016/j.bcmd.2018.03.001 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 109. Glassberg J, Minnitti C, Cromwell C, et al. : Inhaled Steroids Reduce Pain and sVCAM Levels in Individuals with Sickle Cell Disease: A Triple-blind, Randomized trial. Am J Hematol. 2017;92:622–31. 10.1002/ajh.24742 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 110. Wallace KL, Linden J: Adenosine A2A Receptors Induced on iNKT and NK Cells Reduce Pulmonary Inflammation and Injury in Mice with Sickle Cell Disease. Blood. 2010;116:5010–20. 10.1182/blood-2010-06-290643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. 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 regadenoson. Blood. 2013;121(17):3329–34. 10.1182/blood-2012-11-465963 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 112. Field JJ, Majerus E, Gordeuk VR, et al. : Randomized phase 2 trial of regadenoson for treatment of acute vaso-occlusive crises in sickle cell disease. Blood Adv. 2017;1(20):1645–9. 10.1182/bloodadvances.2017009613 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Field JJ, Majerus E, Ataga KI, et al. : NNKTT120, an anti-iNKT cell monoclonal antibody, produces rapid and sustained iNKT cell depletion in adults with sickle cell disease. PLoS One. 2017;12(2):e0171067. 10.1371/journal.pone.0171067 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. 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(5):655–63. 10.1111/j.1365-2141.2010.08480.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Hoppe C, Jacob E, Styles L, et al. : Simvastatin reduces vaso-occlusive pain in sickle cell anaemia: A pilot efficacy trial. Br J Haematol. 2017;177(4):620–9. 10.1111/bjh.14580 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Ataga KI, Wichlan D, Elsherif L, et al. : A pilot study of the effect of atorvastatin on endothelial function and albuminuria in sickle cell disease. Am J Hematol. 2019;94(11):E299–E301. 10.1002/ajh.25614 [DOI] [PubMed] [Google Scholar]
- 117. Daak AA, Ghebremeskel K, Hassan Z, et al. : Effect of omega-3 (n-3) fatty acid supplementation in patients with sickle cell anemia: Randomized, double-blind, placebo-controlled trial. Am J Clin Nutr. 2013;97(1):37–44. 10.3945/ajcn.112.036319 [DOI] [PubMed] [Google Scholar]
- 118. Daak AA, Dampier CD, Fuh B, et al. : Double-blind, randomized, multicenter phase 2 study of SC411 in children with sickle cell disease (SCOT trial). Blood Adv. 2018;2(15):1969–79. 10.1182/bloodadvances.2018021444 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 119. Rees DC, Kilinc Y, Unal S, et al. : Double-Blind, Randomized Study of Canakinumab Treatment in Pediatric and Young Adult Patients with Sickle Cell Anemia. Blood. 2019;134:615 10.1182/blood-2019-123355 [DOI] [Google Scholar]
- 120. Gavins FNE, Russell J, Senchenkova EL, et al. : Mechanisms of enhanced thrombus formation in cerebral microvessels of mice expressing hemoglobin-S. Blood. 2011;117(15):4125–33. 10.1182/blood-2010-08-301366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Chantrathammachart P, Mackman N, Sparkenbaugh E, et al. : Tissue Factor Promotes Activation of Coagulation and Inflammation in a Mouse Model of Sickle Cell Disease. Blood. 2012;120(3):636–46. 10.1182/blood-2012-04-424143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Arumugam PI, Mullins ES, Shanmukhappa SK, et al. : Genetic diminution of circulating prothrombin ameliorates multiorgan pathologies in sickle cell disease mice. Blood. 2015;126(15):1844–55. 10.1182/blood-2015-01-625707 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Ataga KI, Key NS: Hypercoagulability in Sickle Cell Disease: New Approaches to an Old Problem. Hematology Am Soc Hematol Educ Program. 2007;2007:91–6. 10.1182/asheducation-2007.1.91 [DOI] [PubMed] [Google Scholar]
- 124. Pakbaz Z, Wun T: Role of the Hemostatic System on Sickle Cell Disease Pathophysiology and Potential Therapeutics. Hematol Oncol Clin North Am. 2014;28(2):355–74. 10.1016/j.hoc.2013.11.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Greenberg J, Ohene-Frempong K, Halus J, et al. : Trial of Low Doses of Aspirin as Prophylaxis in Sickle Cell Disease. J Pediatr. 1983;102(5):781–4. 10.1016/s0022-3476(83)80258-3 [DOI] [PubMed] [Google Scholar]
- 126. Osamo NO, Photiades DP, Famodu AA: Therapeutic effect of aspirin in sickle cell anaemia. Acta Haematol. 1981;66(2):102–7. 10.1159/000207105 [DOI] [PubMed] [Google Scholar]
- 127. Zago MA, Costa FF, Ismael SJ, et al. : Treatment of Sickle Cell Diseases With Aspirin. Acta Haematol. 1984;72(1):61–4. 10.1159/000206360 [DOI] [PubMed] [Google Scholar]
- 128. Desai PC, Brittain JE, Jones SK, et al. : A Pilot Study of Eptifibatide for Treatment of Acute Pain Episodes in Sickle Cell Disease. Thromb Res. 2013;132(3):341–5. 10.1016/j.thromres.2013.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 129. Cabannes R, Lonsdorfer J, Castaigne JP, et al. : Clinical and Biological Double-Blind-Study of Ticlopidine in Preventive Treatment of Sickle-Cell Disease Crises. Agents Actions Suppl. 1984;15:199–212. [PubMed] [Google Scholar]
- 130. Heeney MM, Hoppe CC, Abboud MR, et al. : A Multinational Trial of Prasugrel for Sickle Cell Vaso-Occlusive Events. N Engl J Med. 2016;374(7):625–35. 10.1056/NEJMoa1512021 [DOI] [PubMed] [Google Scholar]
- 131. Hsu LL, Sarnaik S, Williams S, et al. : A Dose-Ranging Study of Ticagrelor in Children Aged 3-17 Years With Sickle Cell Disease: A 2-part Phase 2 Study. Am J Hematol. 2018;93(12):1493–500. 10.1002/ajh.25273 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 132. Kanter J, Abboud MR, Kaya B, et al. : Ticagrelor Does Not Impact Patient-Reported Pain in Young Adults With Sickle Cell Disease: A Multicentre, Randomised Phase IIb Study. Br J Haematol. 2019;184(2):269–78. 10.1111/bjh.15646 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 133. Heeney MM, Abboud MR, Amilon C, et al. : Ticagrelor Versus Placebo for the Reduction of Vaso-Occlusive Crises in Pediatric Sickle Cell Disease: Rationale and Design of a Randomized, Double-Blind, Parallel-Group, Multicenter Phase 3 Study (HESTIA3). Contemp Clin Trials. 2019;85:105835. 10.1016/j.cct.2019.105835 [DOI] [PubMed] [Google Scholar]
- 134. Kato GJ, Steinberg MH, Gladwin MT: Intravascular Hemolysis and the Pathophysiology of Sickle Cell Disease. J Clin Invest. 2017;127(3):750–60. 10.1172/JCI89741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Gladwin MT, Kato GJ, Weiner D, et al. : Nitric Oxide for Inhalation in the Acute Treatment of Sickle Cell Pain Crisis: A Randomized Controlled Trial. JAMA. 2011;305(9):893–902. 10.1001/jama.2011.235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Maitre B, Djibre M, Katsahian S, et al. : Inhaled Nitric Oxide for Acute Chest Syndrome in Adult Sickle Cell Patients: A Randomized Controlled Study. Intensive Care Med. 2015;41(12):2121–9. 10.1007/s00134-015-4060-2 [DOI] [PubMed] [Google Scholar]
- 137. Morris CR, Kuypers FA, Lavrisha L, et al. : A Randomized, Placebo-Controlled Trial of Arginine Therapy for the Treatment of Children With Sickle Cell Disease Hospitalized With Vaso-Occlusive Pain Episodes. Haematologica. 2013;98(9):1375–82. 10.3324/haematol.2013.086637 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 138. Eleutério RMN, Nascimento FO, Araújo TG, et al. : Double-Blind Clinical Trial of Arginine Supplementation in the Treatment of Adult Patients With Sickle Cell Anaemia. Adv Hematol. 2019;2019:4397150. 10.1155/2019/4397150 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 139. Machado RF, Barst RJ, Yovetich NA, et al. : Hospitalization for Pain in Patients With Sickle Cell Disease Treated With Sildenafil for Elevated TRV and Low Exercise Capacity. Blood. 2011;118(4):855–64. 10.1182/blood-2010-09-306167 [DOI] [PMC free article] [PubMed] [Google Scholar]