Skip to main content

Some NLM-NCBI services and products are experiencing heavy traffic, which may affect performance and availability. We apologize for the inconvenience and appreciate your patience. For assistance, please contact our Help Desk at info@ncbi.nlm.nih.gov.

NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jan 3.
Published in final edited form as: Am J Hematol. 2015 Oct;90(10):934–950. doi: 10.1002/ajh.24116

2015 Clinical trials update in sickle cell anemia

Natasha Archer 1, Frédéric Galacteros 2, Carlo Brugnara 3,*
PMCID: PMC5752136  NIHMSID: NIHMS930159  PMID: 26178236

Abstract

Polymerization of HbS and cell sickling are the prime pathophysiological events in sickle cell disease (SCD). Over the last 30 years, a substantial understanding at the molecular level has been acquired on how a single amino acid change in the structure of the beta chain of hemoglobin leads to the explosive growth of the HbS polymer and the associated changes in red cell morphology. O2 tension and intracellular HbS concentration are the primary molecular drivers of this process, and are obvious targets for developing new therapies. However, polymerization and sickling are driving a complex network of associated cellular changes inside and outside of the erythrocyte, which become essential components of the inflammatory vasculopathy and result in a large range of potential acute and chronic organ damages. In these areas, a multitude of new targets for therapeutic developments have emerged, with several ongoing or planned new therapeutic interventions. This review outlines the key points of SCD pathophysiology as they relate to the development of new therapies, both at the pre-clinical and clinical levels.

Clinical Presentation and Pathophysiology of Sickle Cell Disease

The clinical phenotype of patients with sickle cell disease (SCD) can be exceptionally diverse, despite a finite number of mutations. Clinical manifestations range from almost no symptoms to multiple, potentially fatal, events. Ballas et al. divided the complications secondary to SCD according to three main categories including hematological, pain, and complications affecting major organs, in order to more effectively standardize their definition [1]. While management guidelines are available [2,3], not every SCD patient can be treated exactly the same way given each patients individual manifestations of disease and variable response to therapies.

The most common acute manifestations of the disease include vaso-occlusive crisis (VOC), acute chest syndrome, stroke, priapism, sudden deafness, and acute anemia, particularly from aplastic crisis and splenic sequestration. Individuals with SCD are also more susceptible to stroke and serious bacterial infections. The spectrum of clinical manifestations is age dependent; women with SCD are particularly at risk during pregnancy. In addition to hemolytic anemia, common chronic complications affect major organs, such as brain, kidney, heart, lung, skin, retina, vestibular-cochlear systems, and bone. Some of these complications are seen predominantly in adults. Iatrogenic complications should also be considered, such as delayed hemolytic transfusion reactions and impotence due to inadequate treatment of priapism. Over their lifetime, patients differentially accumulate a wide spectrum of functional defects, which are either passive sequelae of the disease or further reinforce disease pathophysiology and worsen its clinical manifestations, such as renal tubular dysfunctions, which facilitate the development of acidosis.

Acute manifestations of the disease are managed by treating the associated symptoms. In the acute setting, pain crises are treated with hydration, warm packs, and analgesics ranging from nonsteroidal anti-inflammatory drugs (NSAIDs) to opioids. Acute chest syndrome, which typically manifests with respiratory symptoms ranging from an increased respiratory rate to desaturations, is treated with efforts to increase oxygen carrying capacity, i.e. supplemental oxygen, incentive spirometry, and exchange blood transfusion. If pain is associated with acute chest syndrome, analgesics are provided to remove the associated reduction in ventilation. In splenic sequestration, severe, acute anemia is a life-threatening symptom, and is thus treated with top-up blood transfusion. Stroke and serious bacterial infections are treated as they would be in non-SCD but the hemorrhagic risks associated with therapy should be carefully evaluated and glucocorticoids should be used with caution. In addition, SCD patients with stroke receive either simple or exchange transfusions depending on their hemoglobin level and capacity to rapidly achieve a reduction to less than 30% for the remaining SS RBC measured by % HbS after transfusion.

Chronic manifestations sometimes need to be managed symptomatically, but are preferably managed prophylactically in an effort to decrease morbidity associated with each condition. This often requires nonspecific treatment, but treatments that aim at decreasing the pathophysiology associated with SCD, i.e. hemolysis and vaso-occlusion, are particularly needed. The complex clinical nature and evolving pathophysiology of SCD emphasizes the need for very well coordinated follow-up and transition from childhood to adult care.

Preventative measures are currently the hallmark of the management of both acute and chronic manifestations of SCD. Even before disease confirmation, after an initiation newborn screen returns positive for SCD, infants are started on penicillin prophylaxis. The meningococcal conjugate vaccine (MCV4) and pneumococcal polysaccharide vaccine (PPV23), in addition to routine childhood immunizations, are given to all children with SCD for added protection against encapsulated bacteria. These interventions are meant to prevent illness and/or death secondary to severe bacterial infections in the setting of functional asplenia. Another example of prophylaxis is the use of chronic transfusion, and now possibly hydroxyurea, for stroke prevention. Up until recently, children with SCD and elevated Transcranial Doppler (TCD) blood velocities were encouraged to begin a chronic transfusion regimen in efforts to reduce the % HbS containing RBC thus lessening vascular complications. In patients with known stroke or silent cerebral infarcts, chronic transfusions have been shown to result not only in a significant risk reduction of infarct recurrence, but also improved quality of life [4]. However, chronic, monthly blood transfusions are not without potential sequelae, most notably iron overload. The TWiTCH study set out to determine if daily hydroxyurea could lower TCD velocity in children with SCD in a similar way as blood transfusion. This trial was terminated early in November 2014 because hydroxyurea was determined to be noninferior to transfusion therapy in reducing TCD blood velocity. Publication of these data will be an important contribution to the ongoing discussions concerning whether or not hydroxyurea can provide an alternative to chronic blood transfusion for children at risk of developing stroke [5].

The only curative option currently available for SCD is hematopoietic stem cell transplant (HSCT). This approach is mature enough to have resulted in practice guidelines regarding its indications and management [6,7]. However, the protocols currently in use are numerous and the numbers of treated patients in each of the protocols are limited. When one considers the substantial reduction in mortality achieved with noncellular therapies over the last 20 years, long-term mortality for older HSC-based therapies is similar to that of optimal medical care. In addition, long-term complications of HSC therapies are not well documented; the increased long-term risk of solid tumors in transplanted SS patients is an area of particular concern.

Major Therapeutic Strategies for Sickle Cell Disease

HbS polymerization and erythrocyte sickling

Seminal work by the NIH group of Eaton and coworkers has elucidated the interplay of O2 tension and HbS concentration in the kinetics of HbS polymerization [811] (Tables IIV). The extreme dependence of the delay time for HbS polymerization on HbS concentration is a fundamental characteristic underlying SCD pathophysiology. It explains the very low clinical expression in sickle cell trait, the presence of disease in HbSC compound heterozygosity, the beneficial effects of increasing concentrations of HbF, and the modifications in disease severity when the cellular concentration of HbS is decreased by concomitant alpha or beta thalassemia or iron deficiency [18]. Four major avenues of potential therapies directly or indirectly target HbS polymerization, with the final objective of decreasing polymerization and sickling.

TABLE I.

Planned, ongoing or recently completed Phase 1 trials

Company Drug Mechanism Indication Route Clintrials.gov Status
HB F-Anti-sickling
The Cleveland Clinic Decitabine (Dacogen) and tetrahydrouridine HbF induction Prophylaxis
HbSS, HbSβ0, HbSβ+, HbSC
≥18 years
Oral NCT01685515 Open
ASH 2014 # 90
Celgene Pomalidomide (Pomalyst) HbF induction Prophylaxis
HbSS, HbSβ0
18 years > 60 years
Oral NCT01522547 Completed
Novartis Pharmaceuticals Panobinostat (Farydak) HbF induction (HDAC inhibition) Prophylaxis
HbSS, HbSβ0
≥18 years
Oral NCT01245179 Open
AesRx (Baxter International) 5-hydromethyl-2-furfural (AES-103) Increased O2 affinity Prophylaxis
HbSS
18 years ≥ 65 years
Oral NCT01597401 Completed
Prolong Pharmaceuticals PEGylated carboxyhemoglobin bovine (Sanguinate) O2 and CO delivery (oxygenation and anti-inflammation) Prophylaxis
Healthy
18 years ≥ 45 years
IV NCT01847222 Completed;
ASH 2014 #1372
Prolong Pharmaceuticals PEGylated carboxyhemoglobin bovine (Sanguinate) O2 and CO delivery (oxygenation and anti-inflammation) Prophylaxis
HbSS, HbSβ0, HbSβ+, HbSC
18 years ≥ 65 years
IV NCT01374165 Recruitment suspended
Prolong Pharmaceuticals PEGylated carboxyhemoglobin bovine (Sanguinate) O2 and CO delivery (oxygenation and anti-inflammation) Prophylaxis
HbSS
≥18 years
IV NCT01848925 Completed
Invenux, LLC SCD-101 Inhibition of sickling Prophylaxis
HbSS, HbSβ0
18 years ≥ 55 years
Oral NCT02380079 Open
Global Blood Therapeutics GBT440 (formerly GTx011) Increased O2 affinity Prophylaxis
Healthy, 18 years ≥ 55 years
HbSS, 18 years ≥ 60 years
Oral NCT02285088 Open;
ASH 2014 # 217 (mouse) and #1370 (in vitro)
Anti-inflammatory
NKT Therapeutics NKTT120 Anti-inflammation Prophylaxis
HbSS, HbSβ0
18 years ≥ 60 years
IV NCT01783691 Completed;
ASH 2014 #2718
Children’s Hospital Medical Center Zileuton (ZYFLO) Anti-inflammation Prophylaxis
HbSS, HbSβ0, HbSβ+, HbSC
≥12 years
Oral NCT01136941 Completed; [12]
NHLBI Regadenoson (Lexiscan) Anti-inflammation VOC diagnosis
HbSS, HbSβ0
18 years ≥ 70 years
IV NCT01566890 Completed; [13]
Pfizer PF-04447943 Phosphodiesterase 9 inhibitor Prophylaxis
HbSS, HbSβ0
18 years ≥ 70 years
Oral NCT02114203 Open
Vanderbilt University Budenoside Anti-inflammation Post-ACS prophylaxis
HbSS, HbSβ0
1 years > 4 years
Inhaled NCT02187445 Open
Anti-adhesion
University of Miami and Duke University Propranolol Anti-adhesion Prophylaxis
HbSS, HbSβ0
7 years ≥ 17 years
Oral NCT02012777 Open
University of Miami and Duke University Propranolol Anti-adhesion Prophylaxis
HbSS, HbSβ0
≥18 years
Oral NCT01077921 Completed
Albert Einstein College of Medicine Immune Globulin (Gamunex-C) Anti-adhesion Acute VOC treatment
HbSS, HbSβ0, HbSβ+
12 years ≥ 65 years
IV NCT01757418 Completed; [106]
University Hospitals Case Medical Center Sodium bicarbonate Alkali therapy Low serum bicarbonate levels treatment
HbSS
≥18 years
Oral NCT01894594 Open
Other
Sangart MP4CO (pegylated carboxyhemoglobin, > 90% CO-Hb saturation) Anti-sickling and vascular stasis Prophylaxis
HbSS, HbSβ0
≥18 years
IV NCT01356485 Completed
Phase Ib
Discontinued
NHLBI Topical sodium nitrite cream Increased blood flow Leg ulcers treatment
HbSS, HbSβ0, HbSβ+, HbSC
≥18 years
Topical NCT01316796 Completed; [14]

TABLE IV.

Compounds in preclinical development

Company Drug Mechanism Indication/Route Other Status
Acetylon Pharmaceuticals HDAC ½ inhibition HbF induction ASH 2014 #335
EpimedX EdX-17 HbF induction (ferritin heavy chain activation) ASH 2014 #1357
Errant Gene Therapeutics Trichosic HbF induction Early stage of development
Phoenicia Biosciences & NCATSTRND PB-04 HbF induction Prophylaxis ASH 2014 #2687 for other targets
Isis Pharmaceuticals Antisense nucleotides HbF induction via KLF1 reduction ASH 2014 # 4038
NIH (academic, U Chicago RN-1 Inhibition of LSD1, a repressor of gamma-globin expression SC ASH 2014 #336 Baboon-study shows HbF induction
Global Blood Therapeutics GTx011 Increase O2 affinity ORAL ASH 2014 #1370 Sickle mouse study ASH 2014 # 217
Virginia Common- wealth And CHOP INN-270 and TD-7 Increase O2 affinity Sickle mouse study open
Erytech Pharma Enhoxy O2 delivery EU Orphan drug designation 2012
Biogen Idec Tysabri (Natalizumab) Anti-adhesion IV ASH 2014 #221 Approved for relapsing MS
Archemix ARC5690 Aptamer anti-adhesion IV In liquidation
Biogen Idec TYSABRI, Natalizumab VLA4 blocker, anti-adhesion Treatment IV Approved for relapsing MS & Crohn’s disease
Acceleron Pharma Luspatercept (ACE-536) Ligand trap TGFbeta superfamily SC ASH 2014 #4055 Phase 2 in MDS and Beta-thalassemia intermedia
ReveraGen Biopharma VBP15 Anti-inflammation (like prednisolone, NF-kB inhibition, fewer side effects) ORAL Studies in Duchenne muscular dystrophy
Biogen Idec Tecfidera ® (Dimethyl fumarate) Anti-inflammation Prophylaxis
ORAL
Approved for relapsing MS
Addex Therapeutics TFNR1 NAM A2a PAM A2AR, Anti-inflammation ORAL Studies in Parkinson’s disease
Advinus Therapeutics PNQ103 AQ2BAR antagonist ORAL Ex-vivo studies on SCD RBCs
Pfizer PF-04447943 Phosphodiesterase 9 inhibitor ORAL ASH 2014 #2694 Unsuccessful in Alzheimer’s trial. Reduced vaso-occlusion in SS mouse
Duke University TBD MEK1/2 and ERK1/2 inhibition [170]
Alnylam Pharmaceuticals ALN-TMP RNAi Tmprss6, to reduce Fe load Iron overload SC Studies in Beta-thalassemia

Increasing intracellular concentration of HbF

An increasing fraction of HbF inside the sickle erythrocyte is profoundly relevant not only for the concomitant decrease in HbS, but also for the unique capability of HbF to inhibit HbS polymerization. Large studies like the Cooperative Study of Sickle Cell Disease have shown that HbF is among the most important disease modifiers [19,20]. Similarly, studies on a variety of patient cohorts outside of the U.S. have shown lower disease severity in the presence of higher values of HbF. In a study involving Saudi and African American sickle cell patients, despite similar hemoglobin beta (HBB) gene cluster haplotypes, different clinical phenotypes in the Saudi cohort were attributed to their higher values of HbF [21]. Effects of several HbF modifiers have been described on both HbF values and other hematological parameters [2125].

Studies by Platt et al., Charache et al., and others have led to the approval of hydroxyurea as a therapeutic agent in adult patients [26,27]. Early studies of 5-azacytidine and hydroxyurea treatment in SCD patients were notable for an increase in F reticulocytes and HbF elevation. This often led to an increase in hemoglobin despite mild bone marrow suppression noted in other cell lines. More than a decade later, clinical benefits were demonstrated including a reduction in pain crises and an associated reduction in mortality with increasing number of cumulative years on hydroxyurea therapy [27,28].

While hydroxyurea has not yet been approved by the FDA for use in children with SCD, several studies and its more aggressive inclusion in the most recent SCD management recommendations speak to its safety and effectiveness in a pediatric population [2,29]. In the phase 1/2 HUG-KIDS trial, 52 children were treated at Maximum Tolerated Dose (MTD) for 1 year, with no significant clinical adverse effects; hematologic toxicities were found to be mild and reversible [30]. In the Baby Hug phase 3 clinical trial, SCD infants 9 to 18 months of age treated with hydroxyurea and compared with a placebo, had reduced pain scores and fewer pain crises and acute chest syndrome [31,32]. Importantly, hydroxyurea was not associated with an increased risk of serious bacterial infections. Subsequent studies have also shown no short-term effect of hydroxyurea on growth and some positive effects on renal function [3335]. With this supportive evidence, the most recent SCD management guidelines recommend the use of hydroxyurea in both adults with three or more VOCs in any 12-month period and, regardless of symptoms, in children greater than 9 months of age diagnosed with sickle cell anemia [2,29]. Despite the overwhelming evidence supporting the clinical benefits of hydroxyurea, only one out of four adult patients and possibly even fewer are treated with this drug [36]. Extending hydroxyurea therapy to all eligible patients should be a major target for all therapeutic interventions for SCD. Hydroxurea is also being tested in a Phase 2 study in adult and pediatric patients with Hb SC disease (SCYTHE, NCT02336373). Change in quality of life after 6 months at MTD is the primary endpoint of this study. In adults, phlebotomy is allowed if quality of life does not improve after 6 months at MTD.

The overall success of hydroxyurea has led to efforts exploring its potential synergy with other drugs such as magnesium [37]. A study investigating adjuvant magnesium pidolate in patients with HbSC disease treated with hydroxyurea was unfortunately closed early due to slow enrollment; no difference in the primary outcome of hyperdense cells or the secondary outcome of clinical events were observed across the four treatments arms (HU + Mg, HU + placebo, Mg + placebo and placebo + placebo) [38]. In vitro studies have provided evidence that hydroxyurea reduces the endothelial expression of adhesive ligands like Vascular Cell Adhesion Molecule-1 (VCAM-1) and the activation of Lutheran/basal cell-adhesion molecule (Lu/BCAM) [39,40]. Montelukast is a leukotriene receptor antagonist currently approved for prophylaxis and chronic treatment of asthma. In vitro data suggest that it inhibits eosinophil adhesion to VCAM-1 [41]. An ongoing trial is investigating whether the addition of montelukast versus placebo to hydroxyurea leads to a measurable reduction in soluble VCAM-1 (Vanderbilt University, NCT01960413).

NHLBI is currently conducting a trial in 24 patients, age 15 and older, to test a new dosing algorithm to maximize response and reduce side effects of hydroxyurea (NCT02225132, Primary outcome: Maximal HbF values). Baylor College of Medicine is testing a new approach to shorten the time required to achieve MTD for hydroxyurea (NCT02042222, up to 105 patients, age 1–16 years.). It is however not clear how adherence to treatment can be eventually improved by the optimization of MTD for hydroxyurea. While HbF is a widely used endpoint to assess both MTD and therapeutic effectiveness, other endpoints such as the % of dense red blood cells are relevant for disease pathophysiology, have shown value and could be used to guide treatment [42,43].

Other compounds known to increase HbF are currently being investigated clinically. Decitabine (5-aza-2′-deoxycytidine), a drug commonly used to treat myelodysplastic syndrome, inhibits DNA methyltransferase. It is currently 4 years into a phase 2 clinical trial, testing the safety and effectiveness of weekly to biweekly injections administered over 1 year in SCD patients older than 18 years in whom hydroxyurea was not deemed effective or tolerable (NCT01375608, Table II). Oral Decitabine plus Tetrahydrouridine, a cytidine deaminase inhibitor, is also being investigated in a similar patient population and is currently in year 2 of its phase 1 clinical trial. MTD, safety, and effect on HbF induction are being determined for Pomalidomide, a derivative of thalidomide that also inhibits angiogenesis, in SCD patients greater than 18 years of age and who are also hydroxyurea refractory or intolerant (Table I). A phase 2 clinical trial for 2,2-dimethylbutyrate’s (HQK-1001), an oral HbF inducer, was recently terminated early for lack of effects [15,44]. Inhibition of histone deacetylase (HDAC) is being investigated as a mechanism to increase HbF; Vorinostat’s phase 2 clinical trial was recently closed due to the lack of measurable effects, while panobinostat (LBH589) is still in phase 1.

TABLE II.

Planned, ongoing or recently completed Phase 2 trials

Funding Drug/Company Mechanism Indication Route Clintrials.gov Status
HB F-Anti-sickling
NHLBI Decitabine HbF induction Prophylaxis
HbSS, HbSβ0, HbSC
≥18 years
IV NCT01375608 Open
Hema-Quest Pharmaceuticals 2,2-dimethylbutyrate (HQK-1001) HbF induction Prophylaxis
HbSS, HbSβ0
12 years ≥ 60 years
Oral NCT01601340 Completed; [15]
Baxter International Inc. 5-hydromethyl-2-furfural (AES-103) Increased O2 affinity Prophylaxis
HbSS
18 years ≥ 60 years
Oral NCT01987908 Open
Dana Farber Cancer institute Vorinostat (Zolinza) HbF induction Prophylaxis
HbSS, HbSβ0
≥18 years
Oral NCT01000155 Discontinued
Prolong Pharmaceuticals PEGylated carboxyhemoglobin bovine (Sanguinate) O2 and CO delivery (oxygenation and anti-inflammation) Acute VOC treatment
HbSS, HbSβ0, HbSβ+, HbSC
≥12 years
IV NCT02411708 Not yet open
Baylor College of Medicine Hydroxyurea HbF induction; Rheological improvement Prophylaxis
HbSC
5 years ≥ 21 years
Oral NCT02336373 Open (SCYTHE)
Baylor College of Medicine Hydroxyurea ± Phlebotomy HbF induction; Rheological improvement Prophylaxis
HbSC
18 years ≥ 69 years
Oral N/A Not yet open
Anti-adhesion, hemolysis
Novartis (Selexys) SelG1 Anti-P-selectin Prophylaxis
HbSS, HbSβ0, HbSβ+, HbSC
16 years ≥ 65 years
IV NCT01895361 Open (SUSTAIN)
Vanderbilt University Montelukast (Singulair) Anti-adhesion Prophylaxis
HbSS, HbSβ0
16 years ≥ 60 years
Oral NCT01960413 Open
St. Louis University Abciximab (ReoPro) Anti-adhesion Acute VOC treatment
HbSS, HbSβ0, HbSC
5 years ≥ 25 years
IV NCT01932554 Discontinued
Astra Zeneca Ticagrelor (Brilinta) PLT aggregation inhibition Prophylaxis
HbSS, HbSβ0
2 years ≥ 18 years
Oral NCT02214121 Open
University of North Carolina Rivaroxaban (Xarelto) Anticoagulant Prophylaxis
HbSS, HbSβ0
18 years ≥ 55 years
Oral NCT02072668 Open by invitation only
Dilaforette AB (Sweden) Ergomed plc (UH) Sevuparin Anticoagulant Acute VOC treatment IV N/A Study design phase
University of Pittsburgh Unfractionated heparin Anticoagulant Acute Chest Syndrome treatment
HbSS, HbSβ0, HbSC
18 years ≥ 20 years
IV NCT02098993 Open
Boston Children’s Hospital Inhaled NO NO repletion Acute VOC treatment
HbSS, HbSβ0, HbSC
9 years ≥ 22 years
Inhaled NCT00142051 Discontinued
Wake Forest School of Medicine Beet Juice (Unbeetable) NO repletion Prophylaxis
HbSS, HbSβ0
19 years ≥ 65 years
Oral NCT02162225 Open
Bio Products Laboratory (UK) BPL Haptoglobin Free Hb depletion Prophylaxis IV N/A EU Orphan drug designation 2011
University of North Carolina Eptifibatide (Integrilin) PLT aggregation inhibition Acute VOC treatment
HbSS, HbSβ0, HbSC
18 years ≥ 55 years
IV NCT00834899 Discontinued; [16]
Anti-inflammatory
NHLBI Regadenoson (Lexiscan) Anti-inflammation Acute VOC treatment
HbSS, HbSβ0
10 years ≥ 70 years
IV NCT01788631 Open
Children’s Hospital & Research Center Simvastatin (Zocor) Anti-inflammation Prophylaxis
HbSS, HbSβ0
≥10 years
Oral NCT01702246 Open
University of North Carolina Atorvastatin (Lipitor) Anti-inflammation Prophylaxis
HbSS, HbSβ0
18 years ≥ 60 years
Oral NCT01732718 Open by invitation only
Fish oil (Lovaza) Anti-inflammation Prophylaxis Oral NCT01202812 Unknown
Thomas Jefferson University, Mount Sinai School of Medicine Inhaled Mometasone Furoate Anti-inflammation HbSS, HbSβ0
10 years >19 years
Acute VOC treatment
HbSS, HbSβ0
≥15 years
Inhaled NCT02061202 Open
NKT Therapeutics NKTT120 Anti-inflammation Prophylaxis IV N/A Study design phase
Boston University Medical Center Macitentan (Opsumit) Endothelin blocker Pulmonary arterial hypertension treatment Oral NCT02126943 Open
Children’s Hospital Medical Center Losartan (Cozaar) Reduction of intra-glomerular pressure Sickle nephropathy prophylaxis and/or treatment
HbSS, HbSβ0
≥6 years
Oral NCT01479439 Open
University of Alabama at Birmingham Losartan (Cozaar) Reduction of intra-glomerular pressure Sickle nephropathy prophylaxis
HbSS, HbSβ0
6 years ≥ 19 years
Oral NCT02373241 Open
Lagos State University Sorghum bicolor (Jobelyn) Anti-oxidant Prophylaxis
HbSS
14 years ≥ 40 years
Oral NCT01704794 Open

Still in development are a handful of compounds that have some promising preclinical data (Table IV). BCL11A is a transcriptional suppressor of HbF production; a substantial component of HbF variability is associated with genetic polymorphisms in BCL11A [45,46]. Acetylon’s ACY-957, also a selective HDAC ½ inhibitor, is thought to downregulate BCL11A, and upregulate GATA 2, which conversely induces HbF (ASH 2014 #335). Pracinostat (SB939), another HDAC inhibitor, and PB-04 also induced HbF production in erythroid precursors in SCD and beta thalassemic patients (ASH 2014, # 2687). Ferritin heavy chain (FtH), a protein that stimulates gamma globin, is activated by EdX-17, augmenting HbF levels in betaYac mice to greater than 25% (ASH 2014, # 1357). Lysine Specific Demethylase-1 (LSD-1) inhibitor, RN-1, which has previously demonstrated a two to threefold increase in F cells while inducing γ-globin mRNA levels five to eightfold in mice (ASH 2014, # 561) has also induced high levels of HbF, F reticulocytes, and F cells in baboons (ASH 2014, #336). Erythroid Kruppel like factor (KLF1), involved in β-globin induction and γ-globin suppression, has also been targeted using antisense oligonucleotides (ASOs) in both murine and human cell lines. Subcutaneous administration of KLF1 ASOs in mice and rats has been shown to reduce β-globin production (ASH 2014, #4038). Valproic acid and trichostatin have been shown to increase γ-globin gene expression in human liquid erythroid cultures [47]. Trichostatin A (Errant Gene Therapeutics) is a histone deacetylase inhibitor, which may induce HbF expression, but no data have been published or presented so far. For many of the preclinical leads described above, it should be noted that ability of inducing HbF production in vitro in cell lines does not necessarily translate into in vivo efficacy in humans. The limited availability of nonhuman primate models also poses a significant challenge to the pre-clinical development of these compounds.

Reducing intracellular concentration of HbS

This avenue comprises either modification of ion transport across the erythrocyte membrane with the intent of preventing sickle cell dehydration or reduction of HbS concentration by iron deficient erythropoiesis. Given the unique dependence of the kinetics of HbS polymerization on HbS concentration, the presence of erythrocyte dehydration in SCD has important implications for pathophysiology and was originally and unsuccessfully targeted in 1980 in a study aimed to induce red cell swelling with hypo-osmolarity [48]. Over the last 30 years, the major pathways which are responsible for erythrocyte dehydration have been characterized in substantial detail [49]. They include the K-Cl cotransport (KCC1,3,4) [5054], the Ca-activated Gardos channel (KCCN4) [5559], and Psickle (most probably mediated by the mechano-sensitive ion channel Piezo 1) [6062]. Pharmacological inhibition of these pathways has been demonstrated in vitro and in vivo in transgenic mouse models. Clinical trials with dietary magnesium supplementation (Mg-pidolate) in patients with SCD have shown increases in red cell magnesium content, inhibition of K-Cl cotransport, and improvements in erythrocyte hydration, but there have been no completed controlled trials testing clinical efficacy of this approach [63,64]. A phase 1 study of oral Mg-pidolate supplementation established the MTD for children with SCD concomitantly treated with hydroxyurea [36]. As discussed above, a study on oral Mg pidolate supplementation in patients with SC disease was terminated early due to poor enrollment, but the limited data collected showed no changes in erythrocyte Mg or cell dehydration [37]. Clinical studies on the inhibition of the Gardos channel were first conducted in the acute settings with IV cetiedil [65] and subsequently with oral clotrimazole and senicapoc as prophylactic agents. These latter studies resulted in reduction of cell dehydration, improvement of anemia, but no measurable improvements in the relevant clinical endpoints, and an actual increase in painful crises in the subgroup of patients not receiving hydroxyurea [6668]. Since Hb increased in all senicapoc-treated patients, it is possible that the associated increase in blood viscosity negated the benefit of reducing cellular HbS concentration, and resulted in increased vaso-occlusion. However, there was no correlation between Hb values or changes in Hb values and pain rates in patients treated with senicapoc. In vitro and in vivo data have shown a role for endothelin-1 (ET-1) receptor blockade in reducing erythrocyte dehydration, most likely due to a functional connection between activation of this receptor and activation of the Gardos channel [69]. It remains to be determined if targeting ET-1 can achieve a greater reduction in cell dehydration than targeting the Gardos channel itself.

Isolated reports of improvement in SCD due to concomitant hypochromia induced by iron deficiency have so far not translated into viable therapeutic strategies and have not considered potentially associated reduction in HbF [70,241160071]].

Direct inhibition of HbS polymerization

In vivo chemical modification of HbS resulting in inhibition of polymerization has been an elusive therapeutic goal. Obvious challenges to this approach are the high concentration of Hb in the erythrocyte requiring a substantial amount of modifying compound to be absorbed by the GI tract and to cross the erythrocyte membrane without affecting other crucial cellular functions. Several compounds have been tested, all with disappointing results so far. Na cyanate mediates an irreversible carbamylation of the aminoterminal valine of HbS which results in reduced polymerization and sickling [72]. In vivo studies with extracorporeal carbamylation showed measureable changes in several hematological parameters [73], however, significant toxicities were observed most likely due to carbamylation of other targets, resulting in peripheral neuropathy, CNS toxicity, weight loss, and cataract formation. BW12C, a substituted benzaldehyde, showed significant in vivo modification of HbS when administered parenterally to patients with SCD [74,75], but it was not further developed due to significant toxicity observed with a related compound.

The food additive vanillin binds to HbS and reduces polymerization and sickling by both an allosteric shifting of oxygen affinity and a stereospecific inhibition of polymer assembly [76]. Reduced polymerization and sickling were reported following a double-blind, placebo-controlled in vivo study using 1 g vanillin/day for 40 days in 30 patients with SCD in Cuba [77]. However, no additional studies were carried out with this compound. Recent evidence suggests that vanillin may adversely affect red cell ion transport and produce K loss and dehydration of sickle cells [78]. INN-270 and TD-7, two derivatives of vanillin, demonstrate high rates of HbS binding and modification resulting in a shift to a higher oxygen affinity hemoglobin state (ASH 2014, #218) [79]. Sickle mouse studies using these two compounds are ongoing.

Indirect inhibition of HbS polymerization by increasing oxygen affinity

Several compounds have been identified that indirectly inhibit HbS polymerization by shifting the partial pressure of oxygen at which 50% of hemoglobin is saturated with oxygen (P50). For some of the compounds described in the preceding section, such as BW12C, it is not completely understood how much of the antisickling effect is due to inhibition of polymerization and how much to a left-shift in P50. A general concern for this approach is that a reduction in tissue oxygen delivery may produce a compensatory increase in hemoglobin, with concomitant increased viscosity and vaso-occlusion [80]. Sudden interruption of this kind of treatment may also expose patients to substantial complications due to the increased overall mass of circulating sickle cells.

Even including some of the drugs discussed in the preceding section, a limited number of clinical trials have been conducted so far. Oral administration of Tucaresol, a substituted benzaldehyde, resulted in measureable changes in oxygen affinity, but also in significant toxicity [81].

The active ingredient for AES-103 (Baxter International Inc.) is 5-hydroxymethyl-2-furfural (5-HMF), which has been shown to form a high-affinity Schiff-base adduct with HbS and indirectly inhibits sickling via a leftward oxygen curve equilibrium shift (ASH 2014 # 2699) [82]. In vitro 5-HMF reduces sickling-induced dehydration (Gardos and Psickle) while it increases K loss and dehydration mediated by the K-Cl cotransport [83]. A phase 1 clinical trial tested oral doses of AES-103 up to 4,000 mg in 18 patients with SCD, some treated with hydroxyurea, with no significant side effects [84]. AES-103 is currently being tested in a phase 2 clinical trial (Table II).

GBT440 (formerly GTx011, Global Blood Therapeutics, South San Francisco, CA), increases HbS oxygen affinity and diminishes cell sickling in vitro, while it prolongs red cell survival in vivo in sickle (Townes) mice (ASH 2014 #217, # 1370). A phase 1/2 clinical trial is ongoing to assess safety, pharmacokinetics and pharmacodynamics of oral GBT440 (single and multiple doses, administered once daily) in healthy subjects and in patients with SCD (Table I).

While reducing oxygen carrying capabilities, CO has multiple potentially beneficial effects in SCD, which include inhibition of polymerization, increase in oxygen affinity, reduced inflammation, and increase antioxidant responses. Delivery of CO via a pegylated hemoglobin saturated with CO has improved vaso-occlusion in a mouse model of SCD [85]. It remains to be determined if this can be achieved in humans without significant side effects.

The inflammatory vasculopathy of sickle cell disease

Adhesion of sickle cells to endothelium and vaso-occlusion

Adhesion of sickle cells to endothelium plays a major role in SCD pathophysiology, especially as it relates to sickle vaso-occlusion. It is generally believed that vaso-occlusion develops from adhesion of sticky sickle RBCs to endothelium, followed by trapping and polymerization of rigid, less deformable cells [86,87].

The mechanisms supporting the adhesion of sticky sickle erythrocytes to endothelium as well as additional, cooperative adhesive interactions involving activated leukocytes, monocytes, and platelets are of great interests, since interfering with these mechanisms has the potential to greatly impact clinical severity. As we learn more about the specific receptors involved in the adhesion process, new therapeutic targets are coming into play to prevent and/or reverse vaso-occlusion.

Selectins (P, E, and L) play a crucial role in adhesion of leukocytes to endothelium [88]. In sickle erythrocytes, P-selection and E-selectin have therefore been explored as potential targets. Considerable debate has been generated regarding the single selective (P or E) versus pan-selectin blockade. There are also concerns about the fact that selectins mediate adhesion mostly in the presence of shear stress [89,90], and it is not clear how much this mechanism is actively engaged when the blood flow is reduced or stopped as presumably happens in VOC. Matsui et al. demonstrated that the mechanism of decreased sickle cell adhesion via unfractionated heparin is through P-selectin inhibition [91]. Unfractionated heparin was shown to bind to P- and L-selectins and inhibit their function, while low molecular weight heparins did not [92]. Promising results in a small cohort of patients with SCD were obtained with Pentosan Polysulfate Sodium (PPS), an agent used for treatment of pain associated with interstitial cystitis (Elmiron, Janssen Pharmaceuticals). PPS was shown to block P-selection adhesive processes in vitro, to normalize microcirculatory blood flow, and to reduce markers of vascular injury in vivo [93]. Availability of an orally-absorbed selectin inhibitor would be a major advantage compared with those requiring SC injections. In 2007, a randomized, double-blind trial in 253 patients with acute VOC showed positive results on duration of crises, hospitalization and pain intensity with the daily SC administration of 175 IU/Kg of Tinzaparin, a LMWH [94]. US marketing of Tinzaparin, (Innohep, LEO Pharma, Ballerup, Denmark) was discontinued in February 2011.

Sevuparin, a chemically modified heparin, currently being studied as a antimalarial agent, has also been shown to reduce sickle red cell and leukocyte adhesion in vitro, to stimulated endothelial cells [95]. Given promising in vivo studies in nude mice demonstrating decreased vaso-occlusion following TNF-α exposure, Dilaforette AB (Sweden) and Ergomed plc (UK) are planning a phase 2 study using sevuparin as an acute therapy for VOCs (Table II). Hemostatic balance is of critical pathophysiological importance in SCD: hemorrhagic (retinal, renal, brain) disease as well as thrombotic events (pulmonary embolism, phlebitis) are not uncommon. Thus contrary to Tinzaparin, LMWH agents like Sevuparin and pentosan that have limited anticoagulation properties may provide a safer alternative.

GMI-1070 (Rivipansel, GlycoMimetics Inc.) is a E-selectin inhibitor which was shown to be safe and to produce some positive biomarker changes in a phase 1 study when injected IV in 15 SCD patients [96]. GMI-1070 was studied in phase 2 study with the primary outcome measure being a reduction in time to resolution of VOC. No statistically significant effects were observed on the primary outcome measure, while plasma E-selectin (ASH # 2704), opioid and overall pain medication usages were significantly reduced [97100]. Despite having failed its primary outcome, GMI-1070 will be tested in a phase 3 trial, on SCD patients 6 years of age and older hospitalized for pain (Table III). The primary outcome of this study is time to readiness to discharge, while secondary outcomes include amount and duration of opioids and readmission rates. The initiation of this phase 3 trial has been delayed due to manufacturing issues.

TABLE III.

Currently planned, ongoing, or recently completed Phase 3 trials

Company Drug Mechanism Indication Route Enrollment Clintrials.gov Status
Eli Lilly & Daiichi Sankyo Prasugrel (Effient) Platelet inhibition Prophylaxis Oral 240, HbSS, HbSβ0
2 years ≥ 17 years
NCT01794000 Open
Emmaus Medical L-glutamine vs placebo Anti-oxidant Prophylaxis Oral 230, HbSS, HbSβ0
≥5 years
NCT01179217 Completed;
ASH 2014 # 86
MAST Therapeutics MST-188 Anti-adhesion Acute VOC treatment IV 388, HbSS, HbSβ0, HbSβ+, HbSC
4 years ≥ 65 years
NCT01737814 Open (EPIC)
GlycoMimetics GM-1070 (Rivipansel) Anti-P-selectin Acute VOC treatment IV 350, HbSS, HbSβ0, HbSβ+, HbSC
≥6 years
NCT02187003 Phase 2 completed; [100]
BC Children’s Hospital and University of BC, Vancouver MgSO4 Vasodilation? Pain reduction? Acute VOC treatment IV 120 (98 enrolled), HbSS, HbSβ0, HbSβ+, HbSC
4 years ≥ 18 years
NCT00313963 Completed (MAST); [17]
PECARN (Pediatric Emergency Care Applied Research Network) MgSO4 Vasodilation? Pain reduction? Acute VOC treatment IV 208
HbSS, HbSβ0, HbSβ+, HbSC
4 years ≥ 21 years
NCT01197417 Completed (MAGIC);
ASH 2014 # 88
Academisch Medisch Centrum - Universiteit van Amsterdam N-Acetylcysteine Anti-oxidant Prophylaxis Oral 140, HbSS, HbSβ0, HbSβ+, HbSC
≥12 years
NCT01849016 Open
Duke University Medical Center Apixaban (Eliquis) Anticoagulant Prophylaxis Oral 60, HbSS, HbSβ0, HbSC
18 years ≥ 80 years
NCT02179177 Not yet open

The SUSTAIN trial is a phase 2 multicenter trial currently assessing whether SelG1 (Selexys Pharmaceuticals, Oklahoma City, OK and Novartis Pharmaceuticals), a humanized monoclonal antibody to P-selectin, is safe and effective when given IV to patients on or off hydroxyurea (Table II). ARC5690, an anti-P-selectin aptamer, was considered for preclinical development by Archemix, but this company is now in liquidation (Table IV) [101].

Almost 15 years ago, IV Poloxamer 188 (Flocor), purified surfactant, was tested in a large phase 3 clinical trial to assess its efficacy in reducing the duration of painful crises. A limited effect was seen in children and subjects concomitantly treated with hydroxyurea [102]. A smaller study subsequently showed measurable improvement in microcirculatory parameters during VOC [103]. A second phase 3 trial with this compound, now renamed MST-188 is currently under way (Mast Therapeutics, San Diego, CA) to test its effectiveness in shortening VOC in children and adults (Table III).

Very late antigen 4 (VLA-4) or α4β1 is a cell surface integrin that mediates reticulocyte interactions with the endothelium, vascular adhesion molecule 1 (VCAM1), and plasma fibrinogen. Natalizumab, a recombinant humanized antibody currently approved for relapsing multiple sclerosis (MS) and Crohn’s disease, binds to the α4 subunit of VLA-4. Sickle cell reticulocyte and leukocyte adhesion to VCAM-1 was blocked in whole blood samples obtained from subjects with SCD and saturated with Natalizumab at plasma trough concentrations measured in MS and Crohn’s disease patients, (ASH 2014, #221).

RBC have beta adrenergic receptors that may, under adrenergic stress, activate some adhesion molecules like RBC LU-BCAM, favoring initiation of VOC as well as generating vasospasm. Propranolol, a frequently used beta-blocker in children and adults, has been shown to inhibit epinephrine upregulation of sickle RBC endothelial adhesion in animal studies and in a phase 1 clinical study [104].

PF04447943 (PDE9i, Pfizer Inc.), an inhibitor of phosphodiesterase-9A enzyme, was unsuccessfully tested in a Phase 2 trial in Alzheimer’s disease [105]. This compound decreased adhesion in a SCD mouse model (ASH 2014, # 2694). A phase 1 clinical trial is planned for patients with SCD on or off hydroxyurea (Table I).

A phase 1 study in 15 patients with Hb SS or Hb Sβthal experiencing an acute painful crisis showed changes in neutrophil activation markers following a single infusion of intravenous immunoglobulin (IVIG, from 100 to 800 mg/kg). Although not significant, a trend toward increased re-admissions with high dose IVIG was noted in this trial [106].

Vasculopathy of sickle cells and coagulation

Virchow’s triad describes the three broad categories of factors leading to thrombosis. In SCD, endothelial damage (caused by direct vascular damage from ischemic injury and free hemoglobin), stasis (consequence of decreased blood flow secondary to erythrocyte adhesion and increased viscosity), and hypercoagulability (due to externalization of phosphatidlserine on erythrocytes promoting subsequent thrombin generation, platelet adhesion and WBC activation) are potential contributors to thrombus formation. Lastly, platelet elevation and activation in SCD accelerates the hemostasis cascade.

Eptifibatide, an inhibitor of the platelet αIIbβ3 receptor pathway was studied in a small group of patients with SCD during acute VOC but showed no measurable benefits [16]. Abciximab (ReoPro, a platelet glycoprotein IIb/IIIa receptor antagonist), is currently being studied as IV infusion in the treatment of acute VOC in SCD patients 5 to 25 years of age. Ticagrelor and Prasugrel are oral platelet aggregation inhibitors that act via inhibition of the adenosine diphosphate receptor P2Y12. A phase 2 study tested oral administration of Prasugrel (5 mg/day for 30 days, n = 41) vs. placebo (n = 21) in patients with SCD, demonstrating its safety and some changes in markers of platelet activation [107]. Prasugrel is currently in a phase 3, double-blind, placebo-controlled study involving up to 220 pediatric patients, which will test its efficacy in reducing VOC, as a composite point of either painful crisis or acute chest syndrome. This study will use the VerifyNow® P2Y12 test, which quantifies the extent of platelet function inhibition via the P2Y12 pathway, to titrate Prasugrel to a maintenance dose for a treatment period between 9 and 24 months.

Apixaban, a factor Xa inhibitor, is being investigated as a prophylactic agent in a phase 3 study (Table III). The main objective of its study is to reduce mean daily pain scores in patients greater than 18 years of age with SCD. Rivaroxaban, also a factor Xa inhibitor, is under evaluation in a phase 2 trial, to examine its effects on vascular cell adhesion molecule-1 (VCAM-1) and interleukin-6 (IL-6) levels after 4 weeks of therapy (Table II).

White cells and other cellular and soluble mediators of inflammation

Several classic studies have shown that the baseline WBC is a strong predictor of ACS [108], silent strokes [109], and early mortality in patients with SCD [20]. It is well known that inflammation is a key component of the pathophysiology of SCD. Several anti-inflammatory drugs have been and are used in the treatment of acute and chronic events in SCD, including NSAID and steroids. Hydroxyurea therapy significantly decreases WBC counts, but how much this change contributes to the beneficial effects of hydroxyurea remains undetermined. Ischemia-reperfusion injury with release of inflammatory cytokines has also been invoked as a potential mechanism leading to acute and chronic tissue damage. More recent studies have identified regulatory steps leading to the inflammatory state of SCD, which are also potential candidates for therapeutic intervention [110112].

Invariant Natural Killer T (iNKT) cells are currently being investigated as a potential mediator of the inflammatory state of SCD, and activation of these cells has been demonstrated during VOC [13]. The humanized monoclonal antibody NKTT120 has been shown to safely deplete iNKT cells in adult sickle cell patients up to a dose of 0.3 mg/kg (ASH 2014, # 2178). iNKT cells are laden with adenosine receptors. Regadenoson, an adenosine receptor 2A agonist, currently used to increase coronary blood flow during cardiac nuclear stress scan, (Lexiscan, Astellas USLLC) is being studied for the acute treatment of VOC, with the intent to down-regulate the inflammatory cascade that is initiated by iNKT activation. In a phase 1 study in patients with SCD (21 at baseline and 6 during VOC), a 24-h infusion of regadenoson was shown to result in decrease iNKT activation [13]. A double-blind, placebo controlled, phase 2 study is ongoing to determine the effects of a 48-h IV infusion of regadenoson on iNKT cell activation in 96 patients with sickle cell anemia admitted for acute VOC or mild to moderate ACS (Table II). Other adenosine receptor ligands are being considered for potential development in SCD, such as PNQ103 (Advinus Therapeutics, Pune, India), and A2a PAM (Addex Therapeutics, Geneve Switzerland).

Zileuton (ZYFLO CR®, Chiesi) is a structural analog of hydroxyurea, which decreases leukotriene production by inhibiting 5-Lipoxygenase, and is currently marketed for prevention of asthma in children and adults. Zileuton also induces HbF in erythroid progenitors through a mechanism that involves l-arginine/nitric oxide/cyclic GMP [113]. A phase 1 study in 11 patients with SCD showed safety of higher doses than those approved for asthma [12], supporting the feasibility of a future phase 2 study.

Dimethyl fumarate (Tecfidera, Biogen Idec) currently approved in the USA for the treatment of relapsing multiple sclerosis, is being considered for SCD based on both its general anti-inflammatory properties as well as the ability to activate NRF2 signaling which is involved in drug-induced HbF expression [114,115].

The use of the inhaled corticosteroid, Mometasone, in sickle cell patients with cough or wheeze but no diagnosis of asthma, is also currently being investigated to assess if it can decrease general pulmonary inflammation. ReveraGen Biopharma (Silver Spring, MD) is currently developing VBP15, a “dissociative” steroid, which retains steroidal efficacy with reduced side effects, for Duchenne Muscular dystrophy, and has received NIH funding to begin its development for SCD. This is relevant for SCD because there are concerns about oral glucocorticoids possibly promoting VOC and accelerating the development of osteonecrotic complications.

Atorvastatin and Simvastatin, two frequently used lipid lowering medications, have been considered for SCD. However, Atorvastatin treatment (10–20 mg/day) did not produce measurable improvements in vasodilatory responses in a small cohort of SCD patients [116]. Simvastatin increased nitric oxide metabolites while decreasing C-reactive protein (CRP) and interleukin-6 (IL-6) in patients with SCD [117]. Lovaza, an omega-3 fatty acid ethyl ester, is also being investigated for its role in inflammation in pediatric SCD patients (Table II). In sickle mice, a diet supplemented with omega-3 fatty acid (fish oil) reduced several systemic inflammatory biomarkers and improved hypoxia-reoxygenation associated organ damage [118]. Preclinical data show a possible role for inhibition of the mitogen-activated protein kinase ERK1/2 in reducing adhesion and vaso-occlusion (Table IV) [119].

NO, arginine, and hemolysis as a key mediators of vasculopathy

Nitric oxide is a potent vasodilator produced from the metabolism of L-arginine by NO synthase, and plays a key role in vascular physiology and pathophysiology. In SCD, the transfer of NO from HbS to the membrane is impaired, resulting in an impaired capability of red cells to mediate vasodilation [120]. The chronic hemolytic state of SCD increases plasma Hb values and substantially limits NO bio-availability, resulting in a NO-deficient state [121]. This NO-deficient state is believed to be the main determinant for the development of pulmonary hypertension [122124], although there have been different estimates about the true prevalence of this complication [125], and some spirited academic debates about the hemolysis hypothesis [125127]. Initial studies showed some promise for inhaled NO in the treatment of acute VOC [128], but a large randomized, placebo-controlled, double-blind study, showed no effect on the time of crisis resolution [129]. Another study on inhaled NO for the treatment of acute crises in pediatric patients was terminated early due to slow enrollment, but no data are yet available about this trial (Table II). Some case reports have suggested a possible therapeutic role for inhaled NO in ACS [130,131]. A case-controlled trial of inhaled NO in adult patients with ACS has recently been completed in Creteil, France and results have been submitted for publication. Patients with SCD exhibit a dysfunctional regulation of arterial tone, which may impair response to variation in blood flow or shear stress [132,133]. Despite abundant scientific evidence and strong rationale, studies targeting NO and associated regulatory pathways have been largely disappointing.

A trial of sildenafil in SCD disease patients with pulmonary hypertension (TRV ≥2.7 m/s and 6 min walk distance between 150 and 500 m) was terminated early due to serious adverse events in the sildenafil arm (mostly increased hospitalization for pain) [134]. A small study on prevention of recurrent ischemic priapism with sildenafil had inconclusive results [135]. Similarly, studies assessing endothelin receptor blockade (ASSET-1 and -2, with Bosentan) as a potential therapy for pulmonary hypertension in SCD were inconclusive due to poor enrollment [136]. Safety and efficacy of Macitentan (Actelion, Switzerland) will be tested in a single center open label trial for precapillary pulmonary hypertension in SCD at Boston University (Table II).

Low levels of plasma arginine have been demonstrated in steady state SCD, with further decreases due to acute events and chronic vascular damage. Human studies have been performed with arginine, based on the notion that administration of this compound may improve the relative NO deficiency in SCD, with conflicting results [137]. Questions have been raised about what should be the optimal oral arginine dose, suggesting that not enough arginine was used in a negative NIH-sponsored, phase 2 trial, whose results have never been formally published. A more recent study compared parenteral L-arginine (100 mg/kg tid for 5 days) and placebo administration in 38 children with SCD. Although length of stay was not affected, parenteral opioid use (~50%) and pain scores were significantly diminished with L-arginine [138]. It has also been suggested that administration of arginine and hydroxyurea may be superior to arginine monotherapy.

Some key mechanisms associated with hemolysis have been identified as toxic to the body and contributory to the pathophysiology seen in SCD. Hemolysis leads to the release of free hemoglobin into the extravascular space, the depletion of nitric oxide, and the creation of toxic free radicals and hemin [139]. Haptoglobin itself is protective and by binding to hemoglobin prevents movement of hemoglobin across endothelial layers. It also limits NO depletion and thus the release of free radicals and hemin. However, in SCD, haptoglobin is readily depleted and free hemoglobin must be cleared by heme oxygenase-1 (HO-1), as the presence of free hemoglobin can cause multiorgan damage. Preclinical studies show that exogenous haptoglobin decreases the production of HO-1, likely due to a decrease in the presence of free hemoglobin [140]. Bio Products Laboratory has received EU Orphan Drug designation for a preparation of haptoglobin to be administered IV to patients with SCD. Preclinical studies in dogs have shown that this animal model can be used to estimate clearance of haptoglobin-bound Hb [141].

Vasodilation-increased blood flow-perfusion-oxygenation

MgSO4, known for his vasodilatory properties, has also been studied in SCD. However, as detailed above, two randomized clinical trials showed no effect of IV MgSO4 on the resolution of acute painful crises (ASH 2014 # 88) [17].

A hemoglobin based blood substitute was tested in a clinical trial in 1997, under the assumption that this oxygen carrier could potentially reverse VOC [142]. While no toxicity was observed in this first study, subsequent studies for other indications not related to SCD have shown an association with death and myocardial infarction [143], resulting in diminished enthusiasm about these compounds as therapeutic agents.

MP4CO was a human hemoglobin, obtained from blood donors, conjugated with polyethylene glycol (PEG) and saturated with CO. It was produced with the idea that CO delivery to ischemic areas would improve perfusion and reduce microvascular stasis and with the assumption that 5 to 10% HbCO would be nontoxic and could limit or reverse HbS polymerization as well as producing a vasodilatory effect. Preclinical studies in sickle mice showed positive changes in inflammatory parameters and mortality [85], but no data are available on a phase Ib study funded by Sangart, a company which is no longer active (Table I). Sanguinate, a pegylated bovine carboxyhemoglobin was tested in a phase 1 trial (Table I). According to a press release on April 14, 2015 by Prolong Pharmaceuticals (South Plain-field, NJ; http://www.prolongpharma.com/press-14APR2015) phase 2 trials are planned for VOCs and leg ulcers. A topical sodium nitrate 2% cream was shown to be safe and well tolerated in a phase 1 study in 18 patients with SCD [14]. Increases in per-wound cutaneous blood flow and decreased ulcer size were also observed in this trial.

Oxidative damage

Oxidative stress is thought to play a role in the pathophysiology of SCD, both at the erythrocyte level, as well as in the development of VOC. Several commonly used supplements are currently being investigated based on their anti-oxidant properties. Sorghum bicolor extract, Jobelyn is often used as a dietary supplement in Nigeria. Its antioxidant and anti-inflammatory properties have led to its investigation as a potential SCD agent, with an ongoing trial in Lagos, Nigeria comparing the effect of daily Jobelyn at different daily doses (500 mg, 250 mg, and 2 mg) on quality of life (Table II).

It has been hypothesized that N-acetylcysteine (NAC) may help reduce oxidative damage associated with SCD, based on its capability of increasing antioxidant systems like the one based on glutathione. This is currently being tested in a phase 3 trial (Table III).

The amino acid L-glutamine has been shown to increase erythrocyte nicotinamide adenine dinucleotide redox potential (NADH). A randomized, placebo controlled trial evaluated the efficacy and safety in SCD patients >5 years of age and with a history of at least two VOC in the previous 12 months. In the 2014 ASH abstract (ASH 2014, # 86) clinical benefit was reported in the treatment arm with patients receiving L-glutamine having significantly fewer days to first VOC, number of painful crises, and hospital days. However, Form 10-K filed by Emmaus Life Sciences Inc. with the U.S. Securities and Exchange Commission, have disclosed significant concerns raised by the FDA, namely that the primary endpoint of the trial did not reach statistical significance, that the results were inconsistent among regions, and that the reduction in painful crises, was not clinically meaningful and was inconsistent across regions. These concerns resulted in the recommendation that a second trial be conducted in patients with higher baseline levels of painful crises [144].

Cell-based therapies

Cell based therapies currently provide the only curative options for SCD, and they comprise both gene therapies and stem cell transplantation (Table V).

TABLE V.

Cell-based therapies: applications to sickle cell disease

Company Protocol/agent Mechanism Clintrials. gov Enrollment target Status
Gene Therapy
Blue Bird Bio LentiGlobin BB305 Drug Product Gene therapy NCT02140554 8
HbSS, HbSβ0
≥ 18 years
Open
Blue Bird Bio LentiGlobin BB305 Drug Product Gene therapy NCT02151526 7
HbSS, HbSβ0
Beta-thalassemia major
5 years ≥ 35 years
ASH 2014 # 549 & 4797
Children’s Hospital LA Lenti/βAS3-FB lentiviral vector Transduction to express an antisickling (βAS3) gene Gene therapy NCT02247843 6
HbSS, HbSβ0
≥ 18 years
Open
Children’s Hospital Medical Center, Cincinnati Gamma Globin Lentivirus Vector-mediated gene transfer Gene therapy NCT02186418 10
Severe SCD
18 years ≥ 35 years
Open
Errant Gene therapeutics Lentiviral vector, TNS 9.55.3 Gene therapy N/A N/A Open Phase 1 in Beta-thalassemia (NCT01639690)
OrphageniX Targeted Gene Alteration (TGA) Sickle gene repair N/A N/A Preclinical
Sangamo Biosciences (and Biogen Idec?) ZFN-mediated gene correction Sickle gene repair N/A N/A Preclinical
Editas Medicine CRISPR-Cas9 & TALENs Sickle gene repair-orincrease HbF N/A N/A Preclinical
Boston Children’s Hospital BCL11A knockdown HbF induction N/A N/A Not yet open
HSC Transplant
Cellerant Therapeutics CLT-001 Purify stem cells for HSCT N/A N/A Discontinued
Gamida Cell Ltd Nicotinamide (NiCord®) Expand stem cells, HSCT NCT01590628 10
HbSS, HbSβ0, HbSC2 years ≥ 21 years
Open
Morphogenesis SCPF Expand stem cells N/A N/A Discontinued
Assistance Publique -Hôpitaux de Paris Plerixafor Mobilization of stem cells NCT02212535 5
HbSS, HbSβ0
≥ 18 years
Not yet open
University of Louisville, KY Alemtuzumab based conditioning Stable Mixed chimerism induction NCT01419704 30
hemoglobinopathies, bone marrow failure syndromes
≤ 45 years
DIscontinued
Masonic Cancer Center, University of Minnesota Stable Mixed chimerism induction NCT00176852 30HbSS, HbSβ0, Beta-thalassemia major
≤ 50 years
Open but not recruiting
Hackensack University Medical Center Alemtuzumab, fludarabine, and melphalan Reduced intensity HSCT NCT01877837 40
HbSS, HbSβ0, HbSβ+, HbSC
2 years ≥ 30 years
Open
University of Illinois at Chicago Alemtuzumab, 300 cGy TBI, and sirolimus Non-Myeloablative HSCT NCT01499888 15
HbSS, HbSβ0, HbSC
18 years ≥ 60 years
Open
University of Illinois at Chicago Alemtuzumab, 300 cGy TBI, post-SCT cyclophosphamide and sirolimus HLA-Haploidentical HSCT NCT02013375 110
Severe SCD
16 years ≥ 60 years
Open
Sidney Kimmel Comprehensive Fludarabine, cyclophosphamide, and, post-SCT sirolimus Partially HLA-Mismatched and NCT00489281 50
Severe SCD
2 years ≥ 70 years
Open
Emory University & NHLBI Busulfan, fludarabine, anti-thymocyte globulin Reduced intensity NCT01565616 15
Severe SCD
16 years ≥ 40 years
Open (STRIDE)
University of Texas Southwestern Medical Center Low dose irradiation, Alemtuzumab and sirolimus. Non-Myeloablative HSCT, Mobilized peripheral stem cells NCT02038478 50
Severe SCD
18 years ≥ 45 years
Open
University of Pittsburgh Reduced intensity NCT01962415 30
2 months ≥ 35 years
Open
Fred Hutchinson Cancer Research Center Treosulfan and fludarabine, ± low dose irradiation HSCT NCT00919503 68
≤ 54 years
Open
New York Medical College Hydroxyurea, azathioprine, fludarabine, busulfan, thiotepa, cyclophosphamide, and rabbit ATG Haploidentical T cell- depleted HSCT NCT01461837 35
Severe SCD
2 years ≥ 21 years
Open
NHLBI Low dose irradiation, alemtuzumab, sirolimus, and cyclophosphamide Non-Myeloablative Haploidentical Mobilized peripheral stem cells NCT00977691 192
Sever SCD and Beta-Thalassemia
≥ 2 years
Open
NHLBI Low dose radiation (300 rads), oral cyclophosphamide, pentostatin, and sirolimus Non-Myeloablative Mobilized peripheral stem cells NCT02105766 142 HbSS, HbSβ0, 16 years ≥ 80 years Up to 142 Open
NHLBI Low dose radiation, Alemtuzumab and Sirolimus HSCT NCT00061568 124
Severe SCD
2 years ≥ 65 years
Open
Case Comprehensive Cancer Center Fludarabine HSCT NCT02065596 25 subjects
HbSS, HbSβ0, HbSβ+, HbSC
≥ 18 years
Not yet open
Vanderbilt-Ingram Cancer Center Thymoglobulin, fludarabine cyclophosphamide sirolimus, 200 cGy TBI Non-myeloablative partially HLA matched and fully HLA matched NCT01850108 10
Severe SCD
2 years ≥ 70 years
Open

Gene therapies

Several companies have developed groundbreaking gene therapies for SCD that, using a lentiviral vector, insert a functional human beta or gamma globin gene into a patient’s own hematopoietic stem cells. These are then infused into a patient during an autologous stem cell transplant.

Bluebird Bio is currently assessing the safety and efficacy of the LentiGlobin BB305 in children and adults with SCD (trials NCT 02140554 and NCT02151526). The use of this vector, which contains a B-globin gene (BA-T87Q) and produces hemoglobin BA-T87Q has already been proven successful in patients with Beta-thalassemia major (ASH 2014, # 549). The therapeutic Hb was designed with a unique mutation which, while preserving normal O2 carrying properties, strongly inhibits HbS polymerization. In addition, the lack of a functional LTR, the use of erythroid specific promoters, and chromatin insulator element make the chance of leukemia unlikely. Preliminary results have been presented on the first SCD patient treated under this protocol, showing that after 6 months Hb AT87Q comprised 45% of Hb production with associated improvement in transfusion requirements and hemolytic parameters. One additional SCD patient has been treated under a similar protocol at the NIH.

The University of California, Los Angeles is currently investigating the use of the recombinant beta globin gene (HBBAS3) in a lentiviral vector [145]. It too has the T87Q blocking site. In vitro data have been recently published showing that therapeutic levels can be achieved for the production of an antisickling Hb using insulated, self-inactivating and erythroid-specific lentiviral vectors in cultured CD34+ cells from SCD patients [146].

The Children’s Hospital Medical Center at Cincinnati has developed a novel human gamma-globin gene vector. It is currently recruiting adult SCD patients for its phase 1 trial. Boston Children’s Hospital is set to open a phase 1/2 trial that unlike the approaches highlighted above, involves the insertion of a beta or gamma globin gene, seeks to knockout BCL11A thus increasing fetal hemoglobin and bypassing the mutated sickle globin gene.

Preclinical evidence shows promise for the use of zinc-finger nucleases (ZFNs) in site-specific correction of the HbS mutation in mouse and human sickle CD34+ precursors [147]. However, new powerful genome editing techniques like CRISPR are likely to revolutionize gene therapy for hemoglobinopathies [148].

Several companies, including Errant Gene therapeutics and Editas Medicine are currently working with CRISPR-Cas9 systems to develop strategies to modify the mutated sickle cell gene, while Sangamo Biosciences is attempting to develop Zinc Finger Nuclease (ZFN) mediated gene correction. OrphageniX has tried a different approach and is developing therapies based on targeted gene alteration (TGA) which involves the delivery of strategically mismatched oligonucleotides to the nucleus of the cell, feeding off of the body’s natural DNA repair processes.

Stem cell transplantation

Hematopoietic stem cell transplant (HSCT) is currently the only approved cure for SCD. In centers with considerable experience with this approach, and when patients are treated before transplant with hydroxyurea, or if the transplant took place after January 2000, event-free survival rates of 97.4% and 95.3%, respectively, have been reported [149,150]. Overall survival is similar for either bone marrow transplant or cord blood transplant [151].

However, the potential associated morbidity and mortality often make HSCT a less considered treatment option [7]. For many providers, the difficulty in recommending transplant lies in our inability to predict clinical severity before the advent of significant morbidity, which in some cases can greatly affect transplant outcomes. Guidelines recommending stem cell transplant as a therapy for severe disease as opposed to a cure for all patients with the mutation, has also led to underuse.

In reality, overall survival and disease free survival in select populations has been shown to be equal to that of patients treated symptomatically without transplant [152]. Therefore, experts recommend that symptomatic patients with an HLA-matched sibling undergo transplantation preferably when they are pre-school age [6]. However, a major problem within the sickle cell community is the lack of healthy, HLA-matched siblings. In those without a sibling match, transplantation is only recommended in the setting of a controlled clinical trial.

At present, there are several ongoing clinical trials that are studying the success of diverse myeloablative, nonmyeloablative, and reduced intensity regimens in both matched sibling and unrelated sibling donor transplants. A reduced intensity regimen is meant to decrease morbidity and mortality associated with transplant, especially in those with preexisting co-morbidities. It also increases the likelihood of mixed donor chimerism, which, if high enough in SCD, can result in predominance of donor cells in the circulation [153]. Mixed donor chimerism and the addition of antibodies against mature lymphocytes, namely anti-thymocyte globulin or alemtuzumab, provide additional graft versus host disease prophylaxis. Case Comprehensive Cancer Center is planning a trial that uses solely fludarabine for conditioning (NCT0206559). A preliminary study has provided encouraging evidence that fludarabine can be used to reduce exposure to busulfan and cyclophosphamide in children with SCD undergoing HLA-matched HSCT [154].

A nonmyeloablative regimen composed of low dose radiation, alemtuzumab, and sirolimus is also being studied in older patients by University of Texas Southwestern Medical Center, University of Illinois at Chicago (UIC), and the National Heart Lung and Blood Institute (NHLBI). UT Southwestern is using peripheral stem cells while NHLBI and UIC are using bone marrow as their stem cell source. In another study, NHLBI has proposed using the same conditioning regimen backbone with additional pentostatin and oral cyclophosphamide in patients considered at high risk for transplant failure (i.e. female recipients of male donors those with preexisting allo-antibodies and those receiving peripheral stem cells).

As not all patients have a fully matched HLA sibling for transplant or an unrelated match, haploidentical transplantation has been proposed as an alternative. New York Medical College, Vanderbuilt-Ingram Cancer Center, UIC, and NHLBI all have ongoing trials.

Treatment of pain in sickle cell disease

Ballas et al. described the hallmark of SCD as acute continuous pain as it is the most common reason for inpatient hospitalization and treatment in the emergency department (Table VI) [155]. At present the mainstay of treatment for pain in VOC is opioids. Several interventional trials are ongoing to assess alternative modalities to decrease either pain scores during and between VOC. Nationwide Children’s Hospital recently completed a trial investigating whether warmed saline given to children who present with VOC may decrease hospital admission, IV analgesic usage, and pain scores. The safety and efficacy of lidocaine 5% plasters in pediatric patients with SCD experiencing neuropathic pain is being investigated at the Centre Léon Bérard (Lyon, France). Results are not yet available for either of these trials. Still recruiting are trials comparing different opioids such as morphine, hydromorphone, gabapentin, and intranasal fentanyl to those investigating the benefits of music therapy, cannabis, inhaled steroids and nitrous oxide. All studies are welcomed by the SCD community as opioids alone only address one component of the pathophysiology behind VOC i.e. nociception, but fail to address both vaso-occlusion and inflammation. In addition, trials should focus on early treatment during the prodromal stage of VOC as well as on how to minimize the hyperalgesia and fear that often accompanies recurrent painful episodes.

TABLE VI.

Currently planned, ongoing, or recently completed clinical trials on treatment of pain in sickle cell disease

Company Drug Design Indication Phase Enrollment Endpoints Clintrials.gov Status
Duke University (NHLBI) Morphine Sulfate or Hydromorphone Randomized Patient-specific vs. standard dosing Acute VOC treatment 4 77
HbSS, HbSβ0, HbSβ+, HbSC
≥21 years
Pain scores NCT02222246 Not yet open
Children’s Healthcare of Atlanta Morphine or Nubain Randomized double blind Acute VOC treatment 3 40
HbSS, HbSβ0, HbSβ+, HbSC
6 years > 19 years
Pain control acute chest syndrome NCT01380197 Open but not recruiting; (COMPARE)
University Hospital Case Medical Center Music therapy vs. Music Listening vs. no intervention Randomized Prophylaxis 120
HbSS, HbSβ0, HbSβ+, HbSC
≥18 years
Pain intensity, relief and mood NCT02270060 Open
Centre Leon Berard Lidocaine 5% plaster (versatic 5%) Open-label Acute VOC treatment 2 39
6 years > 21 years
Pain scores NCT01314300 Completed
St. Jude Children’s Research Hospital Gabapentin vs. placebo Randomized Double blind Acute VOC treatment 2 190
1 years > 20 years
Pain scales NCT01954927 Open
University of California, San Francisco (NHLBI) Vaporized cannabis (4.7% THC/5.1% CBD) vs. placebo Randomized Crossover Double blind Chronic pain treatment 1–2 35
HbSS, HbSβ0, HbSβ+, HbSC
≥18 years
Pain scores NCT01771731 Open
Montefiore Medical Center Intranasal fentanyl citrate Randomized Double blind Acute VOC treatment 4 200
HbSS, HbSβ0, HbSβ+, HbSC
3 years > 21 years
Pain scores NCT01482091 Open
Columbia University Nitrous oxide 50% Non-randomized, open label Acute VOC treatment 2 12
HbSS, HbSβ0, HbSβ+, HbSC
8 years > 18 years
Pain scores NCT01891812 Open
Nationwide Children’s Hospital, Columbus, OH Warmed saline Randomized, open label Acute VOC treatment 80
HbSS, HbSβ0, HbSβ+, HbSC
4 years > 21 years
Rate of hospital admission NCT02316366 Completed
Brooklyn Hospital Center IV Ketamine Double blind, placebo-controlled Acute VOC treatment 2/3 106
HbSS, HbSβ0, HbSβ+, HbSC
≥18 years
Pain Scores and admission rates NCT02417298 Open
Makerere University, Kampala Uganda Low-dose Ketamine vs morphine Double blind Acute VOC treatment 4 240
HbSS, HbSβ0, HbSβ+, HbSC
7 years > 18 years
Pain scores NCT02434939 Open

Iron overload in sickle cell disease

Chronic transfusion therapy is still the main therapy for primary and secondary stroke prophylaxis (Table VII). Transfusion therapy is also widely used for acute chest syndrome. Thus, transfusional iron overload is a significant problem for patients with severe SCD. Chelation is typically started 2 to 3 years after chronic transfusion therapy is initiated or once ferritin exceeds 1,000 ng/mL. Unfortunately, current therapies are often not well tolerated, especially in the pediatric setting. Current therapies include oral deferasirox, subcutaneous or IV deferoxamine, and oral deferasirox in combination with deferiprone [156]. Previous studies have demonstrated the safety and efficacy of deferasirox compared with deferoxamine in patients with iron overload and SCD [157]. Apopharma is currently recruiting patients to assess the safety and efficacy of deferiprone compared with deferoxamine. The primary endpoint is change in liver iron concentration. A similar study, yet to open, will take place in Italy. It will include patients as young as 1 month of age and will assess successful chelation based on ferritin and cardiac MRI T2*. Shire recently terminated all clinical trials for deferitazole (SHD602) in the treatment of iron overload.

TABLE VII.

Studies on pharmacological treatment of iron overload in sickle cell disease

Company Drug Design Criteria Phase Enrollment Endpoints Clintrials.gov Status
ApoPharma Deferiprone (Ferriprox) vs deferoxamine (Desferal) Randomized open label ≥6 years; Baseline LIC > 7 mg/g dw 4 300 Liver iron concentration NCT02041299
UCTR2013-002181-39-GB
Open (FIRST)
Consorzio per Valutazioni Biologiche e Farmacologiche (EC) Deferiprone (Ferriprox) vs Deferasirox (Exjade) Randomized open label 1 month< 18 years 3 344 Ferritin and cardiac iron concentration NCT01825512 Not yet open
Shire plc (previously Ferrokin Biosciences) Deferitazole (SPD602 or SSP-004184) Non-randomized open label (NCT01927913 randomized open label) Variable 1 and 2 N/A Liver iron concentration NCT01363908
NCT01604941
NCT01671111
Withdrawn:
NCT02065401
NCT01927913
All trials discontinued July 2014

Additional therapeutic modalities

Orthopedic complications such as osteonecrosis of femur and humeral heads represent an important clinical feature of SCD [158,159]. In recent years, the outcome for procedures like total hip arthroplasty has improved substantially and is not different from patients with osteonecrosis not due to SCD [160].

Renal disease occurs early in SCD and may ultimately lead to chronic renal failure. The pathophysiology of the SCD nephropathy is complex, and the clinical manifestations are variable [161,162]. Hyperfiltration is a common finding in children with SCD, but hypofiltration can also be present in a smaller subset [163]. Hydroxyurea therapy can play a significant role in retarding the progression of renal disease in SCD [164]. The angiotensin II receptor losartan is currently being studied for the prevention of sickle nephropathy in children and young adults with SCD (Table II, CT01479439 and NCT02373241).

Impaired rheology plays a key role in the pathogenesis of the complications of HbSC disease, and is believe to result in the 70% incidence of retinopathy and 29% incidence of sensorineural ontological disease, substantially higher than in non-SCD populations [165,166]. Limited evidence suggests a beneficial effect of phlebotomy in preventing recurrence of acute complications [165].

Despite overwhelming evidence for the benefit of vaccination in children with SCD, pneumococcal vaccination rates are still suboptimal in the US and the incidence of invasive pneumococcal disease is still greater than in the general population [167,168]. Similarly disappointing data have been reported for adherence to penicillin prophylaxis and for influenza vaccination [169].

Relevant endpoints for SCD clinical trials

The clinical trial which resulted in the approval of hydroxyurea for adults with SCD remains so far the only prototype for current trials aiming at pharmacologic reductions of clinical complications of SCD [27]. To the best of our knowledge, the FDA has not allowed phase 3 trials based on biomarkers or other disease indicators outside of transfusion and acute events (acute chest and/or VOCs resulting in patient admission). Alternative assessment of pain and disease severity (pain diaries) have been proposed but not fully validated. Validation of novel endpoints for SCD, especially related to pain, remains an important target for future studies.

Methods

Information on clinical trials was obtained from several sources. (a) Clinical-Trials.gov (https://clinicaltrials.gov) was searched for studies including the term sickle; studies were not included if the status of the study was listed as completed (with a completion date before 2013), terminated, unknown, or withdrawn. Observation trials were not included. (b) The WHO International Clinical Trials registry platform http://apps.who.int/trialsearch/ and the European registry (https://www.clinicaltrialsregister.eu/) were searched for additional studies not listed in the ClinicalTrials.gov database. Studies were then organized based on clinical phase (1, 2, 3), with separate tables for studies on cell-based therapies, pain treatment and iron overload. Information for preclinical studies was obtained from literature searches and review of abstracts presented at the 2013, 2014 American Society of Hematology and 2014 and 2015 European Hematology Association meetings.

Conclusions

A substantial number of clinical trials are ongoing or planned to test new treatments for SCD. The majority of these trials is supported by private companies, which represents a profound and positive change compared with the prior decade. This is most likely the result of the significant investments in fundamental research on SCD pathophysiology that NIH supported in the past 30 years and the realization for many biotechnology and pharmaceutical companies that the orphan status of SCD provides unique advantages for drug development and marketing. We hope that these studies will result in the approval of new treatment for SCD, either as a single agent or in combination with hydroxyurea.

Treatment or prevention of VOC in SCD is the central aim of new therapies. They will permit a better quality of life and facilitate more normal educational or occupational activities and ultimately translate into a significantly longer life expectancy. The development of these novel therapies poses a significant challenge for the so-called curative therapeutic approaches, which will have to demonstrate substantial advantages in cost and short-term and long-term toxicities to be considered as a serious, realistic option. These improved, noncurative therapeutic options also provide unexpected ethical and moral challenges in considering interruption of pregnancy following prenatal diagnosis of SCD.

Acknowledgments

Contract grant sponsor: NIH Training Grants (to N.A.); Contract grant numbers: 5T32HL007574 and 5K12HL087164-09.

Contract grant sponsor: Doris Duke Charitable Foundation; Contract grant number: 2013010.

The authors thank Jonathan Stocker (Selexys Pharmaceuticals, Morrisville NC), and Dr. Martin Steinberg (Boston University Medical Center) for helpful feedback and comments.

Footnotes

Conflict of interest: All authors have no conflicts of interest to disclose

References

  • 1.Ballas SK, Lieff S, Benjamin LJ, et al. Definitions of the phenotypic manifestations of sickle cell disease. Am J Hematol. 2010;85:6–13. doi: 10.1002/ajh.21550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Yawn BP, Buchanan GR, Afenyi-Annan AN, et al. Management of sickle cell disease: Summary of the 2014 evidence-based report by expert panel members. JAMA. 2014;312:1033–1048. doi: 10.1001/jama.2014.10517. [DOI] [PubMed] [Google Scholar]
  • 3.Habibi A, Arlet JB, Stankovic K, et al. French guidelines for the management of adult sickle cell disease: 2015 update. Rev Med Intern. 2015;36:5S3–5S84. doi: 10.1016/S0248-8663(15)60002-9. [DOI] [PubMed] [Google Scholar]
  • 4.Beverung LM, Strouse JJ, Hulbert ML, et al. Health-related quality of life in children with sickle cell anemia: Impact of blood transfusion therapy. Am J Hematol. 2015;90:139–143. doi: 10.1002/ajh.23877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. [Last accessed on July 28, 2015]; http://www.nih.gov/news/health/nov2014/nhlbi-0000.htm.
  • 6.Angelucci E, Matthes-Martin S, Baronciani D, et al. Hematopoietic stem cell transplantation in thalassemia major and sickle cell disease: Indications and management recommendations from an international expert panel. Haematologica. 2014;99:811–820. doi: 10.3324/haematol.2013.099747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hsieh MM, Fitzhugh CD, Tisdale JF. Allogeneic hematopoietic stem cell transplantation for sickle cell disease: The time is now. Blood. 2011;118:1197–1207. doi: 10.1182/blood-2011-01-332510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sunshine HR, Hofrichter J, Eaton WA. Requirements for therapeutic inhibition of sickle hemoglobin gelation. Nature. 1978;275:238–240. doi: 10.1038/275238a0. [DOI] [PubMed] [Google Scholar]
  • 9.Eaton WA, Hofrichter J. Hemoglobin S gelation and sickle cell disease. Blood. 1987;70:1245–1266. [PubMed] [Google Scholar]
  • 10.Eaton WA, Hofrichter J. Sickle cell hemoglobin polymerization. Adv Prot Chem. 1990;40:63–279. doi: 10.1016/s0065-3233(08)60287-9. [DOI] [PubMed] [Google Scholar]
  • 11.Eaton WA, Hofrichter J. The biophysics of sickle cell hydroxyurea therapy. Science. 1995;268:1142–1143. doi: 10.1126/science.7539154. [DOI] [PubMed] [Google Scholar]
  • 12.Quarmyne M-O, Rayes O, Gonsalves CS, et al. A phase I trial of zileuton in sickle cell disease. Blood. 2013;122:993–993. [Google Scholar]
  • 13.Field JJ, Lin G, Okam MM, et al. Sickle cell vaso-occlusion causes activation of iNKT cells that is decreased by the adenosine A2A receptor agonist regadenoson. Blood. 2013;121:3329–3334. doi: 10.1182/blood-2012-11-465963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Minniti CP, Gorbach AM, Xu D, et al. Topical sodium nitrite for chronic leg ulcers in patients with sickle cell anaemia: A phase 1 dose-finding safety and tolerability trial. Lancet Haematol. 2014;1:e95–e103. doi: 10.1016/s2352-3026(14)00019-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Reid ME, El Beshlawy A, Inati A, et al. A double-blind, placebo-controlled phase II study of the efficacy and safety of 2,2-dimethylbutyrate (HQK-1001), an oral fetal globin inducer, in sickle cell disease. Am J Hematol. 2014;89:709–713. doi: 10.1002/ajh.23725. [DOI] [PubMed] [Google Scholar]
  • 16.Desai PC, Brittain JE, Jones SK, et al. A pilot study of eptifibatide for treatment of acute pain episodes in sickle cell disease. Thromb Res. 2013;132:341–345. doi: 10.1016/j.thromres.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Goldman RD, Mounstephen W, Kirby-Allen M, et al. Intravenous magnesium sulfate for vaso-occlusive episodes in sickle cell disease. Pediatrics. 2013;132:E1634–E1641. doi: 10.1542/peds.2013-2065. [DOI] [PubMed] [Google Scholar]
  • 18.Ferrone FA. The delay time in sickle cell disease after 40 years: A paradigm assessed. Am J Hematol. 2015:438–445. doi: 10.1002/ajh.23958. [DOI] [PubMed] [Google Scholar]
  • 19.Platt OS, Thorington BD, Brambilla DJ, et al. Pain in sickle-cell disease - Rates and risk-factors. N Engl J Med. 1991;325:11–16. doi: 10.1056/NEJM199107043250103. [DOI] [PubMed] [Google Scholar]
  • 20.Platt OS, Brambilla DJ, Rosse WF, et al. Mortality in sickle-cell disease - Life expectancy and risk-factors for early death. N Engl J Med. 1994;330:1639–1644. doi: 10.1056/NEJM199406093302303. [DOI] [PubMed] [Google Scholar]
  • 21.Alsultan A, Ngo D, Bae H, et al. Genetic studies of fetal hemoglobin in the Arab-Indian haplotype sickle cell-β0 thalassemia. Am J Hematol. 2013;88:531–532. doi: 10.1002/ajh.23434. [DOI] [PubMed] [Google Scholar]
  • 22.Mtatiro SN, Makani J, Mmbando B, et al. Genetic variants at HbF-modifier loci moderate anemia and leukocytosis in sickle cell disease in Tanzania. Am J Hematol. 2015;90:E1–E4. doi: 10.1002/ajh.23859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Barbosa CG, Aleluia ACM, Pacheco APAS, et al. Genetic modulation of HbF in Brazilians with HbSC disease and sickle cell anemia. Am J Hematol. 2013;88:923–924. doi: 10.1002/ajh.23538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sheehan VA, Luo Z, Flanagan JM, et al. Genetic modifiers of sickle cell anemia in the BABY HUG cohort: Influence on laboratory and clinical phenotypes. Am J Hematol. 2013;88:571–576. doi: 10.1002/ajh.23457. [DOI] [PubMed] [Google Scholar]
  • 25.Akinsheye I, Alsultan A, Solovieff N, et al. Fetal hemoglobin in sickle cell anemia. Blood. 2011;118:19–27. doi: 10.1182/blood-2011-03-325258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Platt OS, Orkin SH, Dover G, et al. Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia. J Clin Invest. 1984;74:652–656. doi: 10.1172/JCI111464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med. 1995;332:1317–1322. doi: 10.1056/NEJM199505183322001. [DOI] [PubMed] [Google Scholar]
  • 28.Steinberg MH, McCarthy WF, Castro O, et al. The risks and benefits of long-term use of hydroxyurea in sickle cell anemia: A 17. 5 year follow-up. Am J Hematol. 2010;85:403–408. doi: 10.1002/ajh.21699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wong TE, Brandow AM, Lim W, et al. Update on the use of hydroxyurea therapy in sickle cell disease. Blood. 2014;124:3850–3857. doi: 10.1182/blood-2014-08-435768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kinney TR, Helms RW, O’Branski EE, et al. Safety of hydroxyurea in children with sickle cell anemia: Results of the HUG-KIDS study, a phase I/II trial. Blood. 1999;94:1550–1554. [PubMed] [Google Scholar]
  • 31.Thornburg CD, Files BA, Luo Z, et al. Impact of hydroxyurea on clinical events in the BABY HUG trial. Blood. 2012;120:4304–4310. doi: 10.1182/blood-2012-03-419879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wang WC, Ware RE, Miller ST, et al. Hydroxycarbamide in very young children with sickle-cell anaemia: A multicentre, randomised, controlled trial (BABY HUG) Lancet. 2011;377:1663–1672. doi: 10.1016/S0140-6736(11)60355-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wang WC, Helms RW, Lynn HS, et al. Effect of hydroxyurea on growth in children with sickle cell anemia: Results of the HUG-KIDS Study. J Pediatr. 2002;140:225–229. doi: 10.1067/mpd.2002.121383. [DOI] [PubMed] [Google Scholar]
  • 34.Aygun B, Mortier NA, Smeltzer MP, et al. Hydroxyurea treatment decreases glomerular hyperfiltration in children with sickle cell anemia. Am J Hematol. 2013;88:116–119. doi: 10.1002/ajh.23365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Rana S, Houston PE, Wang WC, et al. Hydroxyurea and growth in young children with sickle cell disease. Pediatrics. 2014;134:465–472. doi: 10.1542/peds.2014-0917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Stettler N, McKiernan CM, Melin CQ, et al. Proportion of adults with sickle cell anemia and pain crises receiving hydroxyurea. JAMA. 2015;313:1671–1672. doi: 10.1001/jama.2015.3075. [DOI] [PubMed] [Google Scholar]
  • 37.Hankins JS, Wynn LW, Brugnara C, et al. Phase I study of magnesium pidolate in combination with hydroxycarbamide for children with sickle cell anaemia. Br J Haematol. 2008;140:80–85. doi: 10.1111/j.1365-2141.2007.06884.x. [DOI] [PubMed] [Google Scholar]
  • 38.Wang WF, Brugnara C, Snyder C, et al. The effects of hydroxycarbamide and magnesium on haemoglobin SC disease: Results of the multi-centre CHAMPS trial. Br J Haematol. 2011;152:771–776. doi: 10.1111/j.1365-2141.2010.08523.x. [DOI] [PubMed] [Google Scholar]
  • 39.Brun M, Bourdoulous S, Couraud PO, et al. Hydroxyurea downregulates endothelin-1 gene expression and upregulates ICAM-1 gene expression in cultured human endothelial cells. Pharmacogenomics J. 2003;3:215–226. doi: 10.1038/sj.tpj.6500176. [DOI] [PubMed] [Google Scholar]
  • 40.Bartolucci P, Chaar V, Picot J, et al. Decreased sickle red blood cell adhesion to laminin by hydroxyurea is associated with inhibition of Lu/BCAM protein phosphorylation. Blood. 2010;116:2152–2159. doi: 10.1182/blood-2009-12-257444. [DOI] [PubMed] [Google Scholar]
  • 41.Robinson AJ, Kashanin DF, O’Dowd F, et al. Montelukast inhibition of resting and GM-CSF-stimulated eosinophil adhesion to VCAM-1 under flow conditions appears independent of cysLT(1)R antagonism. J Leukoc Biol. 2008;83:1522–1529. doi: 10.1189/jlb.1007717. [DOI] [PubMed] [Google Scholar]
  • 42.Bartolucci P, Brugnara C, Teixeira-Pinto A, et al. Erythrocyte density in sickle cell syndromes is associated with specific clinical manifestations and hemolysis. Blood. 2012;120:3136–3141. doi: 10.1182/blood-2012-04-424184. [DOI] [PubMed] [Google Scholar]
  • 43.Rakotoson MG, Di Liberto G, Audureau E, et al. Biological parameters predictive of percent dense red blood cell decrease under hydroxyurea. Orphanet J Rare Dis. 2015;10:57. doi: 10.1186/s13023-015-0272-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kutlar A, Reid ME, Inati A, et al. A dose-escalation phase IIa study of 2,2-dimethylbutyrate (HQK-1001), an oral fetal globin inducer, in sickle cell disease. Am J Hematol. 2013;88:E255–E260. doi: 10.1002/ajh.23533. [DOI] [PubMed] [Google Scholar]
  • 45.Sankaran VG, Menne TF, Xu J, et al. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science. 2008;322:1839–1842. doi: 10.1126/science.1165409. [DOI] [PubMed] [Google Scholar]
  • 46.Bauer DE, Kamran SC, Lessard S, et al. An erythroid enhancer of BCL11A subject to genetic variation determines fetal hemoglobin level. Science. 2013;342:253–257. doi: 10.1126/science.1242088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Marianna P, Kollia P, Akel S, et al. Valproic acid, trichostatin and their combination with hemin preferentially enhance gamma-globin gene expression in human erythroid liquid cultures. Haematologica. 2001;86:700–705. [PubMed] [Google Scholar]
  • 48.Rosa RM, Bierer BE, Thomas R, et al. A study of induced hyponatremia in the prevention and treatment of sickle-cell crisis. N Engl J Med. 1980;303:1138–1143. doi: 10.1056/NEJM198011133032002. [DOI] [PubMed] [Google Scholar]
  • 49.Lew VL, Bookchin RM. Ion transport pathology in the mechanism of sickle cell dehydration. Physiol Rev. 2005;85:179–200. doi: 10.1152/physrev.00052.2003. [DOI] [PubMed] [Google Scholar]
  • 50.Brugnara C, Bunn HF, Tosteson DC. Regulation of erythrocyte cation and water content in sickle cell anemia. Science. 1986;232:388–390. doi: 10.1126/science.3961486. [DOI] [PubMed] [Google Scholar]
  • 51.Franco RS, Palascak M, Thompson H, et al. KCl cotransport activity in light versus dense transferrin receptor-positive sickle reticulocytes. J Clin Invest. 1995;95:2573–2580. doi: 10.1172/JCI117958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Franco RS, Palascak M, Thompson H, et al. Dehydration of transferrin receptor-positive sickle reticulocytes during continuous or cyclic deoxygenation: Role of KCl cotransport and extracellular calcium. Blood. 1996;88:4359–4365. [PubMed] [Google Scholar]
  • 53.Crable SC, Hammond SM, Papes R, et al. Multiple isoforms of the KC1 cotransporter are expressed in sickle and normal erythroid cells. Exp Hematol. 2005;33:624–631. doi: 10.1016/j.exphem.2005.02.006. [DOI] [PubMed] [Google Scholar]
  • 54.Brugnara C, Van Ha T, Tosteson DC. Acid pH induces formation of dense cells in sickle erythrocytes. Blood. 1989;74:487–495. [PubMed] [Google Scholar]
  • 55.Brugnara C, De Franceschi L, Alper SL. Inhibition of Ca(2+)-dependent K+ transport and cell dehydration in sickle erythrocytes by clotrimazole and other imidazole derivatives. J Clin Invest. 1993;92:520–526. doi: 10.1172/JCI116597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Etzion Z, Tiffert T, Bookchin RM, et al. Effects of deoxygenation on active and passive Ca2+ transport and on the cytoplasmic Ca2+ levels of sickle cell anemia red cells. J Clin Invest. 1993;92:2489–2498. doi: 10.1172/JCI116857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lew VL, Ortiz OE, Bookchin RM. Stochastic nature and red cell population distribution of the sickling-induced Ca2+ permeability. J Clin Invest. 1997;99:2727–2735. doi: 10.1172/JCI119462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lew VL, Tiffert T, Etzion Z, et al. Distribution of dehydration rates generated by maximal Gardos-channel activation in normal and sickle red blood cells. Blood. 2005;105:361–367. doi: 10.1182/blood-2004-01-0125. [DOI] [PubMed] [Google Scholar]
  • 59.Rivera A, Jarolim P, Brugnara C. Modulation of gardos channel activity by cytokines in sickle erythrocytes. Blood. 2002;99:357–363. doi: 10.1182/blood.v99.1.357. [DOI] [PubMed] [Google Scholar]
  • 60.Vandorpe DH, Xu C, Shmukler BE, et al. Hypoxia activates a Ca(2+)-permeable cation conductance sensitive to carbon monoxide and to GsMTx-4 in human and mouse sickle erythrocytes. PLoS One. 2010;5:e8732. doi: 10.1371/journal.pone.0008732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Joiner CH, Dew A, Ge DL. Deoxygenation-induced cation fluxes in sickle cells: Relationship between net potassium efflux and net sodium influx. Blood Cells. 1988;13:339–358. [PubMed] [Google Scholar]
  • 62.Joiner CH, Claussen W, Yasin Z, et al. Dipyridamole inhibits in vitro deoxygenation-induced cation fluxes in sickle red blood cells at the membrane concentration achievable in blood. Blood. 1997;90:125. [Google Scholar]
  • 63.De Franceschi L, Bachir D, Galacteros F, et al. Oral magnesium supplements reduce erythrocyte dehydration in patients with sickle cell disease. J Clin Invest. 1997;100:1847–1852. doi: 10.1172/JCI119713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.De Franceschi L, Bachir D, Galacteros F, et al. Oral magnesium pidolate: Effects of long-term administration in patients with sickle cell disease. Br J Haematol. 2000;108:248–289. doi: 10.1046/j.1365-2141.2000.01861.x. [DOI] [PubMed] [Google Scholar]
  • 65.Benjamin LJ, Berkowitz LR, Orringer E, et al. A collaborative, double-blind randomized study of cetiedil citrate in sickle cell crisis. Blood. 1986;67:1442–1447. [PubMed] [Google Scholar]
  • 66.Ataga KI, DeCastro LM, Swerdlow P, et al. Efficacy and safety of the Gardos channel inhibitor, ICA-17043, in patients with sickle cell anemia. Blood. 2004;104:33. doi: 10.1182/blood-2007-08-110098. [DOI] [PubMed] [Google Scholar]
  • 67.Brugnara C, Gee B, Armsby CC, et al. Therapy with oral clotrimazole induces inhibition of the Gardos channel and reduction of erythrocyte dehydration in patients with sickle cell disease. J Clin Invest. 1996;97:1227–1234. doi: 10.1172/JCI118537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ataga KI, Reid M, Ballas SK, et al. Improvements in haemolysis and indicators of erythrocyte survival do not correlate with acute vaso-occlusive crises in patients with sickle cell disease: A phase III randomized, placebo-controlled, double-blind study of the Gardos channel blocker senicapoc (ICA-17043) Br J Haematol. 2011;153:92–104. doi: 10.1111/j.1365-2141.2010.08520.x. [DOI] [PubMed] [Google Scholar]
  • 69.Rivera A. Reduced sickle erythrocyte dehydration in vivo by endothelin-1 receptor antagonists. Am J Physiol Cell Physiol. 2007;293:C960–966. doi: 10.1152/ajpcell.00530.2006. [DOI] [PubMed] [Google Scholar]
  • 70.Castro O, Pollen WN, Finke H, et al. Improvement of sickle cell anemia by iron-limited erythropoiesis. Am J Hematol. 1994;47:74–81. doi: 10.1002/ajh.2830470203. [DOI] [PubMed] [Google Scholar]
  • 71.Boucher N, Manigne P, Kanfer A, et al. Prevention of sickle cell crises with multiple phlebotomies. Arch Pediatr. 2000;7:249–255. doi: 10.1016/s0929-693x(00)88740-0. [DOI] [PubMed] [Google Scholar]
  • 72.De Furia Fg, Miller DR, Cerami A, Cerami A, Manning JM, et al. The effects of cyanate in vitro on red blood cell metabolism and function in sickle cell anemia. J Clin Invest. 1972;51:566–574. doi: 10.1172/JCI106845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Deiderich DA, Trueworthy RC, Gill P, et al. Hematologic and clinical responses in patients with sickle cell anemia after chronic extracorporeal red cell carbamylation. J Clin Invest. 1976;58:642–653. doi: 10.1172/JCI108511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Keidan AJ, White RD, Huehns ER, et al. Effect of BW12C on oxygen affinity of haemoglobin in sickle-cell disease. Lancet. 327:831–834. doi: 10.1016/s0140-6736(86)90941-4. [DOI] [PubMed] [Google Scholar]
  • 75.Orringer E, Huffman J, Guaspari L, et al. A clinical-study of the safety, pharmacokinetics, and pharmacodynamics of intravenous infusions of 12C79 in sickle-cell disease patients not in crisis. Clin Res. 1991;39:238–238. [Google Scholar]
  • 76.Abraham D, Mehanna A, Wireko F, et al. Vanillin, a potential agent for the treatment of sickle cell anemia. Blood. 1991;77:1334–1341. [PubMed] [Google Scholar]
  • 77.Garcia AF, Cabal C, Losada J, et al. In vivo action of Vanillin on delay time determined by magnetic relaxation. Hemoglobin. 2005;29:181–187. doi: 10.1081/hem-200066305. [DOI] [PubMed] [Google Scholar]
  • 78.Hannemann A, Cytlak UMC, Gbotosho OT, et al. Effects of o-vanillin on K+ transport of red blood cells from patients with sickle cell disease. Blood Cells Mol Dis. 2014;53:21–26. doi: 10.1016/j.bcmd.2014.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Abdulmalik O, Ghatge MS, Musayev FN, et al. Crystallographic analysis of human hemoglobin elucidates the structural basis of the potent and dual antisickling activity of pyridyl derivatives of vanillin. Acta Crystallogr Sect D Biol Crystallogr. 2011;67:920–928. doi: 10.1107/S0907444911036353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Verduzco LA, Nathan DG. Sickle cell disease and stroke. Blood. 2009;114:5117–5125. doi: 10.1182/blood-2009-05-220921. [DOI] [PubMed] [Google Scholar]
  • 81.Arya R, Rolan PE, Wootton R, et al. Tucaresol increases oxygen affinity and reduces haemolysis in subjects with sickle cell anaemia. Br J Haematol. 1996;93:817–821. doi: 10.1046/j.1365-2141.1996.d01-1744.x. [DOI] [PubMed] [Google Scholar]
  • 82.Abdulmalik O, Safo MK, Chen QK, et al. 5-hydroxymethyl-2-furfural modifies intracellular sickle haemoglobin and inhibits sickling of red blood cells. Br J Haematol. 2005;128:552–561. doi: 10.1111/j.1365-2141.2004.05332.x. [DOI] [PubMed] [Google Scholar]
  • 83.Hannemann A, Cytlak UM, Rees DC, et al. Effects of 5-hydroxymethyl-2-furfural on the volume and membrane permeability of red blood cells from patients with sickle cell disease. J Physiol Lond. 2014;592:4039–4049. doi: 10.1113/jphysiol.2014.277681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lawrence MP, Mendelsohn LG, Saiyed R, et al. Phase 1 clinical trial of the candidate antisickling agent Aes-103 in adults with sickle cell anemia. Blood. 2013;122:1009–1009. [Google Scholar]
  • 85.Belcher JD, Young M, Chen CS, et al. MP4CO, a pegylated hemoglobin saturated with carbon monoxide, is a modulator of HO-1, inflammation, and vaso-occlusion in transgenic sickle mice. Blood. 2013;122:2757–2764. doi: 10.1182/blood-2013-02-486282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kaul DK, Chen D, Zhan J. Adhesion of sickle cells to vascular endothelium is critically dependent on changes in density and shape of the cells. Blood. 1994;83:3006–3017. [PubMed] [Google Scholar]
  • 87.Kaul DK. Sickle red cell adhesion: Many issues and some answers. Transfus Clin Biol. 2008;15:51–55. doi: 10.1016/j.tracli.2008.03.012. [DOI] [PubMed] [Google Scholar]
  • 88.Nelson RM, Cecconi O, Roberts WG, et al. Heparin oligosaccharides bind L- and P-selectin and inhibit acute inflammation. Blood. 1993;82:3253–3258. [PubMed] [Google Scholar]
  • 89.Finger EB, Puri KD, Alon R, et al. Adhesion through L-selectin requires a threshold hydrodynamic shear. Nature. 1996;379:266–269. doi: 10.1038/379266a0. [DOI] [PubMed] [Google Scholar]
  • 90.Lawrence MB, Kansas GS, Kunkel EJ, et al. Threshold levels of fluid shear promote leukocyte adhesion through selectins (CD62L,P,E) J Cell Biol. 1997;136:717–727. doi: 10.1083/jcb.136.3.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Matsui NM, Varki A, Embury SH. Heparin inhibits the flow adhesion of sickle red blood cells to P-selectin. Blood. 2002;100:3790–3796. doi: 10.1182/blood-2002-02-0626. [DOI] [PubMed] [Google Scholar]
  • 92.Koenig A, Norgard-Sumnicht K, Linhardt R, et al. Differential interactions of heparin and heparan sulfate glycosaminoglycans with the selectins. Implications for the use of unfractionated and low molecular weight heparins as therapeutic agents. J Clin Invest. 1998;101:877–889. doi: 10.1172/JCI1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kutlar A, Ataga KI, McMahon L, et al. A potent oral P-selectin blocking agent improves microcirculatory blood flow and a marker of endothelial cell injury in patients with sickle cell disease. Am J Hematol. 2012;87:536–539. doi: 10.1002/ajh.23147. [DOI] [PubMed] [Google Scholar]
  • 94.Qari MH, Aljaouni SK, Alardawi MS, et al. Reduction of painful vaso-occlusive crisis of sickle cell anaemia by tinzaparin in a double-blind randomized trial. Thromb Haemost. 2007;98:392–396. [PubMed] [Google Scholar]
  • 95.Batchvarova M, Shan SQ, Zennadi R, et al. Sevuparin reduces adhesion of both sickle red cells and leukocytes to endothelial cells in vitro and inhibits vaso-occlusion in vivo. Blood. 2013;122:2. [Google Scholar]
  • 96.Wun T, Styles L, DeCastro L, et al. Phase 1 study of the E-selectin inhibitor GMI 1070 in patients with sickle cell anemia. PLos One. 2014;9:12. doi: 10.1371/journal.pone.0101301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.De Castro LM, Wun T, Lanzkron S, et al. Effects of GMI 1070, a pan-selectin inhibitor, on pain intensity and opioid utilization in sickle cell disease. Blood. 2013;122:775. [Google Scholar]
  • 98.McCavit TL, Krishnamurti L, Hsu LL, et al. An analysis of the pediatric sub-group from the phase 2 study of GMI 1070-A novel agent for the vaso-occlusive crisis of sickle cell anemia. Blood. 2013;122:2206. [Google Scholar]
  • 99.Telen MJ, Wun T, McCavit TL, et al. GMI 1070: Reduction in time to resolution of vaso-occlusive crisis and decreased opioid use in a prospective, randomized, multi-center double blind, adaptive phase 2 study in sickle cell disease. Blood. 2013;122 doi: 10.1182/blood-2014-06-583351. (abstract) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Telen MJ, Wun T, McCavit TL, et al. Randomized phase 2 study of GMI-1070 in SCD: reduction in time to resolution of vaso-occlusive events and decreased opioid use. Blood. 2015;125:2656–2664. doi: 10.1182/blood-2014-06-583351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Gutsaeva DR, Parkerson JB, Yerigenahally SD, et al. Inhibition of cell adhesion by anti–P-selectin aptamer: A new potential therapeutic agent for sickle cell disease. Blood. 2011;117:727–735. doi: 10.1182/blood-2010-05-285718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Orringer EP, Casella JF, Ataga KI, et al. Purified poloxamer 188 for treatment of acute vaso-occlusive crisis of sickle cell disease - A randomized controlled trial. JAMA. 2001;286:2099–2106. doi: 10.1001/jama.286.17.2099. [DOI] [PubMed] [Google Scholar]
  • 103.Cheung ATW, Chan MS, Ramanujam S, et al. Effects of poloxamer 188 treatment on sickle cell vaso-occlusive crisis: Computer-assisted intravital microscopy study. J Invest Med. 2004;52:402–406. doi: 10.1136/jim-52-06-35. [DOI] [PubMed] [Google Scholar]
  • 104.De Castro LM, Zennadi R, Jonassaint JC, et al. Effect of propranolol as antiadhesive therapy in sickle cell disease. Clin Transl Sci. 2012;5:437–444. doi: 10.1111/cts.12005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Schwam E, Nicholas T, Chew R, et al. A multi-center, double-blind, placebo-controlled trial of the PDE9A inhibitor. Curr Alzheimer Res. 2014;11:413–421. doi: 10.2174/1567205011666140505100858. [DOI] [PubMed] [Google Scholar]
  • 106.Manwani D, Chen G, Carullo V, et al. Single-dose intravenous gamamglobulin can stabilize neutrophil Mac-1 activation in sickle cell pain crises. Am J Hematol. 2015;90:381–385. doi: 10.1002/ajh.23956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wun T, Soulieres D, Frelinger AL, et al. A double-blind, randomized, multicenter phase 2 study of prasugrel versus placebo in adult patients with sickle cell disease. J Hematol Oncol. 2013;6:17. doi: 10.1186/1756-8722-6-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Castro O, Brambilla DJ, Thorington B, et al. The acute chest syndrome in sickle cell disease: incidence and risk factors. The Cooperative Study of Sickle Cell Disease. Blood. 1994;84:643–649. [PubMed] [Google Scholar]
  • 109.Kinney TR, Sleeper LA, Wang WC, et al. Silent cerebral infarcts in sickle cell anemia: A risk factor analysis. The Cooperative Study of Sickle Cell Disease. Pediatrics. 1999;103:640–645. doi: 10.1542/peds.103.3.640. [DOI] [PubMed] [Google Scholar]
  • 110.Li J, Kim K, Hahm E, et al. Neutrophil AKT2 regulates heterotypic cell-cell interactions during vascular inflammation. J Clin Invest. 2014;124:1483–1496. doi: 10.1172/JCI72305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zhang Y, Berka V, Song A, et al. Elevated sphingosine-1-phosphate promotes sickling and sickle cell disease progression. J Clin Invest. 2014;124:2750–2761. doi: 10.1172/JCI74604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Vercellotti GM, Belcher JD. Not simply misshapen red cells: Multimolecular and cellular events in sickle vaso-occlusion. J Clin Invest. 2014;124:1462–1465. doi: 10.1172/JCI75238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Haynes J, 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;103:3945–3950. doi: 10.1182/blood-2003-08-2969. [DOI] [PubMed] [Google Scholar]
  • 114.Macari ER, Lowrey CH. Induction of human fetal hemoglobin via the NRF2 antioxidant response signaling pathway. Blood. 2011;117:5987–5997. doi: 10.1182/blood-2010-10-314096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Promsote W, Makala L, Li B, et al. Monomethylfumarate induces γ-globin expression and fetal hemoglobin production in cultured human retinal pigment epithelial (RPE) and erythroid cells, and in intact retina. Invest Ophthalmol Vis Sci. 2014;55:5382–5393. doi: 10.1167/iovs.14-14179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Bereal-Williams C, Machado RF, McGowan V, et al. Atorvastatin reduces serum cholesterol and triglycerides with limited improvement in vascular function in adults with sickle cell anemia. Haematologica. 2012;97:1768–1770. doi: 10.3324/haematol.2011.054957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hoppe C, Kuypers F, Larkin S, et al. A pilot study of the short-term use of simvastatin in sickle cell disease: Effects on markers of vascular dysfunction. Br J Haematol. 2011;153:655–663. doi: 10.1111/j.1365-2141.2010.08480.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kalish BT, Matte A, Andolfo I, et al. Dietary ω-3 fatty acids protect against vasculopathy in a transgenic mouse model of sickle cell disease. Haematologica. 2015;100:870–880. doi: 10.3324/haematol.2015.124586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zennadi R. MEK inhibitors, novel anti-adhesive molecules, reduce sickle red blood cell adhesion in vitro and in vivo, and vasoocclusion in vivo. PLos One. 2014;9:11. doi: 10.1371/journal.pone.0110306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Pawloski JR, Hess DT, Stamler JS. Impaired vasodilation by red blood cells in sickle cell disease. Proc Natl Acad Sci USA. 2005;102:2531–2536. doi: 10.1073/pnas.0409876102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Reiter CD, Wang XD, Tanus-Santos JE, et al. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat Med. 2002;8:1383–1389. doi: 10.1038/nm1202-799. [DOI] [PubMed] [Google Scholar]
  • 122.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;350:886–895. doi: 10.1056/NEJMoa035477. [DOI] [PubMed] [Google Scholar]
  • 123.Gladwin MT, Vichinsky E. Pulmonary complications of sickle cell disease. N Engl J Med. 2008;359:2254–2265. doi: 10.1056/NEJMra0804411. [DOI] [PubMed] [Google Scholar]
  • 124.Parent F, Bachir D, Inamo J, et al. A hemodynamic study of pulmonary hypertension in sickle cell disease. N Engl J Med. 2011;365:44–53. doi: 10.1056/NEJMoa1005565. [DOI] [PubMed] [Google Scholar]
  • 125.Bunn HF, Nathan DG, Dover GJ, et al. Pulmonary hypertension and nitric oxide depletion in sickle cell disease. Blood. 2010;116:687–692. doi: 10.1182/blood-2010-02-268193. [DOI] [PubMed] [Google Scholar]
  • 126.Gladwin MT, Barst RJ, Castro OL, et al. Pulmonary hypertension and NO in sickle cell. Blood. 2010;116:852–854. doi: 10.1182/blood-2010-04-282095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Nathan DG. Guilt by association. Blood. 2011;118:3758–3759. doi: 10.1182/blood-2011-08-370338. [DOI] [PubMed] [Google Scholar]
  • 128.Weiner DL, Hibberd PL, Betit P, et al. Preliminary assessment of inhaled nitric oxide for acute vaso-occlusive crisis in pediatric patients with sickle cell disease. JAMA. 2003;289:1136–1142. doi: 10.1001/jama.289.9.1136. [DOI] [PubMed] [Google Scholar]
  • 129.Gladwin MT, Kato GJ, Weiner D, et al. Nitric oxide for inhalation in the acute treatment of sickle cell pain crisis a randomized controlled trial. JAMA. 2011;305:893–902. doi: 10.1001/jama.2011.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Atz AM, Wessel DL. Inhaled nitric oxide in sickle cell disease with acute chest syndrome. Anesthesiology. 1997;87:988–990. doi: 10.1097/00000542-199710000-00037. [DOI] [PubMed] [Google Scholar]
  • 131.Sullivan KJ, Goodwin SR, Evangelist J, et al. Nitric oxide successfully used to treat acute chest syndrome of sickle cell disease in a young adolescent. Crit Care Med. 1999;27:2563–2568. doi: 10.1097/00003246-199911000-00039. [DOI] [PubMed] [Google Scholar]
  • 132.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;97:1584–1589. doi: 10.1182/blood.v97.6.1584. [DOI] [PubMed] [Google Scholar]
  • 133.de Montalembert M, Aggoun Y, Niakate A, et al. Endothelial-dependent vasodilation is impaired in children with sickle cell disease. Haematologica. 2007;92:1709–1710. doi: 10.3324/haematol.11253. [DOI] [PubMed] [Google Scholar]
  • 134.Machado RF, Barst RJ, Yovetich NA, et al. Hospitalization for pain in patients with sickle cell disease treated with sildenafil for elevated TRV and low exercise capacity. Blood. 2011;118:855–864. doi: 10.1182/blood-2010-09-306167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Burnett AL, Anele UA, Trueheart IN, et al. Randomized controlled trial of sildenafil for preventing recurrent ischemic priapism in sickle cell disease. Am J Med. 2014;127:664–668. doi: 10.1016/j.amjmed.2014.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Barst RJ, Mubarak KK, Machado RF, et al. Exercise capacity and haemodynamics in patients with sickle cell disease with pulmonary hypertension treated with bosentan: Results of the ASSET studies. Br J Haematol. 2010;149:426–435. doi: 10.1111/j.1365-2141.2010.08097.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Morris CR. Alterations of the arginine metabolome in sickle cell disease: A growing rationale for arginine therapy. Hematol/Oncol Clin North Am. 2014;28:301–321. doi: 10.1016/j.hoc.2013.11.008. [DOI] [PubMed] [Google Scholar]
  • 138.Morris CR, Kuypers FA, Lavrisha L, et al. A randomized, placebo-controlled trial of arginine therapy for the treatment of children with sickle cell disease hospitalized with vaso-occlusive pain episodes. Haematologica. 2013;98:1375–1382. doi: 10.3324/haematol.2013.086637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.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;121:1276–1284. doi: 10.1182/blood-2012-11-451229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Chintagari NR, Nguyen J, Belcher JD, et al. Haptoglobin attenuates hemoglobin-induced heme oxygenase-1 in renal proximal tubule cells and kidneys of a mouse model of sickle cell disease. Blood Cells Mol Dis. 2015;54:302–306. doi: 10.1016/j.bcmd.2014.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Boretti FS, Baek JH, Palmer AF, et al. Modeling hemoglobin and hemoglobin:haptoglobin complex clearance in a non-rodent species–pharmacokinetic and therapeutic implications. Front Physiol. 2014;5:385. doi: 10.3389/fphys.2014.00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Gonzalez P, Hackney AC, Jones S, et al. A phase I/II study of polymerized bovine hemoglobin in adult patients with sickle cell disease not in crisis at the time of study. J Invest Med. 1997;45:258–264. [PubMed] [Google Scholar]
  • 143.Natanson C, Kern SJ, Lurie P, et al. Cell-free hemoglobin-based blood substitutes and risk of myocardial infarction and death: A meta-analysis. JAMA. 2008;299:2304–2312. doi: 10.1001/jama.299.19.jrv80007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. [Last accessed on July 28, 2015]; http://www.sec.gov/Archives/edgar/data/1420031/000104746915003012/a2223903z10-k.htm.
  • 145.Romero Z, Urbinati F, Geiger S, et al. β-globin gene transfer to human bone marrow for sickle cell disease. J Clin Invest. 2013;123:3317–3330. doi: 10.1172/JCI67930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Urbinati F, Hargrove PW, Geiger S, et al. Potentially therapeutic levels of anti-sickling globin gene expression following lentivirus-mediated gene transfer in sickle cell disease bone marrow CD34+ cells. Exp Hematol. 2015;43:346–351. doi: 10.1016/j.exphem.2015.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Hoban MD, Cost GJ, Mendel MC, et al. Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. Blood. 2015;125:2597–2604. doi: 10.1182/blood-2014-12-615948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Ledford H. CRISPR, the disruptor. Nature. 2015;522:20–24. doi: 10.1038/522020a. [DOI] [PubMed] [Google Scholar]
  • 149.Dedeken L, Le PQ, Azzi N, et al. Haematopoietic stem cell transplantation for severe sickle cell disease in childhood: A single centre experience of 50 patients. Br J Haematol. 2014;165:402–408. doi: 10.1111/bjh.12737. [DOI] [PubMed] [Google Scholar]
  • 150.Bernaudin F, Socie G, Kuentz M, et al. Long-term results of related myeloablative stem-cell transplantation to cure sickle cell disease. Blood. 2007;110:2749–2756. doi: 10.1182/blood-2007-03-079665. [DOI] [PubMed] [Google Scholar]
  • 151.Locatelli F, Kabbara N, Ruggeri A, et al. Outcome of patients with hemoglobinopathies given either cord blood or bone marrow transplantation from an HLA-identical sibling. Blood. 2013;122:1072–1078. doi: 10.1182/blood-2013-03-489112. [DOI] [PubMed] [Google Scholar]
  • 152.Lê PQ, Gulbis Dedeken L, Dupont S, et al. Survival among children and adults with sickle cell disease in Belgium: Benefit from hydroxyurea treatment. Pediatr Blood Cancer. doi: 10.1002/pbc.25608. in press. [DOI] [PubMed] [Google Scholar]
  • 153.Hsieh MM, Fitzhugh CD, Weitzel RP, et al. Nonmyeloablative HLA-matched sibling allogeneic hematopoietic stem cell transplantation for severe sickle cell phenotype. JAMA. 2014;312:48–56. doi: 10.1001/jama.2014.7192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Horan JT, Haight A, Dioguardi JL, et al. Using fludarabine to reduce exposure to alkylating agents in children with sickle cell disease receiving busulfan, cyclophosphamide, and antithymocyte globulin transplant conditioning: Results of a dose de-escalation trial. Biol Blood Marrow Transplant. 2015;21:900–905. doi: 10.1016/j.bbmt.2015.01.015. [DOI] [PubMed] [Google Scholar]
  • 155.Ballas SK, Gupta K, Adams-Graves P. Sickle cell pain: A critical reappraisal. Blood. 2012;120:3647–3656. doi: 10.1182/blood-2012-04-383430. [DOI] [PubMed] [Google Scholar]
  • 156.Poggiali E, Cassinerio E, Zanaboni L, et al. An update on iron chelation therapy. Blood Transf. 2012;10:411–422. doi: 10.2450/2012.0008-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Vichinsky E, Torres M, Minniti CP, et al. Efficacy and safety of deferasirox compared with deferoxamine in sickle cell disease: Two-year results including pharmacokinetics and concomitant hydroxyurea. Am J Hematol. 2013;88:1068–1073. doi: 10.1002/ajh.23569. [DOI] [PubMed] [Google Scholar]
  • 158.Mahadeo KM, Oyeku S, Taragin B, et al. Increased prevalence of osteonecrosis of the femoral head in children and adolescents with sickle-cell disease. Am J Hematol. 2011;86:806–808. doi: 10.1002/ajh.22103. [DOI] [PubMed] [Google Scholar]
  • 159.Poignard A, Flouzat-Lachaniette CH, Amzallag J, et al. The natural progression of symptomatic humeral head osteonecrosis in adults with sickle cell disease. J Bone Joint Surg Am. 2012;94:156–162. doi: 10.2106/JBJS.J.00919. [DOI] [PubMed] [Google Scholar]
  • 160.Issa K, Naziri Q, Maheshwari AV, et al. Excellent results and minimal complications of total hip arthroplasty in sickle cell hemoglobinopathy at mid-term follow-up using cementless prosthetic components. J Arthroplast. 2013;28:1693–1698. doi: 10.1016/j.arth.2013.03.017. [DOI] [PubMed] [Google Scholar]
  • 161.Nath KA, Hebbel RP. Sickle cell disease: Renal manifestations and mechanisms. Nat Rev Nephrol. 2015;11:161–171. doi: 10.1038/nrneph.2015.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Gosmanova EO, Zaidi S, Wan JY, et al. Prevalence and progression of chronic kidney disease in adult patients with sickle cell disease. J Invest Med. 2014;62:804–807. doi: 10.1097/01.JIM.0000446836.75352.72. [DOI] [PubMed] [Google Scholar]
  • 163.Bodas P, Huang A, O’Riordan MA, et al. The prevalence of hypertension and abnormal kidney function in children with sickle cell disease -a cross sectional review. BMC Nephrol. 2013;14:6. doi: 10.1186/1471-2369-14-237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Laurin LP, Nachman PH, Desai PC, et al. Hydroxyurea is associated with lower prevalence of albuminuria in adults with sickle cell disease. Nephrol Dial Transplant. 2014;29:1211–1218. doi: 10.1093/ndt/gft295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Lionnet F, Hammoudi N, Stojanovic KS, et al. Hemoglobin sickle cell disease complications: A clinical study of 179 cases. Haematol-Hematol J. 2012;97:1136–1141. doi: 10.3324/haematol.2011.055202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Wanek J, Gaynes B, Lim JI, et al. Human bulbar conjunctival hemodynamics in hemoglobin SS and SC disease. Am J Hematol. 2013;88:661–664. doi: 10.1002/ajh.23475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Nero AC, Akuete K, Reeves SL, et al. Pneumococcal vaccination rates in children with sickle cell disease. J Public Health Manag Pract. 2014;20:587–590. doi: 10.1097/PHH.0000000000000034. [DOI] [PubMed] [Google Scholar]
  • 168.Payne AB, Link-Gelles R, Azonobi I, et al. Invasive pneumococcal disease among children with and without sickle cell disease in the United States, 1998 to 2009. Pediatr Infect Dis J. 2013;32:1308–1312. doi: 10.1097/INF.0b013e3182a11808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Beverung LM, Brousseau D, Hoffmann RG, et al. Ambulatory quality indicators to prevent infection in sickle cell disease. Am J Hematol. 2014;89:256–260. doi: 10.1002/ajh.23627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Soderblom EJ, Thompson JW, Schwartz EA, et al. Proteomic analysis of ERK1/2-mediated human sickle red blood cell membrane protein phophorylation. Clin Proteom. 2013;10:1. doi: 10.1186/1559-0275-10-1. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES