Abstract
Introduction:
Sickle cell disease (SCD) is an orphan disease in the United States, but is highly prevalent worldwide. Only two drugs, hydroxyurea and L-glutamine, are approved for this disease. With an improved understanding of the pathophysiology of SCD as well as the success of several recently approved drugs for other orphan diseases, there is an increased interest in the development of drugs for SCD.
Areas covered:
This review summarizes published studies of drug therapies and ongoing trials of novel agents.
Expert opinion:
The development of drugs with different mechanisms of action offers opportunities for combination and individualized therapy in SCD. In addition to acute pain crisis, the evaluation of other SCD-related complications, exercise capacity, patient reported outcomes and validated surrogate endpoints are necessary to advance drug development. It is important to involve sites in sub-Saharan Africa and India, which have the highest burden of SCD, in trials of novel therapies.
Keywords: Sickle cell disease, Treatment, Novel Drugs, Drug Development, Clinical Trials
Introduction
Sickle cell disease is a genetic disorder that occurs due to a single gene mutation and results in substitution of valine for glutamine in the beta globin chain. Although millions of individuals are affected worldwide, SCD is referred to an orphan disease in the United States (US), with a prevalence of approximately 100,000 individuals. An estimated 242,000 children with sickle cell anemia were born in sub-Saharan Africa in 2010, with approximately 40,000 children born in India [1]. Comparatively, the estimated number of children born in the Americas and Europe in 2010 were much lower, at 11,000 and 2,000, respectively [1]. The vast majority of children who reside in resource-rich countries live to adulthood [2, 3, 4], but there continues to be a high mortality rate associated with SCD in childhood in sub-Saharan Africa. Although they have an increased survival to adulthood, individuals with SCD in resource-rich nations have a shorter life expectancy compared with the general population [5, 6, 7]. Furthermore, despite the success of allogeneic bone marrow transplantation as a curative approach [8, 9] and the curative potential of gene therapy [10], these treatment modalities are not available to the vast majority of individuals with SCD, most of whom reside in resource-poor countries. As such, the availability of drug therapies that are safe, effective and affordable is highly desirable and warranted.
Pathophysiology
The development of effective therapies for SCD and its complications depends on an adequate understanding of its pathophysiology (Figure 1). The pathophysiology of SCD is complex and appears to be due to multiple pathways. The primary event is due to the insolubility of sickle hemoglobin (HbS) when it is deoxygenated, leading to intracellular red blood cell (RBC) polymer formation and subsequent erythrocyte deformity. The rate and extent of HbS polymerization is dependent on the extent and duration of hemoglobin deoxygenation, the presence of fetal hemoglobin, as well as the intracellular HbS concentration [11]. The intracellular HbS concentration can vary greatly in patients with SCD, due to both a high number of circulating reticulocytes, having some of the lowest intracellular hemoglobin concentrations, and a substantial number of very dense RBCs that arise following polymerization-induced membrane damage and dehydration. The dehydration of sickle RBC, following the loss of solute and osmotically obliged water, results from increased potassium efflux through two specific pathways: the calcium-activated potassium efflux (or Gardos) channel and the potassium-chloride co-transport pathway [11].
Mechanisms of action of pharmacologic agents that have been evaluated and/or are being tested in sickle cell disease.
RBCs - Red Blood cells; HbF – Hemoglobin F; NO – Nitric Oxide; Invariant NKT cells – Invariant Natural Killer T cells. Modified with permission from [173]
The clinical manifestations of SCD appear to be driven by two major pathophysiological processes: vaso-occlusion with ischemia-reperfusion injury and hemolytic anemia. Vaso-occlusive crisis likely occurs due to adhesive interactions of sickle RBCs and leukocytes with the endothelium causing vascular obstruction and tissue ischemia [12]. These episodes of microvascular obstruction are followed by the restoration of blood flow, which promotes further tissue injury by reperfusion. Reperfusion injury results in increased oxidant and inflammatory stress, with increased synthesis of inflammatory cytokines and increased expression of endothelial adhesion molecules. The inflammatory cascade from ischemia-reperfusion is amplified by the activation of CD1d-restricted invariant natural killer T (iNKT) cells [13].
Associations are reported between markers of hemolysis and complications of SCD, including cholelithiasis, lower extremity ulcers, priapism, increased echocardiography-derived tricuspid regurgitant jet velocity, chronic kidney disease, stroke and increased mortality [14]. Free plasma hemoglobin, released following intravascular hemolysis, reacts directly to scavenge nitric oxide (NO), and generates reactive oxygen species, such as the hydroxyl and superoxide radical, potent scavengers of NO [15, 16]. NO, normally produced by the endothelium, regulates basal vasodilator tone, inhibits platelet and coagulation activation, and transcriptional expression of nuclear factor κB (NFκB)-dependent adhesion molecules, such as vascular cell-adhesion molecule-1, intercellular cell-adhesion molecule-1, and the selectins [17, 18]. Free plasma hemoglobin potently inhibits endothelial NO signaling, leading to endothelial cell dysfunction and NO resistance [12]. Arginase-1, released following hemolysis, metabolizes plasma arginine into ornithine, resulting in a decrease in the required substrate for NO synthesis and further decreases the bioavailability of NO [15]. Release of hemin following intravascular hemolysis also contributes to inflammation [19]. The contribution of inflammation, as well as platelet and coagulation activation to the pathophysiology of SCD have recently been extensively reviewed [20, 21].
Drug Development in Sickle Cell Disease
Despite an improved understanding of the pathophysiology of SCD, available drug treatments remain limited. Although SCD is referred to as the first molecular disease [22], to date, only two drugs, hydroxyurea and L-glutamine, are approved by regulatory agencies including the Food and Drug Administration (FDA) and European Medicines Agency specifically for this disease. This compares unfavorably with many other prevalent and orphan diseases in the US. First described relatively recently, there were 25 FDA approved individual drugs and 14 approved combination drugs for the treatment of HIV as at November, 2017 (https://aidsinfo.nih.gov/education-materials/fact-sheets/21/58/fda-approved-hiv-medicines). Similarly, there are at least 10 drugs approved for pulmonary arterial hypertension and 9 drugs, including 2 FDA-approved transmembrane conductance regulator modulators, for cystic fibrosis. The disparity in approved drug therapies for SCD compared to many other disease conditions is likely due to multiple factors. There is disparate funding per individual affected by SCD and cystic fibrosis from both the National Institutes of Health and national foundations [23]. With the low prevalence of SCD in resource-rich countries and the limited economic power of the affected patients, there was no great incentive to invest in the development of new drugs for many years. However, there is now a growing interest in the development of new drugs and the repurposing of existing drugs for SCD. Government incentives including the Orphan Drug Act of 1983 and the Rare Diseases Act of 2003, both in the U.S., and the Orphan Regulation adopted by the European Union in 1999 have provided organizational support and financial incentives to defray the costs of drug development [24]. Furthermore, successes in the development of drugs for orphan indications (e.g. eculizumab for paroxysmal nocturnal hemoglobinuria) show that the market for orphan diseases can be lucrative (http://www.pharmaceutical-journal.com/news-and-analysis/news-in-brief/eculizumab-with-340200-price-tag-gets-go-ahead-from-nice/20067720.article). Despite the increased funding since the passage of the National Sickle Cell Anemia Control Act of 1972 and the Sickle Cell Treatment Act of 2003 in the U.S., research funding for SCD remains inadequate. Finally, the successful accrual into several relatively large, multicenter trials in SCD [25, 26, 27, 28, 29] suggests that, given the opportunity, patients are willing to participate in well-designed clinical trials.
Hemoglobin F Inducing and Anti-Sickling Agents
There are several therapeutic approaches to inhibiting HbS polymerization, including blocking intermolecular contacts in the sickle fiber, induction of hemoglobin F (HbF) synthesis, reduction of intracellular hemoglobin S concentration, increase in oxygen affinity and reduction of concentration of 2,3,-diphosphoglycerate [30]. Ongoing studies of anti-sickling agents are shown in Table 1.
Table 1:
Ongoing Clinical Trials of Anti-Sickling Agents in Sickle Cell Disease
Drug | Study Title (Sponsor) | Mechanism of Action | Clinical Phase | NCT Number | Sponsor |
---|---|---|---|---|---|
Sanguinate | A Study of SANGUINATE for the Treatment of Vaso-occlusive Crisis (VOC) in Adult Sickle Cell Disease Patients |
Oxygen transfer agent | Phase 2 | NCT02672540 | Prolong Pharmaceuticals |
Voxelotor | Study to Evaluate the Effect of GBT440 Administered Orally to Patients With Sickle Cell Disease (GBT_HOPE) (GBT_HOPE) |
Increase hemoglobin oxygen affinity (left shift of oxyhemoglobin dissociation curve) |
NCT03036813 | Global Blood Therapeutics |
|
Panobinostat | Study of Panobinostat (LBH589) in Patients With Sickle Cell Disease (LBH589) |
Increase hemoglobin F (histone deacetylase inhibitor) |
Phase 1 | NCT01245179 | Novartis |
INCB059872 | A Study to Evaluate Safety, Pharmacokinetic, and Biological Activity of INCB059872 in Subjects With Sickle Cell Disease |
Increase hemoglobin F by inhibition of lysine- specific demethylase 1 (LSD1) |
Phase 1 | NCT03132324 | Incyte Corporation |
Decitabine and Tetrahydrouridine |
Study of Decitabine and Tetrahydrouridine (THU) in Patients With Sickle Cell Disease |
Increase hemoglobin F (hypomethylating agent) |
Phase 1 | NCT01685515 | The Cleveland Clinic |
Metformin | Fetal Hemoglobin Induction Treatment Metformin (FITMet) |
Increase hemoglobin F | Phase 1 | NCT02981329 | Baylor College of Medicine |
Hemoglobin F Inducing Agents
It is well recognized that high levels of HbF ameliorates the severity of SCD [7, 31, 32, 33]. Hydroxyurea is approved by the FDA, as well as the European Medicines Agency, for treatment of individuals with sickle cell anemia. Hydroxyurea is a potent inducer of HbF [34], although the mechanisms for HbF induction are not fully understood [34, 35, 36, 37]. In addition, it is an NO donor [38, 39], improves erythrocyte deformability, reduces hemolysis, lowers circulating leukocytes and reticulocytes [40], and reduces surface expression of adhesion receptors [41, 42, 43, 44]. A randomized, placebo-controlled, phase 3 trial of adult patients with severe sickle cell anemia showed significant reductions in the frequency of vaso-occlusive crises, acute chest syndrome, need for blood transfusion, and delayed onset of first vaso-occlusive crisis with hydroxyurea treatment [26]. Another phase 3 clinical trial of hydroxyurea (BABY HUG), involving infants with sickle cell anemia randomized to either hydroxyurea or placebo similarly showed significant decreases in pain episodes, acute chest syndrome, hospitalizations, and RBC transfusions, although no benefits were observed on the primary endpoints, splenic function and glomerular filtration rate [28]. By decreasing the transcranial Doppler velocity and albuminuria, hydroxyurea may decrease the risk of stroke and chronic kidney disease in sickle cell anemia [45, 46]. Long-term use of hydroxyurea also appears to reduce mortality in patients with SCD [47, 48, 49, 50]. Recent data demonstrate that treatment with low, fixed weight-based dosing of hydroxyurea (10 mg/kg daily) decreases the rates of acute painful events, hospitalization, and blood transfusions in both children and adults with low toxicity [51, 52], suggesting that limited monitoring for hematological toxicities may be feasible in resource-limited areas. Despite its multiple beneficial effects, a substantial number of patients on hydroxyurea therapy may not obtain an adequate clinical response. In addition, despite its overall favorable safety profile, some patients remain reluctant to take this medication due to its effect on decreasing spermatogenesis [53] and their concerns regarding increased risk for leukemogenesis and teratogenicity.
Decitabine (5-aza-2’-deoxycytidine), a DNA hypomethylating agent which depletes DNA methyltransferase (DNMT1), has been studied as a potential diseasemodifying agent for SCD. Administered via a subcutaneous route, at demonstrably non-cytotoxic doses, decitabine increased HbF and total hemoglobin levels in patients who had no response to hydroxyurea [54]. However, decitabine has a very short plasma half-life of only minutes, and negligible solid tissue distribution and oral bioavailability due to its rapid inactivation to uridine degradation products by cytidine deaminase (CDA), an enzyme which is highly expressed in the intestines and liver [55, 56, 57, 58, 59]). In a recent study combining escalating doses of oral decitabine with an oral CDA inhibitor, tetrahydrouridine (THU), patients were treated twice per week for 8 weeks, with 4 weeks of follow-up [60]. The primary endpoint in this early phase study of grade 3 non-hematologic toxicity was not reached, and adverse events were not significantly different in patients treated with THU-decitabine compared with placebo [60]. However, the highest tested dose decreased DNMT1 protein in peripheral blood mononuclear cells, decreased repetitive element CpG methylation, and increased HbF, doubling F- cells up to approximately 80% of total RBCs, and increased total hemoglobin as reticulocytes simultaneously decreased. Furthermore, biomarkers of hemolysis, coagulation activation, and inflammation improved.
Histone deacetylase inhibitors increase levels of HbF in SCD [61, 62, 63, 64, 65, 66, 67]. A small study of sodium butyrate showed that treatment with intermittent doses, rather than continuous therapy, resulted in sustained increases in the levels of HbF, F-cells and total hemoglobin [61]. Despite initially promising results, the orally available sodium dimethyl butyrate (HQK-1001) did not produce a significant increase in HbF levels, but appeared to result in more pain episodes compared with placebo [67]. Pomalidomide, a third-generation immunomodulatory drug, induces a fetal-like erythroid differentiation program, leading to a reversion of γ-globin silencing in adult human erythroblasts. The transcription networks involved in γ-globin repression, including BCL11A, SOX6, IKZF1, KLF1, and LSD1 are selectively and differentially affected by pomalidomide [68]. In a transgenic mouse model of SCD, 8-weeks of treatment with pomalidomide induced modest increases of HbF, augmented erythropoiesis and preserved bone marrow function [69]. A dose-finding study of pomalidomide in patients with SCD, only available in abstract form, reported increases in HbF and total hemoglobin only at the highest dose or with greater than 6 months of exposure [70].
Other Antisickling and Hemoglobin Modifying Agents
Senicapoc, selectively blocks the calcium-activated, potassium efflux (Gardos) channel, and improves anemia and hemolysis in patients with SCD [71, 72]. However, a phase 3, randomized, placebo-controlled trial, showed no significant improvement in the rate of painful crises in patients treated with senicapoc compared to those on placebo [71]. It was suggested that senicapoc did not decrease the frequency of painful episodes in SCD due to the shift to a “more vaso-occlusive phenotype” with increased hemoglobin and possibly increased whole blood viscosity [73]. However, post-hoc analyses from the phase 3 trial showed no significant correlation between total hemoglobin or change from baseline hemoglobin and the crisis rate experienced by patients receiving senicapoc [71]. Magnesium decreases potassium and water loss by blocking the potassium-chloride co-transport channel in RBCs [11]. A phase 2 trial of hydroxyurea and magnesium pidolate, alone and in combination, in HbSC patients showed no effects of magnesium pidolate on its own [74]. In a multicenter, double-blind, placebo-controlled study of patients with HbSS or HbSβ0 thalassemia, intravenous magnesium sulfate did not shorten the length of stay, lessen opioid use or improve the quality of life in children who required inpatient hospitalization after failing emergency department management for pain [75].
Sanguinate (pegylated bovine carboxyhemoglobin) is a carbon monoxide releasing/oxygen transfer agent that was developed for the treatment of anemic and ischemic hypoxia. Due to its modification by polyethylene glycol and the antivasoconstrictive activity of carbon monoxide, sanguinate has demonstrated no vasoactivity in vivo [76], and appears to be safe in healthy individuals [77] and patients with stable sickle cell anemia [76]. Sanguinate has been granted orphan drug status and two randomized, double-blind placebo-controlled phase 2 studies evaluating its safety and efficacy in individuals with SCD experiencing vaso-occlusive crisis are ongoing (NCT02672540; NCT02411708). MP4CO (pegylated carboxyhemoglobin) markedly induced hepatic heme oxygenase-1 activity and inhibited NF-kB activation and microvascular stasis in sickle mice [78]. Although a phase 1 safety study has been completed (NCT01356485), a planned phase 2 study was withdrawn before enrollment, and the sponsor has ceased operations.
Voxelotor (previously called GBT440) is a novel, orally bioavailable small molecule which binds specifically to hemoglobin, with an approximately 150:1 RBC:plasma partition in various animal species, increases hemoglobin oxygen affinity and stabilizes the oxyhemoglobin state [79]. Voxelotor delays in vitro HbS polymerization and prevents sickling of RBCs. In a murine model of SCD, GBT440 extended the half-life of RBCs, reduced reticulocyte counts and prevented ex vivo RBC sickling [79]. Preliminary results suggest that voxelotor at 900 mg is well tolerated, with marked improvement in hemoglobin and reduction in clinical measures of hemolysis in adolescents with SCD [80]. These hematologic improvements were observed in patients on maximum tolerated dose of hydroxyurea. A phase 3, double-blind, randomized, placebo-controlled trial of voxelotor in patients with SCD is ongoing (NCT03036813). The primary endpoint is a change in hemoglobin, with key secondary outcomes, based on patient-reported outcomes. These endpoints are novel in the setting of SCD. In addition, a phase 2a, open-label, study in pediatric patients is ongoing (NCT02850406).
5-hydroxymethyl-2-furfural (5HMF, also called Aes-103) interacts allosterically with HbS, thereby increasing oxygen affinity and decreasing HbS polymerization and RBC sickling during hypoxia. Short-term administration of high doses in healthy volunteers and lung surgery patients was well tolerated [81, 82]. In addition, a phase 1 trial with oral doses up to 4,000 mg, in patients with SCD, some treated with hydroxyurea, showed no significant side effects [83]. However, a phase 2 clinical trial of Aes-103 in SCD was terminated early by the Sponsor due to unblinding between the study drug and placebo groups at the subject, site and Sponsor levels.
Anti-Adhesion Agents
Several drugs targeting adhesion of blood cells to the endothelium are being investigated as treatments for SCD (Table 2). Purified poloxamer 188, a nonionic block copolymer surfactant with hemorheologic and antithrombotic properties, was shown to significantly decrease the duration of pain episodes, especially in children and patients on hydroxyurea [84]. However, the decrease in the duration of pain episodes did not appear to be clinically meaningful. In another phase 3 study in children and adults with vaso-occlusive crisis, treatment with purified poloxamer 188 (vepoloxamer or MST-188) did not significantly reduce the duration of vaso-occlusive crises compared with placebo (NCT01737814) (https://www.prnewswire.com/news-releases/mast-therapeutics- reports-top-line-results-from-phase-3-study-in-sickle-cell-disease-300331289.html).
Table 2:
Ongoing Clinical Trials of Anti-Adhesion Agents in Sickle Cell Disease
Drug | Study Title | Mechanism of Action | Clinical Phase | NCT Number | Sponsor |
---|---|---|---|---|---|
Crizanlizumab | Pharmacokinetics and Pharmacodynamics Study of SEG101 (Crizanlizumab) in Adult Sickle Cell Disease (SCD) Patients With Vaso- Occlusive Crisis (VOC) |
Anti-adhesive (anti-P- selectin agent) |
Phase 2 | NCT03264989 | Novartis |
Rivipansel | Efficacy and Safety of Rivipansel (GMI- 1070) in the Treatment of Vaso- Occlusive Crisis in Hospitalized Subjects With Sickle Cell Disease |
Anti-adhesive (pan- selectin inhibitor) |
Phase 3 | NCT02187003 | Pfizer |
Sevuparin | Sevuparin Infusion for the Management of Acute VOC in Subjects With SCD |
Anti-adhesive (binds to P- and L-selectins, fibronectin, thrombospondin and von Willebrand factor) |
Phase 2 | NCT02515838 | Modus Therapeutics AB |
Crizanlizumab, an antibody against the adhesion molecule P-selectin, was evaluated in a double-blind, randomized, placebo-controlled, phase 2 trial, in 198 patients with SCD [25]. Treatment with high-dose crizanlizumab resulted in a significantly lower median crisis rate, significantly longer median times to the first and second crises, and a significantly lower median rate of uncomplicated crises per year compared with placebo. The adverse events that occurred in 10% or more of the patients in either active-treatment group and at a frequency that was at least twice as high as that in the placebo group were arthralgia, diarrhea, pruritus, vomiting, and chest pain. A phase 2 study to further evaluate the pharmacokinetics and pharmacodynamics of crizanlizumab (SEG101) in adult patients is ongoing (NCT03264989). Rivipansel sodium (formerly called GMI-1070), a small-molecule pan-selectin inhibitor that binds to E-, P- and L-selectin is being evaluated as treatment for acute vaso-occlusive episodes (NCT02187003). In a randomized, double-blind, adaptive, phase 2 trial, rivipansel was shown to shorten the time to resolution of vaso-occlusive crisis compared with placebo [85]. Although the absolute decreases in times to resolution of vaso-occlusive crisis (mean of 41 hours and median of 63 hours) appeared to be clinically meaningful, the differences were not statistically significant. However, treatment with rivipansel resulted in a large and statistically significant reduction in the mean cumulative intravenous opioid analgesic use compared with placebo.
In addition to their anticoagulant effects, heparins are known to inhibit adhesive interactions via P-selectin [86, 87]. Sevuparin is a derivative of low-molecular-weight heparin that retains the P-selectin-binding domain of heparin but largely lacks anticoagulant properties [88]. Sevuparin binds to P- and L-selectins, thrombospondin, fibronectin and von Willebrand factor and inhibits the adhesion of sickle RBCs to stimulated cultured endothelial cells in vitro [88]. In addition, sevuparin prevents vaso-occlusion and normalizes blood flow in a mouse model of SCD vaso-occlusion [88]. A phase 2 clinical trial of sevuparin for the management of acute vaso-occlusive crisis is ongoing (NCT02515838), the primary endpoint being the time to resolution of vaso-occlusive crisis. In a multicenter study with 253 patients, tinzaparin at its usual therapeutic dose was reported to decrease the total number of hospital days, the overall number of days with pain crisis and the number of days of the most severe pain scores compared with placebo [89]. However, it is uncertain whether the observed benefit of tinzaparin was due to its anticoagulant or its anti-adhesive effect. Furthermore, issues regarding standard of care and potential study bias make the study difficult to interpret [90]. A phase 2, pilot feasibility study of unfractionated heparin (NCT02098993) and a phase 3, randomized, placebo-controlled study of tinzaparin in acute chest syndrome are ongoing (NCT02580773).
Intravenous immunoglobulin (IVIG) reverses vaso-occlusion in sickle mice by rapidly inhibiting neutrophil adhesion to the endothelium and blocking interactions between neutrophils and RBCs [91]. IVIG blocks FcγRIII receptors, leading to inhibition of neutrophil adhesion, reduction of RBC capture by leukocytes, and reduced Mac-1 activity as a result of recruitment of Src homology 2–containing tyrosine phosphatase-1 [92]. Administration of IVIG to patients with SCD during acute vaso-occlusive crises in a phase 1 study resulted in decreased Mac-1 function from baseline and produced no significant adverse events at doses up to 800 mg/kg [93]. A phase 2 clinical trial of IVIG in patients during acute pain episodes is ongoing (NCT01757418).
Anti-Oxidant Agents
Increased oxidant stress plays a major role in the pathophysiology of SCD, and leads to disturbances of cell membranes, expression of adhesion molecules and hemolysis. In a phase 3, multicenter, double-blind, placebo-controlled trial, 230 patients with HbSS or HbSβ0-thalassemia were randomized to receive oral L-glutamine or placebo at a ratio of 2:1 for 48 weeks followed by a 3 week taper. Treatment with L-glutamine produced a significantly lower number of pain crises, hospitalizations, cumulative hospital days and number of patients affected by acute chest syndrome compared with placebo [94]. Adverse events in the L-glutamine treatment arm were similar to those observed in the placebo arm. Based on these data, L-glutamine was recently approved by the FDA for the reduction of acute complications of SCD in adult and pediatric patients 5 years and older (https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm566084.htm).
In an open label pilot study of oral N-acetylcysteine (NAC), 11 consecutive patients were randomly assigned to receive 1,200 mg or 2,400 mg NAC daily for 6 weeks [95]. There was an increase in whole blood glutathione levels and a decrease in erythrocyte outer membrane phosphatidylserine exposure, plasma levels of advanced glycation products and cell-free hemoglobin after 6 weeks of treatment in both groups. No patients experienced painful crises or other significant SCD-or NAC-related complications during the trial. In a randomized, placebo-controlled, double-blind trial, treatment with NAC at a dose of 600 mg twice daily for 6 months did not decrease the rate of SCD-related pain days per patient year compared with placebo [96]. In addition, there were no differences in the days with vaso-occlusive crises, admission days, number of admissions or days with home analgesic use. However, a per-protocol analysis performed in adherent patients revealed a significantly lower rate of vaso-occlusive crisis days in the NAC arm compared with placebo. Another phase 1/2 pilot study of NAC in patients with vaso-occlusive crisis is ongoing (NCT01800526). Ongoing studies of anti-oxidants in SCD are shown in Table 3.
Table 3:
Ongoing Clinical Trials of Anti-Oxidants and Anti-Inflammatory Agents in Sickle Cell Disease
Drug | Study Title | Mechanism of Action | Clinical Phase | NCT Number | Sponsor | |
---|---|---|---|---|---|---|
N-acetyl cysteine | A Pilot Study of N-acetylcysteine in Patients With Sickle Cell Disease (NACinSCD) |
Anti-oxidant (increased glutathione) |
Phase 1/2 | NCT01800526 | Bloodworks; University of Washington |
|
Omega-3 fatty Acids (Docosahexaenoic Acid (DHA) and Eicosapentaenoic Acid (EPA)) |
Omega-3 Fatty Acids in Sickle Cell Disease |
Anti-inflammatory | Phase 1/2 / | NCT02947100; | NIGMS; Thomas Jefferson University; Solutex GC, S.L. |
|
Azithromycin | Macrolide Therapy to Improve Forced Expiratory Volume in 1 Second in Adults With Sickle Cell Disease; A Pilot Study of Azithromycin Prophylaxis for Acute Chest Syndrome in Sickle Cell Disease |
Anti-inflamatory agent (antibiotic) |
Phase 1; Phase 1 |
NCT02960503; NCT02630394 |
Vanderbilt University; University of Mississippi Medical Center |
|
Canakinumab | Study of Efficacy, Safety and Tolerability of ACZ885 (Canakinumab) in Pediatric and Young Adult Patients With Sickle Cell Anemia |
Anti-inflammatory agent (anti-IL-1β) |
Phase 2 | NCT02961218 | Novartis Pharmaceuticals |
|
Omega-3 fatty acid (DHA) |
Sickle Cell Omega-3 Treatment Trial (SCOT Trial) (SCOT) |
Anti-inflammatory | Phase 3 | NCT02604368 | Sancillio and Company, Inc |
|
Atorvastatin | Effect of Atorvastatin on Endothelial Dysfunction and Albuminuria in Sickle Cell Disease (ENDO) |
Anti-inflammatory; pleiotropic (statin) |
Phase 2 | NCT01732718 | University of North Carolina, Chapel Hill |
|
Mometasone Furoate |
Inhaled Mometasone to Reduce Painful Episodes in Patients With Sickle Cell Disease (IMPROVE) |
Anti-inflammatory (steroid) |
Phase 2 | NCT02061202 | Icahn School of Medicine at Mount Sinai |
|
Montelukast | Phase 2 Study of Montelukast for the Treatment of Sickle Cell Anemia |
Anti-inflammatory (leukotriene receptor antagonist) |
Phase 2 | NCT01960413 | Vanderbilt University Medical Center |
Anti-Inflammatory Agents
Vaso-occlusion promotes tissue injury by reperfusion following ischemia-reperfusion, with the resultant inflammatory cascade amplified by the activation of CD1d-restricted iNKT cells. In addition, leukocytes, platelets, and multiple proinflammatory pathways appear to contribute to the pathophysiology of SCD. Several approaches to downregulate inflammatory pathways are being evaluated in patients with SCD (Table 3). Treatment of children and adolescents during acute pain episodes with methylprednisolone resulted in significantly shorter duration of inpatient analgesic therapy compared with placebo, although the patients who received methylprednisolone had more “rebound” pain episodes after treatment was discontinued [97]. Intravenous dexamethasone resulted in a significantly shorter hospital stay, prevented clinical deterioration and reduced the need for blood transfusion in patients with mild to moderately severe cases of acute chest syndrome when compared with placebo [98]. However, more patients treated with dexamethasone were re-admitted within 72 hours of discharge. A more recent study using tapered oral dexamethasone was terminated early due to slow accrual [99].
iNKT cell activation is downregulated by activation of the adenosine A2A receptor (A2AR), and adenosine reduces the activation of iNKT cells as well as cytokine production by iNKT cells through this receptor [100, 101, 102]. Sickle mice and patients with SCD have more activated iNKT cells than controls [103]. Blockade or depletion of iNKT cells with an A2AR agonist or an anti-iNKT cell antibody in mice decreased inflammation and tissue injury following ischemia-reperfusion [102, 103, 104]. Regadenoson, a partially selective A2AR agonist, was safe in children and adults in the steady state and during vaso-occlusive crises in a phase 1 study [105]. iNKT cells were more activated in patients with SCD in steady state than in control subjects, with more marked activation during vaso-occlusive crises. A 24-hour infusion of regadenoson during pain episodes significantly decreased phospho-NF-κΒ p65 activation in iNKT cells [105]. However, in a recently completed phase 2 study in children and adults experiencing pain crises, the proportion of subjects who demonstrated a reduction of greater than 30% in activated iNKT cells was not significantly different between the regadenoson and placebo arms [106]. Furthermore, no differences were observed in the length of hospital stay, mean total opioid use, or pain scores in both treatment groups. In an open-label, multi-center, single-ascending-dose study, treatment with NKTT120, an anti-iNKT cell monoclonal antibody, produced rapid, specific and sustained iNKT cell depletion without any infusional toxicity or attributed serious adverse events [106]. A randomized, placebo-controlled clinical trial of NKTT120 will be required to determine the safety and efficacy of long-term iNKT cell depletion.
Statins exhibit benefits independent of their cholesterol lowering effect. SCD is characterized by progressive vascular injury and endothelial dysfunction similar to that observed in atherosclerosis [107, 108, 109]. A pilot study showed that treatment with simvastatin was well tolerated and increased levels of nitric oxide metabolites and decreased levels of both C-reactive protein (CRP) and interleukin-6 in patients with SCD [110]. Treatment of 19 patients with a single daily dose of simvastatin for 3 months in an open label study resulted in a significant reduction in the frequency of pain episodes, oral analgesic use, and levels of circulating high sensitivity CRP, soluble E-selectin, soluble ICAM-1, soluble ICAM-3 and VEGF [111]. These results provide supporting clinical data for the conduct of a randomized, placebo-controlled trial of simvastatin in sickle cell anemia. A study of the effect of atorvastatin on endothelial dysfunction and albuminuria in sickle cell anemia is ongoing (NCT01732718).
The cysteinyl leukotrienes (CysLTs) are lipid inflammatory mediators that cause bronchoconstriction and play a role in leukocyte recruitment, endothelial cell adhesion, vasoconstriction, increased vascular permeability, and smooth muscle proliferation [112]. These pro-inflammatory and vascular effects provide biologic plausibility for a role of CysLTs in the process of vaso-occlusion [113]. Elevated urinary levels of leukotriene E4 are observed during the steady state, and are associated with an increased risk of pain crisis and acute chest syndrome [114, 115, 116]. The leukotriene receptor antagonist, montelukast, is being tested in a phase 2, randomized, placebo-controlled trial to evaluate its effect, in combination with hydroxyurea, on the level of soluble VCAM-1, lung function and forearm microvascular flow (NCT01960413). Zileuton targets leukotriene synthesis by inhibiting 5-lipoxygenase (5-LO). 5-LO causes increased production of leukotrienes, including leukotriene B4, a very potent chemoattractant and neutrophil and endothelial cell activator, and leukotriene E4, which is associated with lung inflammation. Zileuton has been shown to reduce activated neutrophil-mediated sickle RBC adhesion to rat pulmonary vasculature [117], and to reduce IL-13 production by murine splenic lymphocytes [118]. Zileuton has also been reported to increase HbF in erythroid progenitor cells via an L-arginine/NO-dependent mechanism [119]. A phase 1 trial of zileuton to examine its safety, pharmacokinetics and effect on inflammatory biomarkers in patients with SCD has been completed (NCT01136941), but the results have not yet been published.
Long-chain omega-3 polyunsaturated fatty acids, such as eicosapentaenoic acid (EPA [n-3]) and docosahexaenoic acid (DHA [n-6]), have beneficial effects in chronic inflammatory disorders. Patients with SCD have a high n-6/n-3 ratio in blood cells and plasma and this fatty acid abnormality may contribute to the chronic inflammatory state observed in SCD [120, 121]. Supplementation with omega-3 fatty acids improve cell membrane abnormalities, and decrease markers of coagulation activation, oxidative stress, pro-inflammatory cytokines and adhesive molecules [122, 123, 124, 125]. Furthermore, in a randomized, single center study, treatment with daily omega-3 capsules containing 277.8 mg DHA and 39.0 mg EPA significantly reduced the median rate of clinical vaso-occlusive events, severe anemia, blood transfusion requirements, and absence from school compared with placebo [126]. Top line results from a phase 2, randomized, double-blind, placebo-controlled study of children with SCD, showed that treatment with DHA resulted in a statistically significant change from baseline in blood cell membrane fatty acids concentration within 4 weeks (https://www.sancilio.com/altemia-achieves-successful-clinical-results-in-pediatric-patients-with-sickle-cell-disease-scd/). In addition, DHA was reported to produce a clinically meaningful reduction of pain crises, with statistically significant improvements in markers of coagulation, inflammation and adhesion after 8 weeks of treatment. No treatment-related serious adverse events were reported.
Anticoagulant Agents
There is abundant evidence that SCD represents a hypercoagulable state. In addition to an increased risk of thrombotic complications, patients exhibit increased thrombin generation, abnormal activation of fibrinolysis, decreased levels of natural anticoagulant proteins, activation of platelets, as well as increased tissue factor (TF) antigen and TF procoagulant activity in the non-crisis, “steady state” [21]. Increasing evidence suggest that coagulation activation may play a role in the pathophysiology of SCD. Data from mice that express hemoglobin S show that inhibition of TF activity effectively prevented the accelerated thrombus formation in a light/dye-induced model of cerebral microvascular thrombosis [131]. Inhibition of TF in another mouse model of SCD significantly reduced plasma levels of TAT, interleukin-6, soluble VCAM-1, and serum amyloid protein, as well as neutrophil infiltration in the lung [132]. More recently, reduction of prothrombin level to approximately 10% activity in sickle mice not only resulted in lower plasma levels of coagulation activation and inflammatory biomarkers, but also diminished early mortality, as well as damage to organs, including the lung, kidney, heart and liver, with no increase in bleeding complications [134].
Most of the published clinical studies of anticoagulant agents in SCD have been small, poorly controlled and of relatively low quality [135, 136, 137]. With the current availability of direct oral anticoagulants, an ongoing study is evaluating the effect of rivaroxaban in SCD (NCT02072668) (Table 4). A randomized, double-blind, placebo controlled study of SCD patients during acute pain episodes, reported clinical benefit for the low molecular weight heparin, tinzaparin, compared with placebo [89]. However, it is uncertain whether these reported beneficial effects were a result of the anticoagulant effect or the P-selectin blocking effect of tinzaparin. Two studies in acute chest syndrome, one a phase 2 feasibility study of therapeutic dose unfractionated heparin (NCT02098993) and another a phase 3 study of tinzaparin (NCT02580773), are ongoing.
Table 4:
Ongoing Clinical Trials of Antiplatelet Agents and Anticoagulants in Sickle Cell Disease
Drug | Study Title | Mechanism of Action | Clinical Phase | NCT Number | Sponsor |
---|---|---|---|---|---|
Ticagrelor | A Study to Assess the Effect of Ticagrelor in Reducing the Number of Days With Pain in Patients With Sickle Cell Disease (Hestia2) |
Antiplatelet agent | Phase 2 |
NCT02482298 | AstraZeneca |
Rivaroxaban | The Effect of Rivaroxaban in Sickle Cell Disease |
Anticoagulant | Phase 2 |
NCT02072668 | University of North Carolina, Chapel Hill |
Apixaban | Apixaban in Patients With Sickle Cell Disease |
Anticoagulant | Phase 3 | NCT02179177 | Duke University Medical Center |
Unfractionated heparin |
Feasibility Study of Unfractionated Heparin in Acute Chest Syndrome |
Anticoagulant; antiadhesive agent |
Phase 2 | NCT02098993 | University of Pittsburgh |
Tinzaparin | Therapeutic Anticoagulation Strategy for Acute Chest Syndrome (TASC) |
Anticoagulant; antiadhesive agent |
Phase 3 | NCT02580773 | Assistance Publique– Hopitaux de Paris |
Antiplatelet Agents
Platelets are activated in SCD and may contribute to disease pathophysiology [138, 139]. Multiple relatively small studies have evaluated the clinical effects of aspirin in SCD [140, 141, 142]. Although one study reported an increase in oxygen affinity, hemoglobin level and RBC life span [141], other studies showed only modest or no benefit for aspirin on hemoglobin levels or acute pain episodes [135, 140, 142]. A pilot study of eptifibatide showed that this glycoprotein IIb/IIIa inhibitor was safe when administered during acute pain episodes, but did not reduce the time to resolution of such pain episodes [143]. Ticlopidine, a P2Y12 ADP-receptor antagonist, reportedly decreased the number of pain episodes, the mean duration of pain episodes and the severity of such episodes in a multicenter study of 140 sickle cell anemia patients [144]. However, a more recent phase 3 trial of prasugrel, a newer generation P2Y12 receptor blocker, in children with SCD showed no significant reduction of the frequency of pain episodes [29]. A phase 2 study to investigate the efficacy of two different doses of ticagrelor has recently been completed and the results are awaited (NCT02482298).
Nitric Oxide and Related Agents
With the role of hemolysis in NO scavenging and subsequent endothelial dysfunction in SCD, several studies have evaluated the effect of NO and related agents as treatments for SCD. Inhaled NO gas for up to 72 hours did not decrease the median time to resolution of pain crisis, length of hospitalization, visual analog pain scale scores, cumulative opioid usage, or rate of acute chest syndrome compared with placebo in an adequately powered placebo-controlled trial [145]. Treatment of children hospitalized for acute pain episodes with L-arginine for 5 days or until discharge significantly decreased total parenteral opioid use and resulted in lower pain scores at discharge compared to placebo, although no significant difference in hospital length of stay was observed [146]. Sildenafil, a phosphodiesterase 5 inhibitor, increases NOmediated effects by inhibiting cyclic guanosine monophosphate (cGMP) degradation. A double-blind, placebo-controlled trial of sildenafil was stopped early due to a higher percentage of subjects experiencing serious adverse events in the sildenafil arm, predominantly hospitalization for pain, compared with placebo [147]. Furthermore, no evidence of a treatment effect of sildenafil was observed on 6-minute walk distance, echocardiography-derived tricuspid regurgitant jet velocity, or N-terminal pro-brain natriuretic peptide. Although myalgia and back pain are recognized side effects of chronic administration of sildenafil, evidence of a role for NO and cGMP in the processing of inflammatory and neuropathic pain suggests that the increased pain may be due to inhibition of cGMP degradation [148, 149]. A phase 2 multicenter study of riociguat, a soluble guanylate cycle stimulator, is ongoing (NCT02633397) (Table 5).
Table 5:
Ongoing Clinical Trials of Nitric Oxide-Related Drugs and Vasodilators in Sickle Cell Disease
Drug | Study Title | Mechanism of Action | Clinical Phase | NCT Number | Sponsor |
---|---|---|---|---|---|
Ambrisentan | The Role of Endothelin-1 in Sickle Cell Disease |
Vasodilator (endothelin receptor A antagonist) |
Phase 1 | NCT02712346 | Augusta University; Gilead Sciences; NHLBI |
Citrulline | A Phase 1 Study of Continuous Intravenous L-citrulline During Sickle Cell Pain Crisis or Acute Chest Syndrome |
Increased nitric oxide synthesis |
Phase 1 | NCT02697240 | University of Mississippi Medical Center |
Riociguat | A Multi-Center Study of Riociguat in Patients With Sickle Cell Diseases |
Vasodilator (soluble guanylate cyclase stimulator) |
Phase 2 | NCT02633397 | University of Pittsburgh |
Sodium Nitrite | Topical Sodium Nitrite in Sickle Cell Disease and Leg Ulcers |
Vasodilator (local nitric oxide donor) |
Phase 2 | NCT02863068 | Montefiore Medical Center; Food and Drug Administration |
Arginine | Arginine Therapy for the Treatment of Pain in Children With Sickle Cell Disease (R34 pK/PD); Arginine Therapy for Sickle Cell Disease Pain |
Increased nitric oxide synthesis |
Phase 1/2; Phase 2 |
NCT02447874; NCT02536170 |
Emory University |
Macitentan | Macitentan in Pulmonary Hypertension of Sickle Cell Disease (MENSCH) |
Vasodilator (endothelin receptor antagonist) |
Phase 1 | NCT02651272 | Boston University |
IMR-687 | A Study of IMR-687 in Adult Patients With Sickle Cell Anaemia (Homozygous HbSS or Sickle-β0 Thalassemia) |
Phosphodiesterase-9 inhibitor |
Phase 2 | NCT03401112 | Imara, Inc |
Losarían | Preventing Sickle Cell Kidney Disease | Vasodilator (angiotensin receptor blocker) |
Phase 2 | NCT02373241 | University of Alabama at Birmingham |
Clinical Endpoints and Biomarkers in Sickle Cell Disease
Despite the fact that SCD involves multiple organs and systems, most drug trials have focused on acute pain episodes as the primary clinical endpoint. This is not surprising as these painful episodes represent the most frequent reason for medical contact by patients with SCD. However, with the complex pathophysiology of vaso-occlusive crises, combined with the difficulty in accurately quantifying these episodes, the emphasis on pain crisis as a clinical endpoint may be severely limiting. Most studies of acute pain episodes have evaluated only those episodes requiring healthcare utilization, i.e. episodes requiring visits to emergency departments, hospitalizations or other physician contact at which time patients are treated with parenteral or oral analgesics [25, 26, 71]. With the evidence that the bulk of pain episodes are treated at home [150], it is important that studies incorporate these episodes which substantially contribute to disease morbidity and decreased quality of life in SCD [29]. These pain episodes that do not require healthcare utilization may be measured using patient-reported instruments, such as in the ongoing phase 3 trial of voxelotor.
Clinical trials in SCD should also include such complications as ischemic stroke, chronic kidney disease, priapism, pulmonary hypertension, leg ulcers and bone disease as clinical endpoints. However, as these events usually occur less frequently than acute pain episodes, studying each of these complications may require larger numbers of study subjects and/or a longer duration in order to observe and quantify treatment effects. Multiple relatively large studies have evaluated overt and silent stroke as clinical endpoints in SCD in the setting of RBC transfusion therapy [27, 151, 152]. Despite the challenge of drug trials in a setting where chronic RBC transfusion therapy is the accepted standard of care, the successful conduct of the SWITCH and TWITCH trials [45, 153], as well as the ongoing SPRING trial (NCT02560935) in sub-Saharan Africa suggests that the utility of stroke or clinical endpoints other than acute pain episodes is feasible.
Patient-reported outcomes, including health-related quality of life (HRQoL), are suitable endpoints in drug development [154]. Significant impairment of HRQoL is present in SCD patients at baseline and in association with multiple complications [155]. Low hemoglobin and HbF levels are also associated with impaired HRQoL in adult patients [156]. The use of hydroxyurea is associated with improved HRQoL in individuals who are high HbF responders [157]. SCD-specific HRQoL instruments have been developed and provide opportunities to explore the impact of the disease and its treatment on patients [156]. Exercise capacity is also a clinical endpoint for drug development [158, 159]. Decreased exercise capacity is common in SCD due to anemia, cardiac disease, restrictive and obstructive lung disease, pulmonary vascular disease, as well as bone and muscle abnormalities. The 6-minute walk test is a useful measure of functional capacity, and is decreased with elevated tricuspid regurgitant jet velocity and pulmonary hypertension in SCD [160, 161, 162].
Surrogate endpoints may be targets for drug development. A biomarker is defined as a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention [163]. Although multiple laboratory biomarkers are described in SCD [164], many of these are of limited clinical value and require further assessment in prospective studies to validate their prognostic importance before they are acceptable as surrogate endpoints. Increased transcranial Doppler ultrasound (TCD) velocity of at least 200 cm/s is associated with an increased risk of stroke in children with sickle cell anemia [152, 165], and is a validated surrogate endpoint in children [151, 152]. Increased tricuspid regurgitant jet velocity is associated with increased mortality and identifies adult patients at higher risk of having pulmonary hypertension defined by right heart catheterization [7, 162, 166, 167, 168], but it has not been validated as a suitable surrogate endpoint in SCD. Although SCD is characterized by a chronic anemia, increase in hemoglobin may not always be associated with clinical benefit as was observed in the phase 3 trial of senicapoc [71]. The increase in hemoglobin following hydroxyurea therapy is associated with decreased frequency of vaso-occlusive complications as well as RBC transfusion requirements [26, 28]. However, decreased hemolysis with subsequent increases in hemoglobin is likely to be associated with improvements in fatigue, and may be associated with a decreased risk of such complications as gallstones, leg ulcers, chronic kidney disease and possibly mortality risk [7, 14, 169]. Increase in hemoglobin is the primary endpoint in an ongoing phase 3 trial of voxelotor (NCT03036813), with evaluation of patient-reported outcomes as key secondary endpoints.
Clinical Trials in Sub-Saharan Africa and India
More than 300,000 infants with sickle cell anemia are born worldwide every year, but less than 1% of these births occur in North America and Europe where the vast majority of SCD research is undertaken [170]. As a result of scarce healthcare resources as well as limited awareness by healthcare providers and the general public in resource-poor countries, substantial early mortality rates are observed in SCD populations in these countries [171]. Whilst much needs to be done to decrease the morbidity and mortality due to SCD in low-income nations, it is important that new drugs are tested in these environments, in parallel with such testing in high- and middle-income nations. Hydroxyurea has been shown to be efficacious in resource-rich nations [26, 28, 45]. Until recently, there was substantial concern regarding the safety of hydroxyurea in the setting of a high burden of infectious disease and malnutrition. With demonstration of the safety of hydroxyurea in sub-Saharan Africa [172], multiple studies evaluating its efficacy, including the REACH (NCT01966731) and SPRING (NCT02560935) trials, are ongoing. The present enthusiasm for the development of new drugs for SCD provides an opportunity to involve sites in low- and middle-income nations, particularly sub-Saharan Africa and India, which have the highest burden of disease, in these clinical studies. Not only does this offer patients the opportunity to participate in cutting-edge research to assess the safety and efficacy of novel drugs, it also contributes substantially to capacity building by improving local infrastructure, strengthening of health systems and training of local providers.
Conclusion
Although a rare disease in the U.S and Europe, SCD is highly prevalent worldwide. There have been significant advances in our understanding of the pathophysiology of SCD over the past several decades. With its high morbidity and mortality, improved treatments are required, not only to prevent the occurrence of complications, but also to shorten the duration of acute complications. Hydroxyurea, and more recently, L-glutamine, are approved for the treatment of sickle cell anemia. An improved understanding of the pathophysiology of SCD, combined with the success of several recently approved drugs for other orphan diseases has increased interest in the development of drugs for SCD. Drugs that are being evaluated include hemoglobin F inducers, anti-sickling agents, anti-adhesive agents, anti-inflammatory agents, antioxidants, anticoagulants and anti-platelet agents. The enthusiasm for development of new drugs offers hope for the development of multiple pharmacotherapies for the treatment of SCD-related complications.
Expert Opinion
Despite the success of allogeneic bone marrow transplantation and the curative potential of gene therapy, these treatments are not widely available, especially in resource-poor countries. The development of drug therapies for SCD that are safe, effective and affordable is highly warranted. The increased number of drugs in testing offers hope for the availability of more pharmacologic therapies for SCD. With its complex pathophysiology, it is unlikely that individual drugs will ameliorate all of the complications of this disease. The availability of multiple drugs offers an opportunity for individualized therapy based on the presence of SCD-related complications, as well as for combination drug therapy based on different mechanisms of action and side effect profiles. It is important that clinical trials of novel drugs for SCD involve individuals in resource-poor countries, where the burden of disease is high. In addition to acute pain episodes, evaluation of the effect of novel agents on other SCD-related clinical complications, PROs, exercise capacity and validated surrogate endpoints is warranted.
Article Highlights.
While multiple agents have been tested, only two drugs are approved by regulatory agencies for treatment of sickle cell disease (SCD)
The development of effective drug therapies for SCD is based on an adequate understanding of its pathophysiology
Multiple drugs, including anti-sickling agents, anti-adhesive agents, antiinflammatory agents, anti-oxidants, anticoagulants and anti-platelet agents are being evaluated as treatments for SCD
The development of drugs with different mechanisms of action offers opportunities for combination and personalized therapy
In addition to acute pain crisis, the evaluation of other SCD-related complications, exercise capacity, patient reported outcomes and validated surrogate endpoints are necessary to advance drug development in SCD
The present enthusiasm for drug development provides an opportunity to involve sites in resource-limited countries, which have the highest burden of SCD, in trials of novel drug therapies.
Acknowledgments
Funding
This work is supported by the National Heart, Lung, and Blood Institute U01HL117659 (KIA), the Food and Drug Administration R01FD006030 (KIA, PCD) and the North Carolina State Sickle Cell Program (KIA).
Footnotes
Declaration of Interests
KIA has served on clinical advisory boards and/or served as a consultant for Global Blood Therapeutics, Modus Therapeutics, Novartis and Bioverativ. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
REFERENCES
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
- 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. doi: 10.1371/journal.pmed.1001484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.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. June;173(6):927–37. doi: 10.1111/bjh.14015. [DOI] [PubMed] [Google Scholar]
- 3.Quinn CT, Rogers ZR, McCavit TL, et al. Improved survival of children and adolescents with sickle cell disease. Blood. 2010. April 29;115(17):3447–52. doi: 10.1182/blood-2009-07-233700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.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. July;92(7):905–12. [DOI] [PubMed] [Google Scholar]
- 5.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. May;89(5):530–5. doi: 10.1002/ajh.23683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gardner K, Douiri A, Drasar E, et al. Survival in adults with sickle cell disease in a high-income setting. Blood. 2016. September 8;128(10):1436–8. doi: 10.1182/blood-2016-05-716910. [DOI] [PubMed] [Google Scholar]
- 7.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. April;102(4):626–636. doi: 10.3324/haematol.2016.153791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.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 North Am. 2014. December;28(6):1171–85. doi: 10.1016/j.hoc.2014.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hsieh MM, Fitzhugh CD, Weitzel RP, et al. Nonmyeloablative HLA-matched sibling allogeneic hematopoietic stem cell transplantation for severe sickle cell phenotype. JAMA. 2014. July 2;312(1):48–56. doi: 10.1001/jama.2014.7192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ribeil JA, Hacein-Bey-Abina S, Payen E, et al. Gene Therapy in a Patient with Sickle Cell Disease. N Engl J Med. 2017. March 2;376(9):848–855. doi: 10.1056/NEJMoa1609677. [DOI] [PubMed] [Google Scholar]
- 11.Bunn HF. Pathogenesis and treatment of sickle cell disease. N Engl J Med. 1997. September 11;337(11):762–9. doi: 10.1056/NEJM199709113371107. [DOI] [PubMed] [Google Scholar]
- 12. •.Rees DC, Williams TN, Gladwin MT. Sickle-cell disease. Lancet. 2010. December 11;376(9757):2018–31. doi: 10.1016/s0140-6736(10)61029-x. eng.Reference 12 is an excellent review of the clinical manifestations, pathophysiology and management of sickle cell disease [DOI] [PubMed] [Google Scholar]
- 13.Lawson V Turned on by danger: activation of CD1d-restricted invariant natural killer T cells. Immunology. 2012. September;137(1):20–7. doi: 10.1111/j.1365-2567.2012.03612.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kato GJ, Steinberg MH, Gladwin MT. Intravascular hemolysis and the pathophysiology of sickle cell disease. J Clin Invest. 2017. March 1;127(3):750–760. doi: 10.1172/JCI89741. [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. December;8(12):1383–9. doi: 10.1038/nm799. [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. November 15;78(10):2753–8. [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. July;96(1):60–8. doi: 10.1172/JCI118074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Palmer RM, Ashton DS, Moncada S. Vascular endothelial cells synthesize nitric oxide from L-arginine. Nature. 1988. June 16;333(6174):664–6. doi: 10.1038/333664a0. [DOI] [PubMed] [Google Scholar]
- 19.Schaer DJ, Buehler PW, Alayash AI, et al. Hemolysis and free hemoglobin revisited: exploring hemoglobin and hemin scavengers as a novel class of therapeutic proteins. Blood. 2013. February 21;121(8):1276–84. doi: 10.1182/blood-2012-11-451229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Manwani D, Frenette PS. Vaso-occlusion in sickle cell disease: pathophysiology and novel targeted therapies. Blood. 2013. doi: 10.1182/blood-2013-05-498311. [DOI] [PMC free article] [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 reviews. 2016. July;30(4):245–56. doi: 10.1016/j.blre.2015.12.003. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pauling L, Itano HA, et al. Sickle cell anemia a molecular disease. Science. 1949. November 25; 110(2865):543–8. [DOI] [PubMed] [Google Scholar]
- 23.Lobner K, Lanzkron S, MHS, et al. NIH and National Foundation Expenditures For Sickle Cell Disease and Cystic Fibrosis Are Associated With Pubmed Publications and FDA Approvals. Blood. 2013;122(21):1739–1739. [Google Scholar]
- 24.Humphries C Drug development: a complicated path. Nature. 2014. November 13;515(7526):S4–5. doi: 10.1038/515S4a. [DOI] [PubMed] [Google Scholar]
- 25. ••.Ataga KI, Kutlar A, Kanter J, et al. Crizanlizumab for the Prevention of Pain Crises in Sickle Cell Disease. N Engl J Med. 2017. February 2;376(5):429–439. doi: 10.1056/NEJMoa1611770. Reference 25 is an important study that demonstrated the efficacy of the anti-P-selectin antibody, crizanlizumab, in decreasing the frequency of acute painful crisis in sickle cell disease [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. ••.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. May 18;332(20):1317–22. doi: 10.1056/NEJM199505183322001. Reference 26 is a landmark study that demonstrated the efficacy of hydroxyurea in adult patients with sickle cell anemia.2 [DOI] [PubMed] [Google Scholar]
- 27.DeBaun MR, Gordon M, McKinstry RC, et al. Controlled trial of transfusions for silent cerebral infarcts in sickle cell anemia. N Engl J Med. 2014. August 21;371(8):699–710. doi: 10.1056/NEJMoa1401731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. •.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. May 14;377(9778):1663–72. doi: 10.1016/S0140-6736(11)60355-3. Reference 28 is an important study that confirmed the effect of hydroxyurea on vaso-occlusive complications in children with sickle cell anemia [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. •.Heeney MM, Hoppe CC, Rees DC. Prasugrel for Sickle Cell Vaso-Occlusive Events. N Engl J Med. 2016. July 14;375(2):185–6. doi: 10.1056/NEJMc1603499. Reference 29 was a phase 3 trial that evaluated the efficacy of prasugrel on vaso-occlusive events in children with sickle cell anemia. Study participants were recruited from 13 countries, including countries in sub-Saharan Africa [DOI] [PubMed] [Google Scholar]
- 30.Eaton WA, Bunn HF. Treating sickle cell disease by targeting HbS polymerization. Blood. 2017. May 18;129(20):2719–2726. doi: 10.1182/blood-2017-02-765891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.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. July 15;84(2):643–9. [PubMed] [Google Scholar]
- 32.Platt OS. Sickle cell paths converge on hydroxyurea. Nat Med. 1995. April;1(4):307–8. [DOI] [PubMed] [Google Scholar]
- 33.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. June 9;330(23):1639–44. doi: 10.1056/NEJM199406093302303. [DOI] [PubMed] [Google Scholar]
- 34.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. September;36(9):1057–72. doi: 10.1016/j.exphem.2008.06.014. [DOI] [PubMed] [Google Scholar]
- 35.Alcain FJ, Low H, Crane FL, et al. Iron chelators hydroxyurea and bathophenanthroline disulfonate inhibit DNA synthesis by different pathways. Biochem Mol Biol Int. 1994. September;34(2):273–9. [PubMed] [Google Scholar]
- 36.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. January;111(2):231–9. doi: 10.1172/JCI16672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Huang J, Yakubu M, Kim-Shapiro DB, et al. Rat liver-mediated metabolism of hydroxyurea to nitric oxide. Free Radic Biol Med. 2006. May 1;40(9):1675–81. doi: 10.1016/j.freeradbiomed.2006.01.002. [DOI] [PubMed] [Google Scholar]
- 38.Gladwin MT, Shelhamer JH, Ognibene FP, et al. Nitric oxide donor properties of hydroxyurea in patients with sickle cell disease. Br J Haematol. 2002. February;116(2):436–44. [DOI] [PubMed] [Google Scholar]
- 39.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. June;55(6):1006–10. [DOI] [PubMed] [Google Scholar]
- 40.McGann PT, Ware RE. Hydroxyurea for sickle cell anemia: what have we learned and what questions still remain? Curr Opin Hematol. 2011. May;18(3):158–65. doi: 10.1097/MOH.0b013e32834521dd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bridges KR, Barabino GD, Brugnara C, et al. A multiparameter analysis of sickle erythrocytes in patients undergoing hydroxyurea therapy. Blood. 1996. December 15;88(12):4701–10. [PubMed] [Google Scholar]
- 42.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. May;109(2):322–7. [DOI] [PubMed] [Google Scholar]
- 43.Odievre MH, Bony V, Benkerrou M, et al. Modulation of erythroid adhesion receptor expression by hydroxyurea in children with sickle cell disease. Haematologica. 2008. April;93(4):502–10. doi: 10.3324/haematol.12070. [DOI] [PubMed] [Google Scholar]
- 44.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. April;96(4):534–42. doi: 10.3324/haematol.2010.026740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.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. February 13;387(10019):661–70. doi: 10.1016/S0140-6736(15)01041-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bartolucci P, Habibi A, Stehle T, et al. Six Months of Hydroxyurea Reduces Albuminuria in Patients with Sickle Cell Disease. J Am Soc Nephrol. 2016. June;27(6):1847–53. doi: 10.1681/ASN.2014111126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.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. June;161(6):852–60. doi: 10.1111/bjh.12323. [DOI] [PubMed] [Google Scholar]
- 48.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. April 2;289(13):1645–51. doi: 10.1001/jama.289.13.1645. [DOI] [PubMed] [Google Scholar]
- 49.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. June;85(6):403–8. doi: 10.1002/ajh.21699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. •.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. March 25;115(12):2354–63. doi: 10.1182/blood-2009-05-221333. Reference 50 is a prospective study which suggests beneficial effect of hydroxyurea on survival in patients with sickle cell disease [DOI] [PubMed] [Google Scholar]
- 51.Dehury S, Purohit P, Patel S, et al. Low and fixed dose of hydroxyurea is effective and safe in patients with HbSbeta(+) thalassemia with IVS1–5(G-->C) mutation. Pediatr Blood Cancer. 2015. June;62(6):1017–23. doi: 10.1002/pbc.25391. [DOI] [PubMed] [Google Scholar]
- 52.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. October;50(10):929–33. [DOI] [PubMed] [Google Scholar]
- 53.Berthaut I, Bachir D, Kotti S, et al. Adverse effect of hydroxyurea on spermatogenesis in patients with sickle cell anemia after six months of treatment. Blood. 2017. doi: 10.1182/blood-2017-03-771857. [DOI] [PubMed] [Google Scholar]
- 54.Saunthararajah Y, Molokie R, Saraf S, et al. Clinical effectiveness of decitabine in severe sickle cell disease. Br J Haematol. 2008. April;141(1):126–9. doi: 10.1111/j.1365-2141.2008.07027.x. [DOI] [PubMed] [Google Scholar]
- 55.Jh Beumer , Eiseman JL, Parise RA, et al. Modulation of gemcitabine (2’,2’- difluoro-2’-deoxycytidine) pharmacokinetics, metabolism, and bioavailability in mice by 3,4,5,6-tetrahydrouridine. Clin Cancer Res. 2008. June 1;14(11):3529–35. doi: 10.1158/1078-0432.CCR-07-4885. [DOI] [PubMed] [Google Scholar]
- 56.Camiener GW, Smith CG. Studies of the enzymatic deamination of cytosine arabinoside. I. Enzyme distribution and species specificity. Biochem Pharmacol. 1965. October;14(10):1405–16. [DOI] [PubMed] [Google Scholar]
- 57.DeSimone J, Heller P, Molokie RE, et al. Tetrahydrouridine, cytidine analogues, and hemoglobin F. Am J Hematol. 1985. March;18(3):283–8. [DOI] [PubMed] [Google Scholar]
- 58.Ebrahem Q, Mahfouz RZ, Ng KP, et al. High cytidine deaminase expression in the liver provides sanctuary for cancer cells from decitabine treatment effects. Oncotarget. 2012. October;3(10):1137–45. doi: 10.18632/oncotarget.597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Neil GL, Moxley TE, Kuentzel SL, et al. Enhancement by tetrahydrouridine (NSC-112907) of the oral activity of 5-azacytidine (NSC-102816) in L1210 leukemic mice. Cancer Chemother Rep. 1975. May-Jun;59(3):459–65. [PubMed] [Google Scholar]
- 60. •.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. September;14(9):e1002382. doi: 10.1371/journal.pmed.1002382. Reference 60 reports on the safety and efficacy of oral tetrahydrouridine and decitabine in high-risk patients with sickle cell disease [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Atweh GF, Sutton M, Nassif I, et al. Sustained induction of fetal hemoglobin by pulse butyrate therapy in sickle cell disease. Blood. 1999. March 15;93(6):1790–7. [PMC free article] [PubMed] [Google Scholar]
- 62.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. March 25;115(12):2483–90. doi: 10.1182/blood-2009-02-204990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.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. June 30;850:87–99. [DOI] [PubMed] [Google Scholar]
- 64.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. November;87(11):1017–21. doi: 10.1002/ajh.23306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.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. November;88(11):E255–60. doi: 10.1002/ajh.23533. [DOI] [PubMed] [Google Scholar]
- 66.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. June 4;125(23):3668–9. doi: 10.1182/blood-2015-03-635391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.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. July;89(7):709–13. doi: 10.1002/ajh.23725. [DOI] [PubMed] [Google Scholar]
- 68.Dulmovits BM, Appiah-Kubi AO, Papoin J, et al. Pomalidomide reverses gammaglobin silencing through the transcriptional reprogramming of adult hematopoietic progenitors. Blood. 2016. March 17;127(11):1481–92. doi: 10.1182/blood-2015-09-667923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.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. July 28;118(4):1109–12. doi: 10.1182/blood-2010-11-319137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kutlar ASP, Meiler SW, et al. Pomalidomide in Sickle cell disease: Phase 1 study of a novel anti-switching agent [abstract]. Blood. 2013;122(Abstract 777):21. [Google Scholar]
- 71.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. April;153(1):92–104. doi: 10.1111/j.1365-2141.2010.08520.x. [DOI] [PubMed] [Google Scholar]
- 72.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. April 15;111(8):3991–7. doi: 10.1182/blood-2007-08-110098. [DOI] [PubMed] [Google Scholar]
- 73.Castro OL, Gordeuk VR, Gladwin MT, et al. Senicapoc trial results support the existence of different sub-phenotypes of sickle cell disease with possible drug-induced phenotypic shifts. Br J Haematol. 2011. December;155(5):636–8. doi: 10.1111/j.1365-2141.2011.08758.x. [DOI] [PubMed] [Google Scholar]
- 74.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. March;152(6):771–6. doi: 10.1111/j.1365-2141.2010.08523.x. [DOI] [PubMed] [Google Scholar]
- 75.Badaki-Makun O, Scott JP, Panepinto JA, et al. Intravenous magnesium for pediatric sickle cell vaso-occlusive crisis: methodological issues of a randomized controlled trial. Pediatr Blood Cancer. 2014. June;61(6):1049–54. doi: 10.1002/pbc.24925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.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. Jan-Mar;39(1):20–27. doi: 10.1016/j.bjhh.2016.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Misra H, Lickliter J, Kazo F, et al. PEGylated carboxyhemoglobin bovine (SANGUINATE): results of a phase I clinical trial. Artif Organs. 2014. August;38(8):702–7. doi: 10.1111/aor.12341. [DOI] [PubMed] [Google Scholar]
- 78.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. October 10;122(15):2757–64. doi: 10.1182/blood-2013-02-486282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.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. October;175(1):141–53. doi: 10.1111/bjh.14214. [DOI] [PubMed] [Google Scholar]
- 80.Hoppe CI,AC, Brown C, Wang W, Gordeuk V, Liem R, Woods G, Piccone CM, Fong E, Balaratnam G, Dixon S, Tonda ME, Washington CB, Yaron Y, Lehrer J. Initial Results from a Cohort in a Phase 2a Study (GBT440–007) Evaluating Adolescents with Sickle Cell Disease Treated with Multiple Doses of GBT440, a HbS Polymerization Inhibitor [Abstract 689 - Oral presentation]. Blood. 2017;ASH Annual Meeting. [Google Scholar]
- 81.Gatterer H, Greilberger J, Philippe M, et al. Short-term supplementation with alpha-ketoglutaric acid and 5-hydroxymethylfurfural does not prevent the hypoxia induced decrease of exercise performance despite attenuation of oxidative stress. Int J Sports Med. 2013. January;34(1):1–7. doi: 10.1055/s-0032-1312584. [DOI] [PubMed] [Google Scholar]
- 82.Matzi V, Lindenmann J, Muench A, et al. The impact of preoperative micronutrient supplementation in lung surgery. A prospective randomized trial of oral supplementation of combined alpha-ketoglutaric acid and 5- hydroxymethylfurfural. Eur J Cardiothorac Surg. 2007. November;32(5):776–82. doi: 10.1016/j.ejcts.2007.07.016. [DOI] [PubMed] [Google Scholar]
- 83.Lawrence MP, Mendelsohn LG, Saiyed R, et al. Phase 1 Clinical Trial Of The Candidate Anti-Sickling Agent Aes-103 In Adults With Sickle Cell Anemia. Blood. 2013;122(21):1009–1009. [Google Scholar]
- 84.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. November 7;286(17):2099–106. [DOI] [PubMed] [Google Scholar]
- 85. •.Telen MJ, Wun T, McCavit TL, et al. Randomized phase 2 study of GMl-1070 in SCD: reduction in time to resolution of vaso-occlusive events and decreased opioid use. Blood. 2015. April 23;125(17):2656–64. doi: 10.1182/blood-2014-06-583351. Reference 85 reports on the effect of GMI-1070 (rivipansel) on the duration of acute pain episodes and intravenous opioid analgesic use in patients with sickle cell disease [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.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. November 15;104(10):3378–85. doi: 10.1182/blood-2004-02-0713. [DOI] [PubMed] [Google Scholar]
- 87.Matsui NM, Varki A, Embury SH. Heparin inhibits the flow adhesion of sickle red blood cells to P-selectin. Blood. 2002. November 15;100(10):3790–6. doi: 10.1182/blood-2002-02-0626. [DOI] [PubMed] [Google Scholar]
- 88.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. December;175(5):935–948. doi: 10.1111/bjh.14303. [DOI] [PubMed] [Google Scholar]
- 89.Qari MH, Aljaouni SK, Alardawi MS, et al. Reduction of painful vaso-occlusive crisis of sickle cell anaemia by tinzaparin in a double-blind randomized trial. Thromb Haemost. 2007. August;98(2):392–6. [PubMed] [Google Scholar]
- 90.van Zuuren EJ, Fedorowicz Z. Low-molecular-weight heparins for managing vaso-occlusive crises in people with sickle cell disease. Cochrane Database Syst Rev. 2013. June 12(6):CD010155. doi: 10.1002/14651858.CD010155.pub2. [DOI] [PubMed] [Google Scholar]
- 91.Chang J, Shi PA, Chiang EY, et al. Intravenous immunoglobulins reverse acute vaso-occlusive crises in sickle cell mice through rapid inhibition of neutrophil adhesion. Blood. 2008. January 15;111(2):915–23. doi: 10.1182/blood-2007-04-084061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Jang JE, Hidalgo A, Frenette PS. Intravenous immunoglobulins modulate neutrophil activation and vascular injury through FcgammaRIII and SHP-1. Circ Res. 2012. April 13;110(8):1057–66. doi: 10.1161/CIRCRESAHA.112.266411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.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. May;90(5):381–5. doi: 10.1002/ajh.23956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Niihara Y, Koh HA, Tran L, et al. A Phase 3 Study of L-Glutamine Therapy for Sickle Cell Anemia and Sickle ß0-Thalassemia. Blood. 2014;124(21):86–86. [Google Scholar]
- 95.Nur E, Brandjes DP, Teerlink T, et al. N-acetylcysteine reduces oxidative stress in sickle cell patients. Ann Hematol. 2012. July;91(7):1097–105. doi: 10.1007/s00277-011-1404-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.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. 2017. June 23. doi: 10.1111/bjh.14809. eng. [DOI] [PubMed] [Google Scholar]
- 97.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. March 17;330(11):733–7. doi: 10.1056/NEJM199403173301101. [DOI] [PubMed] [Google Scholar]
- 98.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. November 1;92(9):3082–9. [PubMed] [Google Scholar]
- 99.Quinn CT, Stuart MJ, Kesler K, et al. Tapered oral dexamethasone for the acute chest syndrome of sickle cell disease. Br J Haematol. 2011. October;155(2):263–7. doi: . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Linden J Regulation of leukocyte function by adenosine receptors. Adv Pharmacol. 2011;61:95–114. doi: 10.1016/B978-0-12-385526-8.00004-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Nowak M, Lynch L, Yue S, et al. The A2aR adenosine receptor controls cytokine production in iNKT cells. Eur J Immunol. 2010. March;40(3):682–7. doi: 10.1002/eji.200939897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.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. December 2;116(23):5010–20. doi: 10.1182/blood-2010-06-290643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Wallace KL, Marshall MA, Ramos SI, et al. NKT cells mediate pulmonary inflammation and dysfunction in murine sickle cell disease through production of IFN-gamma and CXCR3 chemokines. Blood. 2009. July 16;114(3):667–76. doi: 10.1182/blood-2009-02-205492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Pei H, Schaub R, Scheuplein F, et al. Antibody Mediated Depletion of iNKT Cells Protects Against Hypoxia-Induced Pulmonary Injury in a Murine Model of Sickle Cell Disease. Blood. 2014;124(21):2697–2697. [Google Scholar]
- 105.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. April 25;121(17):3329–34. doi: 10.1182/blood-2012-11-465963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.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. September 12;1(20):1645–1649. doi: 10.1182/bloodadvances.2017009613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Belhassen L, Pelle G, Sediame S, et al. Endothelial dysfunction in patients with sickle cell disease is related to selective impairment of shear stress-mediated vasodilation. Blood. 2001. March 15;97(6):1584–9. [DOI] [PubMed] [Google Scholar]
- 108.de Montalembert M, Aggoun Y, Niakate A, et al. Endothelial-dependent vasodilation is impaired in children with sickle cell disease. Haematologica. 2007. December;92(12):1709–10. doi: 10.3324/haematol.11253. [DOI] [PubMed] [Google Scholar]
- 109.Eberhardt RT, McMahon L, Duffy SJ, et al. Sickle cell anemia is associated with reduced nitric oxide bioactivity in peripheral conduit and resistance vessels. Am J Hematol. 2003. October;74(2):104–11. doi: 10.1002/ajh.10387. [DOI] [PubMed] [Google Scholar]
- 110.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. June;153(5):655–63. doi: 10.1111/j.1365-2141.2010.08480.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.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. May;177(4):620–629. doi: 10.1111/bjh.14580. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Peters-Golden M, Henderson WR, Jr., Leukotrienes. N Engl J Med. 2007. November 1;357(18):1841–54. doi: 10.1056/NEJMra071371. [DOI] [PubMed] [Google Scholar]
- 113.Knight-Perry J, DeBaun MR, Strunk RC, et al. Leukotriene pathway in sickle cell disease: a potential target for directed therapy. Expert Rev Hematol. 2009. February;2(1):57–68. doi: 10.1586/17474086.2.1.57. [DOI] [PubMed] [Google Scholar]
- 114.Field JJ, Krings J, White NL, et al. Urinary cysteinyl leukotriene E(4) is associated with increased risk for pain and acute chest syndrome in adults with sickle cell disease. Am J Hematol. 2009. March;84(3):158–60. doi: 10.1002/ajh.21348. [DOI] [PubMed] [Google Scholar]
- 115.Field JJ, Strunk RC, Knight-Perry JE, et al. Urinary cysteinyl leukotriene E4 significantly increases during pain in children and adults with sickle cell disease. Am J Hematol. 2009. April;84(4):231–3. doi: 10.1002/ajh.21370. [DOI] [PubMed] [Google Scholar]
- 116.Jennings JE, Ramkumar T, Mao J, et al. Elevated urinary leukotriene E4 levels are associated with hospitalization for pain in children with sickle cell disease. Am J Hematol. 2008. August;83(8):640–3. doi: 10.1002/ajh.21199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Haynes J, Jr., Obiako B, King JA, et al. Activated neutrophil-mediated sickle red blood cell adhesion to lung vascular endothelium: role of phosphatidylserine-exposed sickle red blood cells. Am J Physiol Heart Circ Physiol. 2006. October;291(4):H1679–85. doi: 10.1152/ajpheart.00256.2006. [DOI] [PubMed] [Google Scholar]
- 118.Kuvibidila S, Baliga BS, Gardner R, et al. Differential effects of hydroxyurea and zileuton on interleukin-13 secretion by activated murine spleen cells: implication on the expression of vascular cell adhesion molecule-1 and vasoocclusion in sickle cell anemia. Cytokine. 2005. June 7;30(5):213–8. doi: 10.1016/j.cyto.2005.01.010. [DOI] [PubMed] [Google Scholar]
- 119.Haynes J, Jr., Baliga BS, Obiako B, et al. Zileuton induces hemoglobin F synthesis in erythroid progenitors: role of the L-arginine-nitric oxide signaling pathway. Blood. 2004. May 15;103(10):3945–50. doi: 10.1182/blood-2003-08-2969. [DOI] [PubMed] [Google Scholar]
- 120.Aslan M, Celmeli G, Ozcan F, et al. LC-MS/MS analysis of plasma polyunsaturated fatty acids in patients with homozygous sickle cell disease. Clin Exp Med. 2015. August;15(3):397–403. doi: 10.1007/s10238-014-0293-6. [DOI] [PubMed] [Google Scholar]
- 121.Daak AA, Ghebremeskel K, Elbashir MI, et al. Hydroxyurea therapy mobilises arachidonic Acid from inner cell membrane aminophospholipids in patients with homozygous sickle cell disease. J Lipids. 2011;2011:718014. doi: 10.1155/2011/718014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Daak AA, Elderdery AY, Elbashir LM, et al. Omega 3 (n-3) fatty acids down-regulate nuclear factor-kappa B (NF-kappaB) gene and blood cell adhesion molecule expression in patients with homozygous sickle cell disease. Blood Cells Mol Dis. 2015. June;55(1):48–55. doi: 10.1016/j.bcmd.2015.03.014. [DOI] [PubMed] [Google Scholar]
- 123.Daak AA, Ghebremeskel K, Mariniello K, et al. Docosahexaenoic and eicosapentaenoic acid supplementation does not exacerbate oxidative stress or intravascular haemolysis in homozygous sickle cell patients. Prostaglandins Leukot Essent Fatty Acids. 2013. October;89(5):305–11. doi: 10.1016/j.plefa.2013.09.006. [DOI] [PubMed] [Google Scholar]
- 124.Okpala I, Ibegbulam O, Duru A, et al. Pilot study of omega-3 fatty acid supplements in sickle cell disease. APMIS. 2011. July;119(7):442–8. doi: 10.1111/j.1600-0463.2011.02751.x. [DOI] [PubMed] [Google Scholar]
- 125.Tomer A, Kasey S, Connor WE, et al. Reduction of pain episodes and prothrombotic activity in sickle cell disease by dietary n-3 fatty acids. Thromb Haemost. 2001. June;85(6):966–74. [PubMed] [Google Scholar]
- 126.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. January;97(1):37–44. doi: 10.3945/ajcn.112.036319. [DOI] [PubMed] [Google Scholar]
- 127.Key NS, Slungaard A, Dandelet L, et al. Whole blood tissue factor procoagulant activity is elevated in patients with sickle cell disease. Blood. 1998. June 1;91(11):4216–23. eng. [PubMed] [Google Scholar]
- 128.Freedman ML, Karpatkin S. Elevated platelet count and megathrombocyte number in sickle cell anemia. Blood. 1975. October;46(4):579–82. eng. [PubMed] [Google Scholar]
- 129.Alkjaersig N, Fletcher A, Joist H, et al. Hemostatic alterations accompanying sickle cell pain crises. The Journal of laboratory and clinical medicine. 1976. September;88(3):440–9. eng. [PubMed] [Google Scholar]
- 130.Ca Hillery , Foster TD Holzhauer SL, et al. Tissue Factor Deficiency Decreases Sickle Cell-Induced Vascular Stasis in a Hematopoietic Stem Cell Transplant Model of Murine Sickle Cell Disease. Blood. 2004;104(11):236–236. [Google Scholar]
- 131.Gavins FN, Russell J, Senchenkova EL, et al. Mechanisms of enhanced thrombus formation in cerebral microvessels of mice expressing hemoglobin-S. Blood. 2011. April 14;117(15):4125–33. doi: 10.1182/blood-2010-08-301366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.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. doi: 10.1182/blood-2012-04-424143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.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. July 19;120(3):636–46. doi: 10.1182/blood-2012-04-424143. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Arumugam PI, Mullins ES, Shanmukhappa SK, et al. Genetic diminution of circulating prothrombin ameliorates multiorgan pathologies in sickle cell disease mice. Blood. 2015. October 8;126(15):1844–55. doi: 10.1182/blood-2015-01-625707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Chaplin H, Jr., Alkjaersig N, Fletcher AP, et al. Aspirin-dipyridamole prophylaxis of sickle cell disease pain crises. Thromb Haemost. 1980. July 15;43(3):218–21. [PubMed] [Google Scholar]
- 136.Salvaggio JE, Arnold CA, Banov CH. Long-term anti-coagulation in sickle-cell disease. A clinical study. N Engl J Med. 1963. July 25;269:182–6. doi: 10.1056/NEJM196307252690403. [DOI] [PubMed] [Google Scholar]
- 137.Schnog JB, Kater AP, Mac Gillavry MR, et al. Low adjusted-dose acenocoumarol therapy in sickle cell disease: a pilot study. Am J Hematol. 2001. November;68(3):179–83. [DOI] [PubMed] [Google Scholar]
- 138.Ataga KI, Key NS. Hypercoagulability in sickle cell disease: new approaches to an old problem. Hematology Am Soc Hematol Educ Program. 2007:91–6. doi: 10.1182/asheducation-2007.1.91. [DOI] [PubMed] [Google Scholar]
- 139.Pakbaz Z, Wun T. Role of the hemostatic system on sickle cell disease pathophysiology and potential therapeutics. Hematol Oncol Clin North Am. 2014. April;28(2):355–74. doi: 10.1016/j.hoc.2013.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Greenberg J, Ohene-Frempong K, Halus J, et al. Trial of low doses of aspirin as prophylaxis in sickle cell disease. J Pediatr. 1983. May;102(5):781–4. [DOI] [PubMed] [Google Scholar]
- 141.Osamo NO, Photiades DP, Famodu AA. Therapeutic effect of aspirin in sickle cell anaemia. Acta Haematol. 1981;66(2):102–7. [DOI] [PubMed] [Google Scholar]
- 142.Zago MA, Costa FF, Ismael SJ, et al. Treatment of sickle cell diseases with aspirin. Acta Haematol. 1984;72(1):61–4. [DOI] [PubMed] [Google Scholar]
- 143.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. September;132(3):341–5. doi: 10.1016/j.thromres.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Cabannes R, Lonsdorfer J, Castaigne JP, et al. Clinical and biological doubleblind-study of ticlopidine in preventive treatment of sickle-cell disease crises. Agents Actions Suppl. 1984;15:199–212. [PubMed] [Google Scholar]
- 145.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. March 2;305(9):893–902. doi: 10.1001/jama.2011.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.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. September;98(9):1375–82. doi: 10.3324/haematol.2013.086637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.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. July 28;118(4):855–64. doi: 10.1182/blood-2010-09-306167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Boettger MK, Uceyler N, Zelenka M, et al. Differences in inflammatory pain in nNOS-, iNOS- and eNOS-deficient mice. Eur J Pain. 2007. October;11(7):810–8. doi: 10.1016/j.ejpain.2006.12.008. [DOI] [PubMed] [Google Scholar]
- 149.Schmidtko A, Tegeder I, Geisslinger G. No NO, no pain? The role of nitric oxide and cGMP in spinal pain processing. Trends Neurosci. 2009. June;32(6):339–46. doi: 10.1016/j.tins.2009.01.010. [DOI] [PubMed] [Google Scholar]
- 150.Smith WR, Penberthy LT, Bovbjerg VE, et al. Daily assessment of pain in adults with sickle cell disease. Ann Intern Med. 2008. January 15;148(2):94–101. [DOI] [PubMed] [Google Scholar]
- 151.Adams RJ, Brambilla D, Optimizing Primary Stroke Prevention in Sickle Cell Anemia Trial I. Discontinuing prophylactic transfusions used to prevent stroke in sickle cell disease. N Engl J Med. 2005. December 29;353(26):2769–78. doi: 10.1056/NEJMoa050460. [DOI] [PubMed] [Google Scholar]
- 152.Adams RJ, McKie VC, Hsu L, et al. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography. N Engl J Med. 1998. July 2;339(1):5–11. doi: 10.1056/NEJM199807023390102. [DOI] [PubMed] [Google Scholar]
- 153.Ware RE, Helms RW, Investigators SW. Stroke With Transfusions Changing to Hydroxyurea (SWiTCH). Blood. 2012. April 26;119(17):3925–32. doi: 10.1182/blood-2011-11-392340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.DeMuro C, Clark M, Doward L, et al. Assessment of PRO label claims granted by the FDA as compared to the EMA (2006–2010). Value Health. 2013. December;16(8):1150–5. doi: 10.1016/j.jval.2013.08.2293. [DOI] [PubMed] [Google Scholar]
- 155.Panepinto JA, Bonner M. Health-related quality of life in sickle cell disease: past, present, and future. Pediatr Blood Cancer. 2012. August;59(2):377–85. doi: 10.1002/pbc.24176. [DOI] [PubMed] [Google Scholar]
- 156.Panepinto JA. Health-related quality of life in patients with hemoglobinopathies. Hematology Am Soc Hematol Educ Program. 2012;2012:284–9. doi: 10.1182/asheducation-2012.1.284. [DOI] [PubMed] [Google Scholar]
- 157.Ballas SK, Barton FB, Waclawiw MA, et al. Hydroxyurea and sickle cell anemia: effect on quality of life. Health Qual Life Outcomes. 2006. August 31;4:59. doi: 10.1186/1477-7525-4-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Ferrantino M, White RJ. Inhaled treprostinil sodium for the treatment of pulmonary arterial hypertension. Expert Opin Pharmacother. 2011. November;12(16):2583–93. doi: 10.1517/14656566.2011.622269. [DOI] [PubMed] [Google Scholar]
- 159.Hrometz SL, Shields KM. Role of ambrisentan in the management of pulmonary hypertension. Ann Pharmacother. 2008. November;42(11):1653–9. doi: 10.1345/aph.1L014. [DOI] [PubMed] [Google Scholar]
- 160.Anthi A, Machado RF, Jison ML, et al. Hemodynamic and functional assessment of patients with sickle cell disease and pulmonary hypertension. Am J Respir Crit Care Med. 2007. June 15;175(12):1272–9. doi: 10.1164/rccm.200610-1498OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Machado RF, Martyr S, Kato GJ, et al. Sildenafil therapy in patients with sickle cell disease and pulmonary hypertension. Br J Haematol. 2005. August;130(3):445–53. doi: 10.1111/j.1365-2141.2005.05625.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Parent F, Bachir D, Inamo J, et al. A hemodynamic study of pulmonary hypertension in sickle cell disease. N Engl J Med. 2011. July 7;365(1):44–53. doi: 10.1056/NEJMoa1005565. [DOI] [PubMed] [Google Scholar]
- 163.Biomarkers Definitions Working G. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001. March;69(3):89–95. doi: 10.1067/mcp.2001.113989. [DOI] [PubMed] [Google Scholar]
- 164.Rees DC, Gibson JS. Biomarkers in sickle cell disease. Br J Haematol. 2012. February;156(4):433–45. doi: 10.1111/j.1365-2141.2011.08961.x. [DOI] [PubMed] [Google Scholar]
- 165.Adams R, McKie V, Nichols F, et al. The use of transcranial ultrasonography to predict stroke in sickle cell disease. N Engl J Med. 1992. February 27;326(9):605–10. doi: 10.1056/NEJM199202273260905. [DOI] [PubMed] [Google Scholar]
- 166.Ataga KI, Moore CG, Jones S, et al. Pulmonary hypertension in patients with sickle cell disease: a longitudinal study. Br J Haematol. 2006. July;134(1):109–15. doi: 10.1111/j.1365-2141.2006.06110.x. [DOI] [PubMed] [Google Scholar]
- 167.De Castro LM, Jonassaint JC, Graham FL, et al. Pulmonary hypertension associated with sickle cell disease: clinical and laboratory endpoints and disease outcomes. Am J Hematol. 2008. January;83(1):19–25. doi: 10.1002/ajh.21058. [DOI] [PubMed] [Google Scholar]
- 168.Gladwin MT, Sachdev V, Jison ML, et al. Pulmonary hypertension as a risk factor for death in patients with sickle cell disease. N Engl J Med. 2004. February 26;350(9):886–95. doi: 10.1056/NEJMoa035477. [DOI] [PubMed] [Google Scholar]
- 169.Nouraie M, Lee JS, Zhang Y, et al. The relationship between the severity of hemolysis, clinical manifestations and risk of death in 415 patients with sickle cell anemia in the US and Europe. Haematologica. 2013. March;98(3):464–72. doi: 10.3324/haematol.2012.068965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.McGann PT, Hernandez AG, Ware RE. Sickle cell anemia in sub-Saharan Africa: advancing the clinical paradigm through partnerships and research. Blood. 2017. January 12;129(2):155–161. doi: 10.1182/blood-2016-09-702324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Makani J, Cox SE, Soka D, et al. Mortality in sickle cell anemia in Africa: a prospective cohort study in Tanzania. PLoS One. 2011. February 16;6(2):e14699. doi: 10.1371/journal.pone.0014699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.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. doi: 10.1182/blood-2017-06-788935. [DOI] [PubMed] [Google Scholar]
- 173.Ataga K and Stocker K. The trials and hopes for drug development in sickle cell disease. Br. J Haematol. 2015. June 30. doi.org/ 10.1111/bjh.13548 [DOI] [PubMed] [Google Scholar]