Sickle cell disease (SCD) places a heavy burden on a global and increasing population predominantly resident in resource-poor and developing countries. Progress continues to be made in preventing childhood mortality, and increasing numbers of chronically ill adults with disease are requiring care for disease sequelae. Curative therapies for SCD are therefore attractive to physicians and investigators focused on SCD. Gene therapies are being developed, and several are now in various stages of early-phase human clinical trials. However, we must also pursue avenues through which we can do the most good for the most people alive today. Such efforts include improving our understanding of disease mechanisms and which disease sequelae most strongly affect survival and interfere with quality of life. The pathways leading to disease sequelae are multiple, complex, and highly interactive. Four drugs have now been approved by the US Food and Drug Administration for SCD; however, each has a distinct mechanism and a measurable but limited effect on the many clinical sequelae of SCD. We therefore need to learn how to approach multi-agent therapy for SCD. The order of addition of each agent to treat a specific patient will need to be guided by response to previous therapy, risk factors identified for specific disease outcomes, and clinical studies to determine more comprehensively how the 4 currently approved drugs might interact and produce (or not) additive effects. Moreover, this will have to be accomplished with defined end points in mind, according to which pose the greatest threats to quality of life as well as survival.

Sickle cell disease (SCD) places a heavy burden on an increasingly widespread population throughout the world. Although only ∼100 000 to 120 000 of the ∼330 million people in the United States (0.036%) live with SCD,1  ∼20 million people are affected by SCD worldwide. Globally, ∼312 000 children are born with SCD each year.2  Most of the people affected by SCD live in developing countries with scarce resources to devote to health care. Thus, although survival is improving in India and in African countries, and adults with SCD are no longer highly unusual in those settings, the average survival with SCD still means that death during childhood is far more likely than survival to adulthood, with mortality under the age of 5 years estimated to be 50% to 90% in low-income countries.3  In addition, as we have learned in more resource-rich countries, survival to adulthood results in a high burden of disease-related complications during adult life, with multiple types of end-organ damage causing both shortened survival as well as substantially impaired quality of life. In addition, although death from SCD during childhood is relatively rare (<4%) in the United States,4  the nation spends approximately $1 billion annually on care for individuals with SCD.5 

Curative therapies for SCD are therefore attractive to physicians and investigators focused on SCD, although such therapy offers both potential benefits and risks to patients. The first curative therapy to arrive on the horizon was hematopoietic stem cell transplant (HSCT). However, it became clear early on that this procedure was extremely challenging when performed in patients with SCD. Initial success was observed in young children, whereas success in older children and adults came at the price of a great deal of experimentation and high mortality rates during the early years of this effort. Although we now can perform HSCT for both children and adults with increasing success,6,7  HSCT has thus far reached only ∼2000 individuals worldwide, with overall survival of ∼95% and an average age at HSCT of 10 years.8  Thus, under the best circumstances, for the next few decades, HSCT will likely remain available to only a minority of patients due to donor availability and high resource requirements, although progress is being made in utilization of alternative donors, such as haploidentical family members.9 

Meanwhile, gene therapies are being developed, and several are now in various stages of early-phase human clinical trials. Countries with robust medical research enterprises, including the United States, are increasingly focusing on gene therapy for hemoglobinopathies.10-14 

Generally, gene therapy may take a variety of approaches, including: (1) addition of a helpful gene; (2) gene knockdown (eg, Bcl11A) to improve the phenotype; (3) direct globin gene editing to “correct” the mutation present (eg, changing a hemoglobin S [HbS]–encoding gene to one encoding hemoglobin A); and (4) gene editing of globin regulatory elements, to at least partially reverse the normal hemoglobin switching from fetal to adult hemoglobin. The gene addition approach may entail adding a gene encoding γ globin, a β/γ hybrid, an anti–sickling β-globin [eg, βA(T87Q)], or simply β-globin.15  Nonetheless, although gene therapy is improving relatively fast, there is still a long way to go. Continuous iterative improvement remains needed in every aspect of gene therapy development, including: (1) quality of viral delivery systems; (2) quality and quantity of hematopoietic stem cells harvested; (3) optimization of the gene modification system in hematopoietic stem cells; (4) choice of recombination pathway (homologous vs non-homologous); (5) identification of the best gene targets; (6) cell manufacturing; (7) preparation regimens to allow the bone marrow to receive genetically modified cells with minimized toxicity; (8) problems related to off-target effects; (9) optimization of preclinical models for testing of developing gene therapy strategies; and (10) parameters that should be used to define cure.

Given the current infrastructure present where most SCD patients are located, and to reach a significant fraction of people living with SCD today, gene therapy will also need to involve minimal bone marrow ablation and in vivo transduction (ie, Star Trek medicine). As in the Starship Enterprise sickbay, the patient would ideally be successfully treated by one injection of a healing factor and require no further care. Alternatively, if we will not be able to offer curative therapies to the vast majority of people currently living with SCD during their lifetimes, we must provide those patients alive today with alternative therapies to improve survival and quality of life.

Realizing the difficulties confronting gene therapy at this time, pharmaceutical companies and investigators have also been trying to develop pharmacologic methods to affect the genes controlling hemoglobin switching and thus increase fetal hemoglobin (HbF) expression. At least 2 loci independently and strongly affect HbF (γ/δ globin) suppression: Bcl11A and LRF/ZBTB7A. Potentially therapeutic small molecules have reportedly been identified, and early-phase clinical trials are in the planning stages to determine both their safety and whether they can indeed reactivate HbF expression in vivo.16  Other molecules may bind to and affect the globin control region, facilitating expression of γ globin.17  Although not curative per se, such therapeutic agents might be able to accomplish what is targeted by many gene therapies; that is, using increased HbF expression to improve cell biology and forestall the damage caused by sickle red cell sickling, hemolysis, and abnormal circulatory behavior.

Of course, the root of the problem in SCD is the sickle red blood cell (SS RBC), a red blood cell containing predominantly HbS or HbS and another abnormal hemoglobin that participates in hemoglobin polymerization in the setting of hemoglobin deoxygenation. HbS is an inherently unstable hemoglobin in the circulation; it undergoes deoxygenation under higher oxygen tension than normal hemoglobin, forms polymers when deoxygenated, and ultimately leads to deformation of red blood cells (sickling) and hemolysis. However, exacerbating this process is a panoply of additional effects arising from perturbation of the RBCs by HbS (Figure 1).18  These effects include but are not limited to the following pathophysiologic processes, as partly illustrated in Figure 1:

  • Abnormal cation content and dehydration,19  leading to increased cellular viscosity;

  • Surface extracellular phosphatidylserine exposure, leading to activation of coagulation pathways20-22 ;

  • Metabolic abnormalities, including adenosine triphosphate and glutathione deficiency and high 2,3-diphosphoglycerate23-25 ;

  • Oxidative damage to membranes, accompanied by membrane instability and abnormal deformability and rheology26,27 ;

  • Abnormally high intracellular signaling activity, likely due to young RBC age and altered gene expression in stress erythropoiesis27-32 ;

  • Abnormally increased adhesive properties33-35 ;

  • Abnormal cell–cell signaling, leading to activation of leukocytes,36  endothelial cells,37  and probably platelets; and

  • Deficient expression of antioxidant compounds due to alteration in gene regulation and metabolism.32 

Figure 1.

Pathophysiologic pathways and drug interventions in SCD. The pathologic RBCs containing HbS and microparticles derived from them give rise to a multiplicity of highly interactive pathways that contribute to hemolysis and organ damage. Various FDA-approved drugs as well as other classes of compounds may serve to reduce one or more of these processes and thus ameliorate the symptoms of the disease. O2, oxygen.

Figure 1.

Pathophysiologic pathways and drug interventions in SCD. The pathologic RBCs containing HbS and microparticles derived from them give rise to a multiplicity of highly interactive pathways that contribute to hemolysis and organ damage. Various FDA-approved drugs as well as other classes of compounds may serve to reduce one or more of these processes and thus ameliorate the symptoms of the disease. O2, oxygen.

Close modal

Thus, the SS RBC stands at the center of the vicious cycle of SCD that gives rise to hemolysis and anemia; vaso-occlusion with attendant pain, acute organ damage, and vasculopathy; and chronic tissue damage and vasculopathy leading to irreversible end-organ dysfunction. Importantly, the pathways leading to these downstream sequelae are multiple, complex, and highly interactive.

Given our limited ability to affect the genetic root cause of SCD at this time, how can we do the most good for the most people alive today? To improve survival, we first must answer a critical question: Which sequelae are most associated with mortality? The answers, however, likely depend on geography and patient age. Although childhood mortality is low in the United States and countries with similar resources, it is common in Africa and India. Causes of disability also vary by both age and geography.

SCD results in multiple varieties of acute and chronic end-organ damage. These comprise cardiovascular and cardiopulmonary effects, including congestive heart failure and pulmonary hypertension; pulmonary effects, including acute chest syndrome; musculoskeletal effects, including dactylitis and avascular necrosis; neurologic effects, including both hemorrhagic and infarctive strokes, seizure disorders, and cognitive impairment; renal effects, including hyposthenuria, proteinuria, and end-stage renal disease; splenic sequelae, including splenic infarcts, splenic sequestration, and immunologic deficiencies associated with asplenia; hepatobiliary effects, including cholelithiasis and hepatic sequestration; and priapism. Among these sequelae, the ones most associated with mortality are acute chest syndrome,38  central nervous system effects (both strokes and presence of a seizure disorder, presumably reflecting silent infarcts),39-41  sickle cell nephropathy, as identified by the presence of macroalbuminuria,39,42  and pulmonary hypertension, as defined by elevated tricuspid regurgitant jet velocity. In addition, however, the frequency of vaso-occlusive (VOC) episodes is a predictor of mortality.43,44  Thus, therapeutic maneuvers that address these sequelae of SCD should extend survival. However, at the same time, we must remember that patients do not only want to live longer, they want to live better lives.

Four drugs have now been approved by the US Food and Drug Administration (FDA) for treatment of SCD. In reviewing the benefits of their use (Figure 2), however, we see that there is a long way to go to achieve both symptom relief and improved survival. Hydroxyurea seems to ameliorate many aspects of SCD, including anemia, VOC frequency, and acute chest syndrome frequency, while also improving overall survival. However, hydroxyurea is only partially effective in preventing stroke and may not have a major effect on the development of nephropathy and pulmonary hypertension, although definitive evidence is lacking. The 3 other more recently approved drugs for SCD are all individually somewhat less broad in their effects. l-glutamine decreased VOC frequency by ∼25%, while also reducing the frequency of acute chest syndrome significantly and hospitalization frequency by about one-third.45  However, the study leading to FDA approval failed to show any change in hematologic parameters such as anemia, and there was also no effect documented for well-being and quality of life measures. In addition, the mechanism whereby l-glutamine exerts its effects remains uncharacterized. Voxelotor, which binds to and shifts the oxygen dissociation curve of HbS and thereby inhibits deoxygenation and sickling, seems to substantially reduce hemolysis and ameliorate anemia in most patients with SCD.46,47  However, the study leading to its approval by the FDA for SCD treatment did not show any salutatory effects on VOC or acute chest syndrome frequency,47  nor is there evidence that the drug might be effective prophylaxis for stroke, nephropathy, or pulmonary hypertension. Crizanlizumab, which is a humanized monoclonal antibody directed against P-selectin (one of the mediators of both erythrocyte and leukocyte adhesion in animal models of vaso-occlusion), was effective in reducing VOC frequency.48,49  However, it did not affect hematologic parameters or quality of life.

Figure 2.

Effects of FDA-approved drugs for SCD. Four currently approved drugs for SCD each have distinct mechanisms and incompletely overlapping benefits. ↓, evidence for decrease in degree or frequency; →, evidence for no effect; QoL, quality of life; Fatigue and QoL ↓, evidence that fatigue is decreased and QoL is improved.

Figure 2.

Effects of FDA-approved drugs for SCD. Four currently approved drugs for SCD each have distinct mechanisms and incompletely overlapping benefits. ↓, evidence for decrease in degree or frequency; →, evidence for no effect; QoL, quality of life; Fatigue and QoL ↓, evidence that fatigue is decreased and QoL is improved.

Close modal

Furthermore, none of these 3 newer agents has been comprehensively studied in multiple age groups for an extended period of time. l-glutamine was studied in both children aged ≥5 years and adults, but the follow-up period was <1 year. Voxelotor was studied at 2 dose levels in patients aged >12 years for up to 72 weeks, although most end points were reported only after 24 weeks of treatment. Crizanlizumab was likewise administered over 52 weeks only to patients aged ≥16 years. In summary, therefore, all 3 drugs together have been studied in <500 persons receiving study drug (rather than placebo), and no long-term follow-up, particularly regarding end-organ damage and mortality, is available for any of them.

Thus, to achieve further therapeutic progress, we need to continue to develop a better understanding of mechanisms leading to organ dysfunction associated with poor quality of life and shortened survival, devise an evidence-based assessment system of risk for acute and chronic organ dysfunction, and adopt multi-agent therapy based on a better understanding of risk factors and disease mechanisms. Although vaso-occlusion occurs throughout the body, processes contributing to specific types of organ damage may vary. We need to understand the organ-specific characteristics of these events so that we can identify druggable targets and develop drugs and treatment modalities addressing those targets. With that point of view, and considering the pathophysiologic pathways shown in Figure 1, we might find additional drugs, either already FDA-approved drugs, such as anticoagulants, or drugs still in development, such as pyruvate kinase activators, that can be beneficial in combination with hydroxyurea, either with or without the other drugs shown in Figure 2. However, given the limitations of studies to date, insufficient data are likely to make difficult both the choices of multi-drug regimens as well as how to judge their efficacy and safety.

Acute events continue to bring high risk of morbidity and mortality in SCD. Among those seen with high frequency, acute chest syndrome remains the most common acute event leading to mortality in both children and adults.38,50 

Acute chest syndrome typically presents with hypoxemia and pulmonary infiltrates consistent with pneumonia on chest radiography. This syndrome can have a rapid downhill course, which may result from the effect hypoxia has on the pulmonary endothelium, including upregulation of adhesion molecules and downregulation of protective mechanisms. Recently, in its approval of l-glutamine, the FDA cited not only its ability to reduce the frequency of VOC but also its efficacy in reducing the incidence of acute chest syndrome.45  Nonetheless, we cannot currently offer our patients pharmacologic therapies for acute chest syndrome. At this time, treatment usually consists of oxygen supplementation (and ventilatory support when needed), antibiotics (despite the paucity of evidence that acute chest syndrome often involves bacterial infection38 ), and blood transfusion. The latter, although never tested in a randomized clinical trial, seems to be the most effective treatment we have; simple transfusion is usually used in mild cases, and RBC exchange is used in severe cases, as well as in milder cases38 in patients with relatively higher baseline hemoglobin levels, such as with HbSC.

Over the past several years, investigators have developed a better understanding of acute chest syndrome that may lead to real therapeutic advances. Etiologic factors are not identifiable in nearly 50% of cases,38  although intravascular sickling,51  fat emboli, hypoxia,52  microvascular in situ thrombosis,52  infection,53  and acute painful VOC episodes may precede acute chest syndrome. Ghosh et al54  created an animal model of acute chest syndrome that relies on hemolysis as the precipitating factor. They note that available data support the idea that, whatever the cause, acute chest syndrome is most often preceded by an increase in hemolysis and thus in free heme.50,52,55  Research with this animal model has suggested both new pathophysiologic pathways as well as possible new treatment modalities. The authors found that pharmacologic inhibition of Toll-like receptor 4 (TLR4), as well as infusion of hemopexin before hemin infusion, protected sickle mice from developing acute chest syndrome.54 

In somewhat similar studies, Belcher et al56  showed that infusion of hemoglobin or heme triggered vaso-occlusion in sickle mice, and that this effect was blocked by infusion of haptoglobin or the heme-binding protein hemopexin. They also found that blockade of TLR4 was protective, and that TLR4 knockout mice transplanted with sickle bone marrow were resistant to heme-induced vaso-occlusion. These groups have also shown that agents which interfere with cell adhesion or that increase antioxidant stress capacity also protect against the deleterious consequences of hemin infusion.56-58  These studies are examples of how modeling of specific complications of SCD may lead to promising therapeutic investigations. Indeed, l-glutamine, which is believed to improve antioxidant capacity, was approved by the FDA in part due to the reduction in acute chest syndrome seen with the drug.45,59  Animal models suggest that improvement in antioxidant response helps prevent heme oxygenase decline and thereby facilitates the breakdown of heme and reduction of its downstream toxic effects.58 

Clinically apparent chronic end-organ damage has a strong negative effect on overall survival in SCD, especially among adults. Several recent studies of survival in US SCD cohorts have documented that both the overall number of organs affected as well as specific types of end-organ damage strongly affect life expectancy. Chaturvedi et al60  showed that the presence of more than one organ affected by SCD greatly reduced survival from 14.0 to 7.8 years in adults, even after adjusting for age at enrollment, sex, sickle cell genotype (sickle cell anemia vs other), and hydroxyurea therapy. Elmariah et al39  showed that a history of central nervous system events, including stroke and seizure, as well as elevated tricuspid regurgitant jet velocity (emblematic of pulmonary hypertension), and sickle cell nephropathy have especially negative effects on survival; requirement for hospital admission for VOC in the past year, as well as several other clinical characteristics, are also associated with a higher risk of mortality. Four or more VOC episodes per year are especially strongly correlated to mortality.

Sickle cell nephropathy and pulmonary hypertension, at least as indicated by high tricuspid regurgitant jet velocity measured by echocardiogram, are 2 types of chronic end-organ damage that are highly linked to accelerated mortality. Nephropathy is highly associated with premature mortality (hazard ratio, 1.86), and survival with chronic kidney disease is severely limited.39  SCD affects both glomerular and tubular function of the kidney. Hyposthenuria, the inability to concentrate urine to >450 mOsm/kg with water deprivation, is seen in infancy and persists throughout life. Hyperfiltration, often defined as a measured glomerular filtration rate (GFR) >110 mL/min/1.73 m2, is present in >50% of infants from 9 months to 1 year of age42  and increases during childhood and into young adulthood. Nearly all adults age 18 to 30 years also exhibit high creatinine-based estimated GFR (130 mL/min/1.73 m2).61  Hyperfiltration also seems to result in glomerular injury, and hyperfiltration (estimated GFR >130-140 mL/min/1.73 m2) is associated with the development of microalbuminuria. Progression to macroalbuminuria affects nearly 40% of adults with HbSS by age 40 years.42  Tubular functional defects, in addition to hyposthenuria, have recently been shown to include impaired urinary acidification and distal tubular acidosis.62  Macroalbuminuria is a common finding (26%-28%) in SCD and is a known risk factor for decline in renal function. Current SCD guidelines suggest that any patient with macroalbuminuria or modest elevations in creatinine (>1.0 mg/dL in adults) should be considered as having significant renal disease and receive treatment with an angiotensin-converting enzyme inhibitor or angiotensin receptor blocker therapy.63  However, these therapies are used in analogy with other causes of proteinuria, although their true efficacy in SCD is uncertain. To study sickle cell nephropathy and potential therapeutic strategies, we first have to improve our understanding of risk factors and rate of progression. A growing body of evidence suggests that the rate of annual GFR decline is predictive of progression to end-stage renal disease. In a retrospective study, Xu et al64  asked: Can we identify SCD patients with a high risk of rapid GFR decline, to target therapies to this population and optimize the power of clinical studies to detect efficacy? Their study was able to identify 7 variables that increased the risk of rapid renal functional decline, including hemoglobin genotype (eg, HbSS/Sβ0 vs HbSC/Sβ+), presence of proteinuria, higher platelet count, higher reticulocyte count, higher systolic blood pressure, and lower body mass index. These and other factors, including the ApoL1 genetic variants thoroughly demonstrated to be risk factors for sickle nephropathy,65,66  must now be verified prospectively, so that future therapeutic trials can most efficiently identify valuable pharmacologic approaches by studying the proportion of patients at highest risk for significant renal disease.

Although curative approaches such as gene therapy may eventually be a universally applicable way to cure SCD, the advances needed to make gene therapy simpler and transportable to resource-poor countries are likely many years in the future and possibly beyond the lifespans of the majority of those already living with the disease. Hydroxyurea, which inhibits HbS polymerization by inducing HbF expression and has multiple, potential other modes of action as well, has been very effective in altering the course of SCD for many patients63,67-71 ; however, its multiple effects also pose a spectrum of challenges for patients and caregivers (Figure 3). Despite higher HbF levels, even “responders” may still experience debilitating episodes of vaso-occlusion, strokes, organ dysfunction, and pain. Hydroxyurea is not recommended for use in female or male patients while they are planning to conceive children and may also reduce ovarian reserve. Hydroxyurea may also cause long-term damage to hematopoietic cells, reducing stem cell number and in some cases causing myelodysplasia.72  Physicians in most cases have managed hydroxyurea by titrating dosing to maximum tolerated dose, followed by frequent blood count checks to ensure safety. Only recently have studies shown that a standard dose can be both safe and beneficial.70,73-75 

Figure 3.

Hydroxyurea and multi-agent therapy. Although hydroxyurea (HU) has been used for decades in a variety of diseases, and its overall safety is well documented, there are still some unresolved questions regarding its long-term use. Guidelines continue to advise against HU when male or female patients plan to conceive a child, and questions remain about the effect of HU on spermatogenesis77,78  and ovarian reserve. In industrialized countries, management of HU treatment most often involves titration to maximum tolerated dose (MTD), but recent studies in Africa have documented the feasibility and benefit of fixed dosing without such resource-intensive monitoring.70,73,75  Concerns also persist regarding the possibility that long-term HU therapy reduces the number and viability of pluripotent stem cells and may predispose to myelodysplasia.72  The efficacy of HU in preventing organ damage, such as cardiac dysfunction, also remains uncertain. Now, to improve overall SCD therapy, we need to learn how to prescribe HU in combination with other, differently targeted therapies, such as those already FDA approved. Hopefully, additional therapies with additive or different therapeutic effects will be added to this list in the future. ACS, acute chest syndrome; MDS, myelodysplastic syndrome.

Figure 3.

Hydroxyurea and multi-agent therapy. Although hydroxyurea (HU) has been used for decades in a variety of diseases, and its overall safety is well documented, there are still some unresolved questions regarding its long-term use. Guidelines continue to advise against HU when male or female patients plan to conceive a child, and questions remain about the effect of HU on spermatogenesis77,78  and ovarian reserve. In industrialized countries, management of HU treatment most often involves titration to maximum tolerated dose (MTD), but recent studies in Africa have documented the feasibility and benefit of fixed dosing without such resource-intensive monitoring.70,73,75  Concerns also persist regarding the possibility that long-term HU therapy reduces the number and viability of pluripotent stem cells and may predispose to myelodysplasia.72  The efficacy of HU in preventing organ damage, such as cardiac dysfunction, also remains uncertain. Now, to improve overall SCD therapy, we need to learn how to prescribe HU in combination with other, differently targeted therapies, such as those already FDA approved. Hopefully, additional therapies with additive or different therapeutic effects will be added to this list in the future. ACS, acute chest syndrome; MDS, myelodysplastic syndrome.

Close modal

Three other drugs have recently been approved for SCD treatment, although each has only limited effects currently documented by data (Figure 2). Each of the newly approved drugs seems effective when used alone or concurrently with hydroxyurea. Moreover, each of these new drugs targets different pathophysiologic processes, and more differently targeted drugs are in the pipeline.

In this new world, however, we have yet to learn how to approach multi-agent therapy for SCD. The order of addition of each agent to treat a specific patient will need to be guided by response to previous therapy, risk factors identified for specific disease outcomes, and clinical studies to determine more thoroughly how the 4 currently approved drugs “mix and match.” This goal will have to be accomplished with an improved understanding of both disease pathophysiology and drug mechanisms of action, as well as defined end points in mind. Depending in part on the drugs being studied, appropriate end points may need to include HbF content; hemoglobin p50; markers of oxidative stress; inflammatory markers; white blood cell count, RBC, platelet, and reticulocyte counts; markers of hemolysis, including such measures as plasma free hemoglobin, and haptoglobin and hemopexin levels; frequency and duration of pain episodes; presence of end-organ damage and risk assessment; and quality-of-life measures. To date, however, although a variety of biomarkers predict higher mortality, only transcranial Doppler measurements have correlated with both prediction of risk as well as a therapeutic outcome (prevention of stroke).76 

The health of millions of patients living with SCD today depends on near-term success in developing and delivering targeted and multi-agent therapeutic approaches to the sequelae of having RBCs with predominantly HbS. In the long run, and perhaps not during the lives of most patients with SCD alive today, gene therapy will become an easily accomplished and economically feasible cure. Although Judah Folkman, a true pioneer in medicine, cited E. L. Doctorow, who had originally referred to the process of writing, Folkman found it equally true of research. He said it is “like driving at night. You cannot see beyond the headlights, but you can make the whole trip that way.”79  We may not now be able to see exactly where we will end up, but we can nevertheless get there and save lives along the way.

M.J.T. acknowledges research support from the Doris Duke Charitable Foundation and from the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (DK110104).

Contribution: This manuscript was conceived and solely written by M.J.T.

Conflict-of-interest disclosure: M.J.T. has received research funds in support of basic research from Forma Therapeutics and CSL Behring; has conducted clinical research sponsored by Pfizer Inc. and Forma Therapeutics; and serves on the Executive Committee of the Cure Sickle Cell Initiative of the National Institutes of Health and on a Data Monitoring Committee for a study of crizanlizumab sponsored by Novartis.

Correspondence: Marilyn J. Telen, School of Medicine, Duke University, Box 2615, DUMC MSRB1, Research Dr, Durham, NC 27710; e-mail: marilyn.telen@duke.edu.

1.
Hassell
KL
.
Population estimates of sickle cell disease in the U.S
.
Am J Prev Med
.
2010
;
38
(
suppl 4
):
S512
-
S521
.
2.
Piel
FB
,
Patil
AP
,
Howes
RE
, et al
.
Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model-based map and population estimates
.
Lancet
.
2013
;
381
(
9861
):
142
-
151
.
3.
Uyoga
S
,
Macharia
AW
,
Mochamah
G
, et al
.
The epidemiology of sickle cell disease in children recruited in infancy in Kilifi, Kenya: a prospective cohort study
.
Lancet Glob Health
.
2019
;
7
(
10
):
e1458
-
e1466
.
4.
Quinn
CT
,
Rogers
ZR
,
Buchanan
GR
.
Survival of children with sickle cell disease
.
Blood
.
2004
;
103
(
11
):
4023
-
4027
.
5.
Kauf
TL
,
Coates
TD
,
Huazhi
L
,
Mody-Patel
N
,
Hartzema
AG
.
The cost of health care for children and adults with sickle cell disease
.
Am J Hematol
.
2009
;
84
(
6
):
323
-
327
.
6.
Bernaudin
F
,
Socie
G
,
Kuentz
M
, et al;
SFGM-TC
.
Long-term results of related myeloablative stem-cell transplantation to cure sickle cell disease
.
Blood
.
2007
;
110
(
7
):
2749
-
2756
.
7.
Tisdale
JF
,
Thein
SL
,
Eaton
WA
.
Treating sickle cell anemia
.
Science
.
2020
;
367
(
6483
):
1198
-
1199
.
8.
Walters
MC
,
De Castro
LM
,
Sullivan
KM
, et al
.
Indications and results of HLA-identical sibling hematopoietic cell transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2016
;
22
(
2
):
207
-
211
.
9.
Eapen
M
,
Brazauskas
R
,
Walters
MC
, et al
.
Effect of donor type and conditioning regimen intensity on allogeneic transplantation outcomes in patients with sickle cell disease: a retrospective multicentre, cohort study
.
Lancet Haematol
.
2019
;
6
(
11
):
e585
-
e596
.
10.
Brendel
C
,
Williams
DA
.
Current and future gene therapies for hemoglobinopathies
.
Curr Opin Hematol
.
2020
;
27
(
3
):
149
-
154
.
11.
Curtis
SA
,
Shah
NC
.
Gene therapy in sickle cell disease: possible utility and impact
.
Cleve Clin J Med
.
2020
;
87
(
1
):
28
-
29
.
12.
Leonard
A
,
Tisdale
J
,
Abraham
A
.
Curative options for sickle cell disease: haploidentical stem cell transplantation or gene therapy?
Br J Haematol
.
2020
;
189
(
3
):
408
-
423
.
13.
Motta
I
,
Ghiaccio
V
,
Cosentino
A
,
Breda
L
.
Curing hemoglobinopathies: challenges and advances of conventional and new gene therapy approaches
.
Mediterr J Hematol Infect Dis
.
2019
;
11
(
1
):
e2019067
.
14.
Tachere
RO
,
Umunna
AO
.
Gene therapy renews hope to lower the global rural sickle cell disease burden
.
Rural Remote Health
.
2017
;
17
(
3
):
4291
.
15.
Demirci
S
,
Uchida
N
,
Tisdale
JF
.
Gene therapy for sickle cell disease: an update
.
Cytotherapy
.
2018
;
20
(
7
):
899
-
910
.
16.
Vinjamur
DS
,
Bauer
DE
,
Orkin
SH
.
Recent progress in understanding and manipulating haemoglobin switching for the haemoglobinopathies
.
Br J Haematol
.
2018
;
180
(
5
):
630
-
643
.
17.
Dai
Y
,
Chen
T
,
Ijaz
H
,
Cho
EH
,
Steinberg
MH
.
SIRT1 activates the expression of fetal hemoglobin genes
.
Am J Hematol
.
2017
;
92
(
11
):
1177
-
1186
.
18.
Telen
MJ
,
Malik
P
,
Vercellotti
GM
.
Therapeutic strategies for sickle cell disease: towards a multi-agent approach
.
Nat Rev Drug Discov
.
2019
;
18
(
2
):
139
-
158
.
19.
Hannemann
A
,
Rees
DC
,
Tewari
S
,
Gibson
JS
.
Cation homeostasis in red cells from patients with sickle cell disease heterologous for HbS and HbC (HbSC genotype)
.
EBioMedicine
.
2015
;
2
(
11
):
1669
-
1676
.
20.
Setty
BN
,
Kulkarni
S
,
Stuart
MJ
.
Role of erythrocyte phosphatidylserine in sickle red cell-endothelial adhesion
.
Blood
.
2002
;
99
(
5
):
1564
-
1571
.
21.
Setty
BN
,
Rao
AK
,
Stuart
MJ
.
Thrombophilia in sickle cell disease: the red cell connection
.
Blood
.
2001
;
98
(
12
):
3228
-
3233
.
22.
Stuart
MJ
,
Setty
BN
.
Hemostatic alterations in sickle cell disease: relationships to disease pathophysiology
.
Pediatr Pathol Mol Med
.
2001
;
20
(
1
):
27
-
46
.
23.
Lopez Domowicz
DA
,
Welsby
I
,
Esther
CR
Jr.
, et al
.
Effects of repleting organic phosphates in banked erythrocytes on plasma metabolites and vasoactive mediators after red cell exchange transfusion in sickle cell disease
.
Blood Transfus
.
2020
;
18
(
3
):
200
-
207
.
24.
McMahon
TJ
.
Red blood cell deformability, vasoactive mediators, and adhesion
.
Front Physiol
.
2019
;
10
:
1417
.
25.
Rogers
SC
,
Ross
JG
,
d’Avignon
A
, et al
.
Sickle hemoglobin disturbs normal coupling among erythrocyte O2 content, glycolysis, and antioxidant capacity
.
Blood
.
2013
;
121
(
9
):
1651
-
1662
.
26.
Caprari
P
,
Massimi
S
,
Diana
L
, et al
.
Hemorheological alterations and oxidative damage in sickle cell anemia
.
Front Mol Biosci
.
2019
;
6
:
142
.
27.
Nader
E
,
Romana
M
,
Connes
P
.
The red blood cell-inflammation vicious circle in sickle cell disease
.
Front Immunol
.
2020
;
11
:
454
.
28.
D’Alessandro
A
,
Xia
Y
.
Erythrocyte adaptive metabolic reprogramming under physiological and pathological hypoxia
.
Curr Opin Hematol
.
2020
;
27
(
3
):
155
-
162
.
29.
Kaestner
L
,
Bogdanova
A
,
Egee
S
.
Calcium channels and calcium-regulated channels in human red blood cells
.
Adv Exp Med Biol
.
2020
;
1131
:
625
-
648
.
30.
Zennadi
R
.
MEK1/2 as a therapeutic target in sickle cell disease
.
Int J Blood Res Disord
.
2019
;
6
:
6
.
31.
Chen
SY
,
Wang
Y
,
Telen
MJ
,
Chi
JT
.
The genomic analysis of erythrocyte microRNA expression in sickle cell diseases
.
PLoS One
.
2008
;
3
(
6
):
e2360
.
32.
Sangokoya
C
,
Telen
MJ
,
Chi
JT
.
microRNA miR-144 modulates oxidative stress tolerance and associates with anemia severity in sickle cell disease
.
Blood
.
2010
;
116
(
20
):
4338
-
4348
.
33.
Eyler
CE
,
Telen
MJ
.
The Lutheran glycoprotein: a multifunctional adhesion receptor
.
Transfusion
.
2006
;
46
(
4
):
668
-
677
.
34.
Telen
MJ
.
Role of adhesion molecules and vascular endothelium in the pathogenesis of sickle cell disease
.
Hematology Am Soc Hematol Educ Program
.
2007
;
2007
:
84
-
90
.
35.
Zennadi
R
,
Moeller
BJ
,
Whalen
EJ
, et al
.
Epinephrine-induced activation of LW-mediated sickle cell adhesion and vaso-occlusion in vivo
.
Blood
.
2007
;
110
(
7
):
2708
-
2717
.
36.
Zennadi
R
,
Chien
A
,
Xu
K
,
Batchvarova
M
,
Telen
MJ
.
Sickle red cells induce adhesion of lymphocytes and monocytes to endothelium
.
Blood
.
2008
;
112
(
8
):
3474
-
3483
.
37.
Novelli
EM
,
Little-Ihrig
L
,
Knupp
HE
, et al
.
Vascular TSP1-CD47 signaling promotes sickle cell-associated arterial vasculopathy and pulmonary hypertension in mice
.
Am J Physiol Lung Cell Mol Physiol
.
2019
;
316
(
6
):
L1150
-
L1164
.
38.
Vichinsky
EP
,
Neumayr
LD
,
Earles
AN
, et al;
National Acute Chest Syndrome Study Group
.
Causes and outcomes of the acute chest syndrome in sickle cell disease
.
N Engl J Med
.
2000
;
342
(
25
):
1855
-
1865
.
39.
Elmariah
H
,
Garrett
ME
,
De Castro
LM
, et al
.
Factors associated with survival in a contemporary adult sickle cell disease cohort
.
Am J Hematol
.
2014
;
89
(
5
):
530
-
535
.
40.
Farooq
S
,
Testai
FD
.
Neurologic complications of sickle cell disease
.
Curr Neurol Neurosci Rep
.
2019
;
19
(
4
):
17
.
41.
Marks
LJ
,
Munube
D
,
Kasirye
P
, et al
.
Stroke prevalence in children with sickle cell disease in Sub-Saharan Africa: a systematic review and meta-analysis
.
Glob Pediatr Health
.
2018
;
5
:
2333794X18774970
.
42.
Naik
RP
,
Derebail
VK
.
The spectrum of sickle hemoglobin-related nephropathy: from sickle cell disease to sickle trait
.
Expert Rev Hematol
.
2017
;
10
(
12
):
1087
-
1094
.
43.
Fitzhugh
CD
,
Lauder
N
,
Jonassaint
JC
, et al
.
Cardiopulmonary complications leading to premature deaths in adult patients with sickle cell disease
.
Am J Hematol
.
2010
;
85
(
1
):
36
-
40
.
44.
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
(
9
):
886
-
895
.
45.
Niihara
Y
,
Smith
WR
,
Stark
CW
.
A phase 3 trial of l-glutamine in sickle cell disease
.
N Engl J Med
.
2018
;
379
(
19
):
1880
.
46.
Blyden
G
,
Bridges
KR
,
Bronte
L
.
Case series of patients with severe sickle cell disease treated with voxelotor (GBT440) by compassionate access
.
Am J Hematol
.
2018
;
93
(
8
):
E188
-
E190
.
47.
Vichinsky
E
,
Hoppe
CC
,
Ataga
KI
, et al;
HOPE Trial Investigators
.
A phase 3 randomized trial of voxelotor in sickle cell disease
.
N Engl J Med
.
2019
;
381
(
6
):
509
-
519
.
48.
Ataga
KI
,
Kutlar
A
,
Kanter
J
, et al
.
Crizanlizumab for the prevention of pain crises in sickle cell disease
.
N Engl J Med
.
2017
;
376
(
5
):
429
-
439
.
49.
Kutlar
A
,
Kanter
J
,
Liles
DK
, et al
.
Effect of crizanlizumab on pain crises in subgroups of patients with sickle cell disease: a SUSTAIN study analysis
.
Am J Hematol
.
2019
;
94
(
1
):
55
-
61
.
50.
Vichinsky
EP
,
Styles
LA
,
Colangelo
LH
,
Wright
EC
,
Castro
O
,
Nickerson
B
;
Cooperative Study of Sickle Cell Disease
.
Acute chest syndrome in sickle cell disease: clinical presentation and course
.
Blood
.
1997
;
89
(
5
):
1787
-
1792
.
51.
Davies
SC
,
Luce
PJ
,
Win
AA
,
Riordan
JF
,
Brozovic
M
.
Acute chest syndrome in sickle-cell disease
.
Lancet
.
1984
;
1
(
8367
):
36
-
38
.
52.
Gladwin
MT
,
Schechter
AN
,
Shelhamer
JH
,
Ognibene
FP
.
The acute chest syndrome in sickle cell disease. Possible role of nitric oxide in its pathophysiology and treatment
.
Am J Respir Crit Care Med
.
1999
;
159
(
5 pt 1
):
1368
-
1376
.
53.
Sprinkle
RH
,
Cole
T
,
Smith
S
,
Buchanan
GR
.
Acute chest syndrome in children with sickle cell disease. A retrospective analysis of 100 hospitalized cases
.
Am J Pediatr Hematol Oncol
.
1986
;
8
(
2
):
105
-
110
.
54.
Ghosh
S
,
Adisa
OA
,
Chappa
P
, et al
.
Extracellular hemin crisis triggers acute chest syndrome in sickle mice
.
J Clin Invest
.
2013
;
123
(
11
):
4809
-
4820
.
55.
van Agtmael
MA
,
Cheng
JD
,
Nossent
HC
.
Acute chest syndrome in adult Afro-Caribbean patients with sickle cell disease. Analysis of 81 episodes among 53 patients
.
Arch Intern Med
.
1994
;
154
(
5
):
557
-
561
.
56.
Belcher
JD
,
Chen
C
,
Nguyen
J
, et al
.
Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease
.
Blood
.
2014
;
123
(
3
):
377
-
390
.
57.
Ghosh
S
,
Flage
B
,
Weidert
F
,
Ofori-Acquah
SF
.
P-selectin plays a role in haem-induced acute lung injury in sickle mice
.
Br J Haematol
.
2019
;
186
(
2
):
329
-
333
.
58.
Ghosh
S
,
Hazra
R
,
Ihunnah
CA
,
Weidert
F
,
Flage
B
,
Ofori-Acquah
SF
.
Augmented NRF2 activation protects adult sickle mice from lethal acute chest syndrome
.
Br J Haematol
.
2018
;
182
(
2
):
271
-
275
.
59.
Neumayr
LD
,
Hoppe
CC
,
Brown
C
.
Sickle cell disease: current treatment and emerging therapies
.
Am J Manag Care
.
2019
;
25
(
suppl 18
):
S335
-
S343
.
60.
Chaturvedi
S
,
Ghafuri
DL
,
Jordan
N
,
Kassim
A
,
Rodeghier
M
,
DeBaun
MR
.
Clustering of end-organ disease and earlier mortality in adults with sickle cell disease: a retrospective-prospective cohort study
.
Am J Hematol
.
2018
;
93
(
9
):
1153
-
1160
.
61.
Vazquez
B
,
Shah
B
,
Zhang
X
,
Lash
JP
,
Gordeuk
VR
,
Saraf
SL
.
Hyperfiltration is associated with the development of microalbuminuria in patients with sickle cell anemia
.
Am J Hematol
.
2014
;
89
(
12
):
1156
-
1157
.
62.
Cazenave
M
,
Audard
V
,
Bertocchio
JP
, et al
.
Tubular acidification defect in adults with sickle cell disease
.
Clin J Am Soc Nephrol
.
2020
;
15
(
1
):
16
-
24
.
63.
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
(
10
):
1033
-
1048
.
64.
Xu
JZ
,
Garrett
ME
,
Soldano
KL
, et al
.
Clinical and metabolomic risk factors associated with rapid renal function decline in sickle cell disease
.
Am J Hematol
.
2018
;
93
(
12
):
1451
-
1460
.
65.
Anderson
BR
,
Howell
DN
,
Soldano
K
, et al
.
In vivo modeling implicates APOL1 in nephropathy: evidence for dominant negative effects and epistasis under anemic stress [published correction appears in PLoS Genet. 2015;11(9):e1005459]
.
PLoS Genet
.
2015
;
11
(
7
):
e1005349
.
66.
Ashley-Koch
AE
,
Okocha
EC
,
Garrett
ME
, et al
.
MYH9 and APOL1 are both associated with sickle cell disease nephropathy
.
Br J Haematol
.
2011
;
155
(
3
):
386
-
394
.
67.
Charache
S
,
Barton
FB
,
Moore
RD
, et al;
The Multicenter Study of Hydroxyurea in Sickle Cell Anemia
.
Hydroxyurea and sickle cell anemia. Clinical utility of a myelosuppressive “switching” agent
.
Medicine (Baltimore)
.
1996
;
75
(
6
):
300
-
326
.
68.
Steinberg
MH
,
McCarthy
WF
,
Castro
O
, et al;
Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia and MSH Patients’ Follow-Up
.
The risks and benefits of long-term use of hydroxyurea in sickle cell anemia: a 17.5 year follow-up
.
Am J Hematol
.
2010
;
85
(
6
):
403
-
408
.
69.
Thornburg
CD
,
Files
BA
,
Luo
Z
, et al
;
BABY HUG Investigators
.
Impact of hydroxyurea on clinical events in the BABY HUG trial [published correction appears in Blood. 2016;128(24):2869]
.
Blood
.
2012
;
120
(
22
):
4304
-
4310
,
quiz 4448
.
70.
Tshilolo
L
,
Tomlinson
G
,
Williams
TN
, et al;
REACH Investigators
.
Hydroxyurea for children with sickle cell anemia in Sub-Saharan Africa
.
N Engl J Med
.
2019
;
380
(
2
):
121
-
131
.
71.
Voskaridou
E
,
Christoulas
D
,
Bilalis
A
, et al
.
The effect of prolonged administration of hydroxyurea on morbidity and mortality in adult patients with sickle cell syndromes: results of a 17-year, single-center trial (LaSHS)
.
Blood
.
2010
;
115
(
12
):
2354
-
2363
.
72.
Li
Y
,
Maule
J
,
Neff
JL
, et al
.
Myeloid neoplasms in the setting of sickle cell disease: an intrinsic association with the underlying condition rather than a coincidence; report of 4 cases and review of the literature
.
Mod Pathol
.
2019
;
32
(
12
):
1712
-
1726
.
73.
Mvalo
T
,
Topazian
HM
,
Kamthunzi
P
, et al
.
Real-world experience using hydroxyurea in children with sickle cell disease in Lilongwe, Malawi
.
Pediatr Blood Cancer
.
2019
;
66
(
11
):
e27954
.
74.
Opoka
RO
,
Ndugwa
CM
,
Latham
TS
, et al
.
Novel use of hydroxyurea in an African region with malaria (NOHARM): a trial for children with sickle cell anemia
.
Blood
.
2017
;
130
(
24
):
2585
-
2593
.
75.
Power-Hays
A
,
Ware
RE
.
Effective use of hydroxyurea for sickle cell anemia in low-resource countries
.
Curr Opin Hematol
.
2020
;
27
(
3
):
172
-
180
.
76.
Kalpatthi
R
,
Novelli
EM
.
Measuring success: utility of biomarkers in sickle cell disease clinical trials and care
.
Hematology Am Soc Hematol Educ Program
.
2018
;
2018
:
482
-
492
.
77.
National Toxicology Program
.
NTP-CERHR monograph on the potential human reproductive and developmental effects of hydroxyurea
.
NTP CERHR MON
.
2008
;
21
(
21
):
vii
-
III1
.
78.
Lukusa
AK
,
Vermylen
C
.
Use of hydroxyurea from childhood to adult age in sickle cell disease: semen analysis
.
Haematologica
.
2008
;
93
(
11
):
e67
,
discussion e68
.
79.
Chabner
BA
,
Koop
CE
,
Niederhuber
JE
,
Pinedo
HM
.
Homage to Judah Folkman
.
Oncol
.
2008
;
13
(
2
):
205
-
211
.