Curative therapies for sickle cell disease include allogeneic hematopoietic stem cell transplantation (HSCT) and gene-modified autologous stem cell transplantation. HSCT has been used for 30 years with success measured by engraftment, symptom control, graft-vs-host disease (GVHD) risk, organ toxicity, and immune reconstitution. While human leukocyte antigen–matched sibling donor (MSD) transplants have excellent outcomes, alternate donor transplants (unrelated/haploidentical) are just beginning to overcome GVHD and engraftment hurdles to match MSD. Gene therapy, a newly developed treatment, is undergoing careful evaluation in many trials with varying approaches. The risk/benefit ratio to the patient in relation to outcomes, toxicities, and mortality risk drives eligibility for curative interventions. Consequently, eligibility criteria for MSD transplants can be less stringent, especially in the young. Posttransplant outcome analysis after the “cure” with respect to organ function recovery is essential. While established damage such as stroke is irreversible, transplant can help stabilize (pulmonary function), prevent further deterioration (stroke), improve (neurocognition), and protect unaffected organs. Tracking organ functions postintervention uniformly between clinical trials and for adequate duration is essential to answer safety and efficacy questions related to curative therapies. Age-appropriate application/outcome analyses of such therapies will be the ultimate goal in overcoming this disease.

Learning Objectives

  • Apply knowledge regarding the current status of curative therapy interventions to determine indications and eligibility

  • Track organ functions across clinical trials following curative therapies to determine components and extent of cure

“George” was 12 years old with hemoglobin Sβ+ thalassemia. He developed silent cerebral infarcts and acute chest syndrome (ACS) × 2, and he had pain-related admissions impairing school attendance/performance despite hydroxyurea therapy. His hemoglobin was 7.5   g/dL. Hemoglobin F was 8.6%.

His 6-year-old brother with similar hemoglobinopathy had occasional fevers and 1 mild ACS. He was on hydroxyurea therapy. His hemoglobin was 7.2  g/dL; hemoglobin F was 7.8%.

After extensive discussion regarding treatment and natural history, per family preference, they underwent reduced intensity conditioning (RIC) and hematopoietic stem cell transplantation (HSCT) 1 year apart following peripheral blood stem cell collection once from their 10-year-old human leukocyte antigen–matched sibling donor (MSD) with sickle trait.

Now 5 and 4 years post-HSCT, respectively, neither has disease-related symptoms or hemolysis, and both have hemoglobin levels of 13.5  g/dL. George's brain magnetic resonance imaging continues to have the preexisting frontal T2/fluid attenuated inversion recovery (FLAIR) hyperintensity. He has an Individualized Education Plan in school but is grade appropriate.

“Adam,” with hemoglobin SS disease, was 21 years old when he decided to consider HSCT. He has been on chronic red cell transfusion therapy (CRTT) since having an ischemic stroke at 17 months of age and chelation therapy since 4 years of age. He had a right thalamic infarct, left-sided hemiparesis that improved gradually, and learning difficulties in school. He underwent a matched unrelated donor RIC transplant and weaned systemic graft-vs-host disease (GVHD) prophylaxis at 1.5 years. The thalamic infarct remains unchanged 6.5 years post-HSCT. He completed law school and is employed. Pre-HSCT, he had severe c-holelithiasis/pancreatitis. Post-HCT, he has mild pancreatic insufficiency and continues enzyme replacement. Hemoglobin/hemolytic parameters are normal, as are other organ functions.

These cases represent the complex age and symptom variability of the sickle cell disease (SCD) phenotype. The pathology of target organ damage is similar although at a variable pace and severity. Table 1 shows the extensive nature of vasculopathy and organs involved. The most frequent, muscular vaso-occlusive pain episodes (VOEs) are reversible with adequate medical management. Symptom frequency and intensity impair physical and mental well-being and quality of life. Vasculopathy involving vital organs can cause irreversible sequelae, morbidity, and premature mortality.1  Recently approved medications decrease red cell polymerization and endothelial damage to decrease the frequency/severity of VOEs, but long-term benefits on organs are unknown. Curative interventions (allogeneic HSCT/autologous gene-modified HSCT) can change disease course. Safety and efficacy signals are better known for the former and under investigation in the latter.

Table 1.

The spectrum of sickle cell disease manifestations and effects of transplant

OrganPathologyClinical manifestationsEffect of transplant
Blood and vessels Red cell polymerization
Red cell shape distortion
↓ membrane fluidity
Abnormal rheology
Red cell–leukocyte–endothelial adhesive interaction
Oxidative stress 
Hemolysis
Chronic anemia
Inflammation
Infarction 
Reversed 
Brain Stenosis
Velocity perfusion imbalance
Vessel beading
Moyamoya 
Posterior reversible encephalopathy syndrome
Ischemic stroke
Hemorrhagic stroke
Silent infarcts 
Improved or stabilized 
Cardiopulmonary Hemolysis
Chronic anemia
Ischemia reperfusion injury
Red cell polymerization 
Increased left ventricular mass
Left ventricular diastolic dysfunction
Pulmonary hypertension
Acute chest syndrome
Obstructive lung disease
Restrictive lung disease 
Improved or stabilized 
Liver Red cell polymerization
Hemolysis
Hyperbilirubinemia
Iron accumulation 
Sickle hepatopathy
Transfusional hemosiderosis
Biliary disease
Hepatic fibrosis/cirrhosis 
Improved or reversed 
Spleen Congestion, hemolysis
Infarction 
Splenic crisis and congestion
Autosplenectomy 
Improved or stabilized 
Kidneys Intramedullary sickling
Papillary necrosis
Hyperfiltration 
Hematuria
Acute kidney injury
Chronic kidney disease
Renal failure 
Improved or reversed 
Gonads Iron deposition
Microvascular occlusion
Ovarian and testicular infarction 
Urinary tract infection
Dysmenorrhea
Low ovarian reserve
Priapism
Low sperm count 
Stabilized 
Bone and joints Perfusion impediment Osteonecrosis
Avascular necrosis 
Progression halted 
Muscles Perfusion impediment Vaso-occlusive episodes Reversed 
OrganPathologyClinical manifestationsEffect of transplant
Blood and vessels Red cell polymerization
Red cell shape distortion
↓ membrane fluidity
Abnormal rheology
Red cell–leukocyte–endothelial adhesive interaction
Oxidative stress 
Hemolysis
Chronic anemia
Inflammation
Infarction 
Reversed 
Brain Stenosis
Velocity perfusion imbalance
Vessel beading
Moyamoya 
Posterior reversible encephalopathy syndrome
Ischemic stroke
Hemorrhagic stroke
Silent infarcts 
Improved or stabilized 
Cardiopulmonary Hemolysis
Chronic anemia
Ischemia reperfusion injury
Red cell polymerization 
Increased left ventricular mass
Left ventricular diastolic dysfunction
Pulmonary hypertension
Acute chest syndrome
Obstructive lung disease
Restrictive lung disease 
Improved or stabilized 
Liver Red cell polymerization
Hemolysis
Hyperbilirubinemia
Iron accumulation 
Sickle hepatopathy
Transfusional hemosiderosis
Biliary disease
Hepatic fibrosis/cirrhosis 
Improved or reversed 
Spleen Congestion, hemolysis
Infarction 
Splenic crisis and congestion
Autosplenectomy 
Improved or stabilized 
Kidneys Intramedullary sickling
Papillary necrosis
Hyperfiltration 
Hematuria
Acute kidney injury
Chronic kidney disease
Renal failure 
Improved or reversed 
Gonads Iron deposition
Microvascular occlusion
Ovarian and testicular infarction 
Urinary tract infection
Dysmenorrhea
Low ovarian reserve
Priapism
Low sperm count 
Stabilized 
Bone and joints Perfusion impediment Osteonecrosis
Avascular necrosis 
Progression halted 
Muscles Perfusion impediment Vaso-occlusive episodes Reversed 

Indications for curative therapy should account for the risk/benefit ratio. Modern supportive care efficiently prevents SCD-related mortality during childhood in high-income countries. Childhood survival is 93% to 98% depending on disease severity.2  Similarly, registry databases and clinical trial outcome data are available for allogeneic HSCT. These allow outcome tracking/toxicities/reasons for failure and comparison between donor sources and transplant methods. With improving results for cure (>90% disease-free survival in recent reports),3-5  indications for curative interventions will continue to evolve.6  An example is “less severe” disease and HSCT in young patients (<13 years) with MSD.7  It also provides impetus for trials expanding donor options (<25% have an MSD), improving conditioning regimens, and incorporating novel GVHD prophylaxis. Gene therapy averts GVHD risk/immune suppression. Higher-risk transplants (older patients, alternative/mismatched donors, preexisting organ dysfunction) require careful eligibility consideration and care. Acceptable indications for all transplants include damaging disease sequelae such as central nervous system (CNS) ischemia/vasculopathy, recurrent ACS, severe/recurrent VOEs, or mortality predictors such as pulmonary hypertension. Morbidity indicators include CRTT and red cell alloimmunization. Inclusions of nephropathy/retinopathy stem from newer natural history studies. Prevention/progression is independent of reversibility. Figure 1 shows the definitive and “gray” zones of eligibility based on disease risks and HSCT benefits, as highlighted in SCD manifestation differences between George, his brother, and Adam. Current trials of gene therapy include patients 5 to 45 years of age with severe symptoms that justify experimental intervention in the absence of MSD. However, the requirement for chemoablation prior to gene therapy can limit this option for those with severe organ function impairment.

Figure 1.

Expanding transplant indications based on risk/benefit ratio. HLA, human leukocyte antigen; QOL, quality of life; TCD, transcranial Doppler velocity.

Figure 1.

Expanding transplant indications based on risk/benefit ratio. HLA, human leukocyte antigen; QOL, quality of life; TCD, transcranial Doppler velocity.

Close modal

HSCT reverses red cell polymerization and eliminates red cell–leukocyte–endothelial cell damage, oxidative stress, and inflammation. Those with unremitting pain despite conservative management are good candidates for curative therapy.8  Successful HSCT eliminates pain early in children and gradually in adults.9  Reversibility is best noted in patients with acute episodic pain that remits with short-acting opioids, as in the case of George. Chronic pain, as well as that requiring prolonged long-acting opioid therapy, is more difficult. In a cohort of 20 adult patients who underwent a nonmyeloablative matched sibling transplant, risk factors such as older age, more severe and persistent pain before HSCT, and more symptoms of anxiety were associated with persistent pain following transplant.10  Pain control, when achieved, decreases hospital admissions and opioid use. Functional and performance scores, as well as health- related quality of life, improve in the months following and are evident 1 year posttransplant in children following MSD HSCT.11  This improvement in health, health perception, self-esteem, and function was evident in unrelated donor HSCT despite GVHD, following haploidentical transplants, and in adults with established pain prior to HSCT, although there is interindividual variation, more so in adults.12-15  Fatigue remained a lingering issue in adults following myeloablative HSCT.13  Elimination of pain is evident after gene therapy and is a primary outcome measure.16 

Individuals with preexisting end-organ damage/vasculopathy are susceptible to transplant complications—hypertension, consequent posterior reversible encephalopathy syndrome (PRES), and hemorrhagic infarcts due to CNS vasculopathy (compounded by calcineurin inhibitors/steroid therapy/thrombocytopenia). Hence, nuanced supportive care, including transfusions to suppress hemoglobin S pre-HSCT, hemoglobin levels maintained in the low-normal range (9-11  g/dL) to avoid hyperviscosity, aggressive hypertension control (<90th percentile for age), and thrombocytopenia prevention (platelets >50 × 109/L), are important. While PRES suggests reversibility, persistent white matter changes and neurologic dysfunction may persist. Modified preparative regimens and supportive care strategies have decreased PRES. A PRES incidence of 11% to 34% decreased to 7% after eliminating corticosteroids, shortening exposure to calcineurin inhibitors, or substituting with sirolimus.4,12  Modern GHVD prophylaxis strategies that include abatacept or cyclophosphamide result in excellent PRES control.

Both George and Adam presented with SCD-related involvement of the brain in varying severity. Small-caliber brain vessels are common vasculopathy targets that manifest as stenosis, beading, moyamoya, increased flow velocity, perfusion imbalance, and eventually overt ischemic or hemorrhagic stroke or silent infarctions. Historically, overt stroke was an established indication for HSCT, given the CRTT for secondary stroke prevention and the incidence of breakthrough despite this.17  Although control of progression of vascular damage is the goal, the brain is also a susceptible site for ischemia or intracerebral hemorrhage during HSCT or in the immediate post-HSCT period due to the inflammatory/thrombocytopenic milieu. Cerebral vasculopathy, acute complications that alter blood oxygen content, and cerebral blood flow autoregulatory exhaustion are thought to contribute to complications. A retrospective study performed in an SCD transplant consortium found that the risk of neurologic complications post-HSCT was higher with pre-HSCT brain involvement.18 

HSCT provides better neuroprotection than CRTT. Following myeloablative MSD HSCT, stroke recurrence was noted in 2 of 36 recipients with pre-HSCT stroke.19  Subsequent reports in >60 patients demonstrated no strokes after ablative or RIC HSCT and good supportive care.20,21  In contrast, 10% to 20% of patients developed strokes despite continuing CRTT.17,22,23 

Strokes and silent cerebral infarcts represent fixed injury. While HSCT prevents damage extension, established lesions are seldom reversible, making a case for predamage intervention. MSD HSCT significantly decreases transcranial Doppler velocities compared with standard of care by 1 year posttransplant.24  Improvement in dynamic measures of brain injury such as axonal integrity and cerebral metabolic stress normalization can also be demonstrated post-HSCT.25-27  Both George and Adam had radiologic CNS stability posttransplant. Furthermore, in a series of 10 patients who underwent transplantation from MSD or alternate donors, abnormal cerebral blood flows and oxygen extraction fractions returned to normal non-SCD levels 12 to 24 months post-HSCT, a result better than that noted in children receiving chronic red cell transfusion therapy.28 

Cognitive deficits are common among children and adults, with some detected before 1 year of age.29,30  Intelligence quotient (IQ) is projected to decline with age, and domain-specific deficits in executive function, attention, and processing speed are prevalent.31-33  Given the association of cognition with anemia and oxygen saturation, a permanent increase in hemoglobin with less sickling should result in stable to improved cognitive function.

Brain imaging is standard in medical centers performing curative interventions. Neuropsychological assessment is more challenging. In the SCURT trial, a secondary outcome included a battery of neuropsychological tests—IQ and several domain-specific measures. Only 13 of 29 children completed IQ assessments before and 2 years post-HSCT. The IQ post-RIC and unrelated donor HSCT was stable after 2 years.34  In a single- institution experience of 9 children who underwent MSD HSCT, IQ before and 3 years after MSD was stable.35  Another cohort of 16 children and adolescents who underwent haploidentical HSCT also demonstrated stable IQ and improved processing speed after 1 year.36  A French cohort (15 children) had the longest follow-up of 5 years post-MSD HSCT. Stable IQ and improved processing speed were demonstrated.37  Taken together, HSCT recipients consistently demonstrate stabilized/improved cognition. Again, HSCT could be more protective at younger ages prior to the development of vasculopathy. To determine the scope of treatment, it is imperative that domain-specific measures of cognition are included in trials as an aspect of the risks and benefits of intervention.38 

Acute and chronic kidney disease evolves with age. Hyperfiltration and microalbuminuria begin in childhood. Acute complications are frequently accompanied by acute kidney injury. Nonsteroidal anti-inflammatory analgesics used for VOE pain can contribute to chronic kidney disease. By the time HSCT is considered, many patients have clinical or subclinical sickle nephropathy. In 18 children with SCD, hyperfiltration was present in 44% pre-HSCT but declined to 22% at 2 years post-HSCT.39  However, one-third developed hypertension, likely exacerbated by nephrotoxic transplant medications, compared with an incidence of 5.6% pre-HSCT. Adults with SCD have a 12% prevalence of renal failure by 37 years of age, an irreversible change contributing to morbidity and mortality.40  However, peri-HSCT, acute kidney injury was noted early in 58% of recipients, 10% of which was severe (stage 3), again likely due to nephrotoxic agents used early post-HSCT. Thus, minimizing nephrotoxic agents coupled with transplant-mediated control of vaso-occlusion within the acidic/osmolar renal medulla by HSCT can aid preservation of renal function.

Sickle-related hepatic disease can include asymptomatic biliary dysfunction, acute hepatic crisis ± red blood cell sequestration, hyperbilirubinemia, hepatocellular injury, and synthetic function failure, a consequence of vaso-occlusion in hepatic sinusoids and liver parenchyma/terminal bile duct ischemia.41,42  Hepatic iron overload and cirrhosis contribute to chronic liver disease. In a single-center review, 10% of adult SCD deaths were attributed to cirrhosis.43  Post-HSCT, gallstones and consequent pancreatitis should decrease due to the elimination of hemolysis and hyperbilirubinemia. In our case 2, pre-HSCT choledochopancreatic compromise was preexisting, and pancreatic insufficiency manifested after, suggesting compromised reserve. The general theme of preexisting pathology (SCD and iron related), HSCT-induced toxicity such as sinusoidal obstructive syndrome (SOS) necessitates awareness, monitoring, and intervention. Although less of a concern with SCD, hepatic iron with bridging fibrosis is a risk for SOS and graft failure in thalassemia. Adolescents and adults treated with a reduced-intensity (fludarabine/melphalan) or reduced-toxicity (busulfan/fludarabine) regimen did not develop SOS.13,21  HSCT should protect against vaso-occlusion–induced hepatic crisis. Serum ferritin levels decrease following successful transplant and cessation of transfusions, although a preexisting iron load may need to be tackled with phlebotomy or post-HSCT chelation.44  The successful recovery to normal hemoglobin levels and eradication of hemolysis following gene therapy should also offset iron-related pathology in similar manner.45 

Life span is compromised by SCD, with a spike in mortality at 35 to 44 years.1  Cardiopulmonary pathology is the commonest culprit.46  Pathology commences during late childhood/adolescence and includes ACS/pneumonia, pulmonary hypertension, systemic hypertension, congestive cardiac failure, left ventricular diastolic dysfunction, myocardial infarction, and arrhythmia. A higher tricuspid regurgitation jet velocity (>3.1  m/s) and lower hemoglobin (< 8.3  g/dL) were mortality predictors.47,48  HSCT can avoid cardiopulmonary deterioration if accomplished before established pathology. ACS risks receded posttransplant; stability or improvement was noted in restrictive and obstructive pulmonary disease in most patients, measured at 2 years post-HSCT with pulmonary function tests (forced expiratory volume, forced expiratory volume in the first second/forced vital capacity, total lung capacity, and diffusing capacity of the lungs for carbon monoxide).49,50  Similarly, cardiac size and diastolic filling continued to improve in the first year, with a trend toward improvement in the 6-minute walk time.51  Twenty- four patients from this group had a high tricuspid regurgitation velocity (a measure of increased pulmonary pressure and a mortality predictor) at a median of 2.7  m/s, which decreased to 2.3  m/s at 1 year post-HSCT, indicating benefit. These functions, however, continue to require diligent follow-up after curative therapy long term.

Curative therapies for SCD commenced with ablative transplant strategies similar to hematopoietic malignancies about 4 decades ago and have evolved since. Clinical trials have targeted lower-intensity/toxicity conditioning, expanded donor sources to unrelated marrow, expanded cord and haploidentical products, modified GVHD prophylaxis, and enhanced supportive care and monitoring to gradually but definitively improve outcomes and increase HSCT access. Consequently, eligibility for transplant has expanded for indications and age. This expansion results in variable pre-HSCT disease baseline and thus variable outcomes post-HSCT ranging from complete elimination of disease manifestations in the young to stemming progression of complications in older recipients. Baseline disease status also determines time to recovery post-HSCT. The trajectory of recovery is likely to differ with gene therapy approaches as well. As transplant and gene therapy trials proliferate, defining outcomes and success demand uniformity in assessment and compartmentalization based on recipient status. Outcome guidelines have been developed within the American Society of Hematology and transplant consortia such as the Pediatric Transplant and Cellular Therapy Consortium.38,52,53  It is also imperative to track recipients long term to determine late outcomes, and formal efforts to achieve this are now under way.

Monica L. Hulbert is a consultant for Bluebird Bio and Global Blood Therapeutics, has received research funding from Forma Therapeutics and Global Blood Therapeutics, and has family member employment with Pfizer Inc.

Allison A. King has received research funding from Global Blood Therapeutics.

Shalini Shenoy is vice chair on the ASH publications committee, DSMB chair with Aruvant Technologies, a member of NHLBI DSMB, and attended the advisory board meeting for Bristol Myers Squibb.

Monica L. Hulbert: nothing to disclose.

Allison A. King: nothing to disclose.

Shalini Shenoy: nothing to disclose.

1.
Lanzkron
S
,
Carroll
CP
,
Haywood
C
Jr
.
Mortality rates and age at death from sickle cell disease: U.S., 1979-2005
.
Public Health Rep
.
2013
;
128
(
2
):
110
-
116
.
doi:10.1177/003335491312800206
.
2.
Quinn
CT
,
Rogers
ZR
,
McCavit
TL
,
Buchanan
GR
.
Improved survival of children and adolescents with sickle cell disease
.
Blood
.
2010
;
115
(
17
):
3447
-
3452
.
doi:10.1182/blood-2009-07-233700
.
3.
Bolaños-Meade
J
,
Cooke
KR
,
Gamper
CJ
, et al
.
Effect of increased dose of total body irradiation on graft failure associated with HLA-haploidentical transplantation in patients with severe haemoglobinopathies: a prospective clinical trial
.
Lancet Haematol
.
2019
;
6
(
4
):
e183
-
e193
.
doi:10.1016/S2352-3026(19)30031-6
.
4.
Ngwube
A
,
Shah
N
,
Godder
K
,
Jacobsohn
D
,
Hulbert
ML
,
Shenoy
S
.
Abatacept is effective as GVHD prophylaxis in unrelated donor stem cell transplantation for children with severe sickle cell disease
.
Blood Adv
.
2020
;
4
(
16
):
3894
-
3899
.
doi:10.1182/bloodadvances.2020002236
.
5.
de la Fuente
J
,
Dhedin
N
,
Koyama
T
, et al
.
Haploidentical bone marrow transplantation with post-transplantation cyclophosphamide plus thiotepa improves donor engraftment in patients with sickle cell anemia: results of an International Learning Collaborative
.
Biol Blood Marrow Transplant
.
2019
;
25
(
6
):
1197
-
1209
.
doi:10.1016/j.bbmt.2018.11.027
.
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
(
5
):
811
-
820
.
doi:10.3324/haematol.2013.099747
.
7.
King
A
,
Shenoy
S
.
Evidence-based focused review of the status of hematopoietic stem cell transplantation as treatment of sickle cell disease and thalassemia
.
Blood
.
2014
;
123
(
20
):
3089
-
3094
.
doi:10.1182/blood-2013-01-435776
.
8.
Walters
M
,
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
.
doi:10.1016/j.bbmt.2015.10.017
.
9.
Hsieh
MM
,
Fitzhugh
CD
,
Weitzel
RP
, et al
.
Nonmyeloablative HLA-matched sibling allogeneic hematopoietic stem cell transplantation for severe sickle cell phenotype
.
JAMA
.
2014
;
312
(
1
):
48
.
doi:10.1001/jama.2014.7192
.
10.
Darbari
DS
,
Liljencrantz
J
,
Ikechi
A
, et al
.
Pain and opioid use after reversal of sickle cell disease following HLA-matched sibling haematopoietic stem cell transplant
.
Br J Haematol
.
2019
;
184
(
4
):
690
-
693
.
doi:10.1111/bjh.15169
.
11.
Bhatia
M
,
Kolva
E
,
Cimini
L
, et al
.
Health-related quality of life after allogeneic hematopoietic stem cell transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2015
;
21
(
4
):
666
-
672
.
doi:10.1016/j.bbmt.2014.12.007
.
12.
Shenoy
S
,
Eapen
M
,
Panepinto
JA
, et al
.
A trial of unrelated donor marrow transplantation for children with severe sickle cell disease
.
Blood
.
2016
;
128
(
21
):
2561
-
2567
.
doi:10.1182/blood-2016-05-715870
.
13.
Krishnamurti
L
,
Neuberg
DS
,
Sullivan
KM
, et al
.
Bone marrow transplantation for adolescents and young adults with sickle cell disease: results of a prospective multicenter pilot study
.
Am J Hematol
.
2019
;
94
(
4
):
446
-
454
.
doi:10.1002/ajh.25401
.
14.
Kelly
MJ
,
Pennarola
BW
,
Rodday
AM
,
Parsons
SK
,
Journeys to Recovery Study
HS
CT-CHESS™
Study
.
Health-related quality of life (HRQL) in children with sickle cell disease and thalassemia following hematopoietic stem cell transplant (HSCT)
.
Pediatr Blood Cancer
.
2012
;
59
(
4
):
725
-
731
.
doi:10.1002/pbc.24036
.
15.
Saraf
SL
,
Oh
AL
,
Patel
PR
, et al
.
Nonmyeloablative stem cell transplantation with alemtuzumab/low-dose irradiation to cure and improve the quality of life of adults with sickle cell disease
.
Biol Blood Marrow Transplant
.
2016
;
22
(
3
):
441
-
448
.
doi:10.1016/j.bbmt.2015.08.036
.
16.
Kanter
J
,
Walters
MC
,
Krishnamurti
L
, et al
.
Biologic and clinical efficacy of lentiglobin for sickle cell disease
.
N Engl J Med
.
2022
;
386
(
7
):
617
-
628
.
doi:10.1056/NEJMoa2117175
.
17.
Hulbert
ML
,
McKinstry
RC
,
Lacey
JL
, et al
.
Silent cerebral infarcts occur despite regular blood transfusion therapy after first strokes in children with sickle cell disease
.
Blood
.
2011
;
117
(
3
):
772
-
779
.
doi:10.1182/blood-2010-01-261123
.
18.
Stenger
EO
,
Chellapandian
D
,
Shah
RK
, et al
. Prevalence of risk factors for cardiac, pulmonary and neurologic dysfunction following hematopoietic cell transplantation for sickle cell disease: a STAR study. Presented at: the Tandem Meeting
2022
; 24 April 2022;
Salt Lake City, UT
.
19.
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
.
doi:10.1182/blood-2007-03-079665
.
20.
Bernaudin
F
,
Verlhac
S
,
Arnaud
C
, et al
.
Long-term treatment follow-up of children with sickle cell disease monitored with abnormal transcranial Doppler velocities
.
Blood
.
2016
;
127
(
14
):
1814
-
1822
.
doi:10.1182/blood-2015-10-675231
.
21.
King
AA
,
Kamani
N
,
Bunin
N
, et al
.
Successful matched sibling donor marrow transplantation following reduced intensity conditioning in children with hemoglobinopathies
.
Am J Hematol
.
2015
;
90
(
12
):
1093
-
1098
.
doi:10.1002/ajh.24183
.
22.
Scothorn
DJ
,
Price
C
,
Schwartz
D
, et al
.
Risk of recurrent stroke in children with sickle cell disease receiving blood transfusion therapy for at least five years after initial stroke
.
J Pediatr
.
2002
;
140
(
3
):
348
-
354
.
doi:10.1067/mpd.2002.122498
.
23.
Brousse
V
,
Hertz-Pannier
L
,
Consigny
Y
, et al
.
Does regular blood transfusion prevent progression of cerebrovascular lesions in children with sickle cell disease?
Ann Hematol
.
2009
;
88
(
8
):
785
-
788
.
doi:10.1007/s00277-008-0670-x
.
24.
Bernaudin
F
,
Verlhac
S
,
Peffault de Latour
R
, et al
; DREPAGREFFE Trial Investigators.
Association of matched sibling donor hematopoietic stem cell transplantation with transcranial doppler velocities in children with sickle cell anemia
.
JAMA
.
2019
;
321
(
3
):
266
-
276
.
doi:10.1001/jama.2018.20059
.
25.
Costa
TCM
,
Chiari-Correia
R
,
Salmon
CEG
, et al
.
Hematopoietic stem cell transplantation reverses white matter injury measured by diffusion-tensor imaging (DTI) in sickle cell disease patients
.
Bone Marrow Transplant
.
2021
;
56
(
11
):
2705
-
2713
.
doi:10.1038/s41409-021-01365-z
.
26.
Whitehead
MT
,
Smitthimedhin
A
,
Webb
J
, et al
.
Cerebral blood flow and marrow diffusion alterations in children with sickle cell anemia after bone marrow transplantation and transfusion
.
AJNR Am J Neuroradiol
.
2018
;
39
(
11
):
2132
-
2139
.
doi:10.3174/ajnr.A5830
.
27.
Jordan
LC
,
Juttukonda
MR
,
Kassim
AA
, et al
.
Haploidentical bone marrow transplantation improves cerebral hemodynamics in adults with sickle cell disease
.
Am J Hematol
.
2019
;
94
(
6
):
E155
-
E158
.
doi:10.1002/ajh.25455
.
28.
Hulbert
ML
,
Fields
ME
,
Guilliams
KP
, et al
.
Normalization of cerebral hemodynamics following hematopoietic stem cell transplant in children with sickle cell disease [published online ahead of print 30 August 2022]
.
Blood
.
doi:10.1182/blood.2022016618
.
29.
Hoyt
CR
,
Varughese
TE
,
Erickson
J
, et al
.
Developmental delay in infants and toddlers with sickle cell disease: a systematic review
.
Dev Med Child Neurol
.
2022
;
64
(
2
):
168
-
175
.
doi:10.1111/dmcn.15048
.
30.
Martin
S
,
Roderick
MC
,
Abel
C
, et al
.
Neurocognitive functioning in symptomatic adults with sickle cell disease: a description and comparison with unaffected siblings
.
Neuropsychol Rehabil
.
2020
;
30
(
9
):
1666
-
1681
.
doi:10.1080/09602011.2019.1598876
.
31.
King
AA
,
Rodeghier
MJ
,
Panepinto
JA
, et al
.
Silent cerebral infarction, income, and grade retention among students with sickle cell anemia
.
Am J Hematol
.
2014
;
89
(
10
):
E188
-
E192
.
doi:10.1002/ajh.23805
.
32.
Wang
W
,
Enos
L
,
Gallagher
D
, et al
; Cooperative Study of Sickle Cell Disease.
Neuropsychologic performance in school-aged children with sickle cell disease: a report from the Cooperative Study of Sickle Cell Disease
.
J Pediatr
.
2001
;
139
(
3
):
391
-
397
.
doi:10.1067/mpd.2001.116935
.
33.
Prussien
KV
,
Siciliano
RE
,
Ciriegio
AE
, et al
.
Correlates of cognitive function in sickle cell disease: a meta-analysis
.
J Pediatr Psychol
.
2020
;
45
(
2
):
145
-
155
.
doi:10.1093/jpepsy/jsz100
.
34.
King
AA
,
McKinstry
RC
,
Wu
J
, et al
.
Functional and radiologic assessment of the brain after reduced-intensity unrelated donor transplantation for severe sickle cell disease: Blood and Marrow Transplant Clinical Trials Network Study 0601
.
Biol Blood Marrow Transplant
.
2019
;
25
(
5
):
e174
-
e178
.
doi:10.1016/j.bbmt.2019.01.008
.
35.
Woodard
P
,
Helton
KJ
,
Khan
RB
, et al
.
Brain parenchymal damage after haematopoietic stem cell transplantation for severe sickle cell disease
.
Br J Haematol
.
2005
;
129
(
4
):
550
-
552
.
doi:10.1111/j.1365-2141.2005.05491.x
.
36.
Prussien
KV
,
Patel
DA
,
Wilkerson
K
, et al
.
Improvement in processing speed following haploidentical bone marrow transplant with posttransplant cytoxan in children and adolescents with sickle cell disease
.
Pediatr Blood Cancer
.
2020
;
67
(
1
):
e28001
.
doi:10.1002/pbc.28001
.
37.
Bockenmeyer
J
,
Chamboredon
E
,
Missud
F
, et al
.
Development of psychological and intellectual performance in transplanted sickle cell disease patients: a prospective study from pretransplant period to 5 years after HSCT
.
Arch Pediatr
.
2013
;
20
(
7
):
723
-
730
.
doi:10.1016/j.arcped.2013.04.012
.
38.
Farrell
AT
,
Panepinto
J
,
Carroll
CP
, et al
.
End points for sickle cell disease clinical trials: patient-reported outcomes, pain, and the brain
.
Blood Adv
.
2019
;
3
(
23
):
3982
-
4001
.
doi:10.1182/bloodadvances.2019000882
.
39.
Pedersen
SJV
,
Monagel
DA
,
Mammen
C
,
Lewis
VA
,
Guilcher
GMT
,
Bruce
AA
.
Stable renal function in children and adolescents with sickle cell disease after nonmyeloablative hematopoietic stem cell transplantation
.
Pediatr Blood Cancer
.
2020
;
67
(
9
):
e28568
.
doi:10.1002/pbc.28568
.
40.
Powars
DR
,
Chan
LS
,
Hiti
A
,
Ramicone
E
,
Johnson
C
.
Outcome of sickle cell anemia: a 4-decade observational study of 1056 patients
.
Medicine (Baltimore)
.
2005
;
84
(
6
):
363
-
376
.
doi:10.1097/01.md.0000189089.45003.52
.
41.
Suddle
AR
.
Management of liver complications in sickle cell disease
.
Hematology Am Soc Hematol Educ Program
.
2019
;
2019
(
1
):
345
-
350
.
doi:10.1182/hematology.2019000037
.
42.
Kyrana
E
,
Rees
D
,
Lacaille
F
, et al
.
Clinical management of sickle cell liver disease in children and young adults
.
Arch Dis
Child,
2021
;
106
(
4
):
315
-
320
.
doi:10.1136/archdischild-2020-319778
.
43.
Darbari
DS
,
Kple-Faget
P
,
Kwagyan
J
,
Rana
S
,
Gordeuk
VR
,
Castro
O
.
Circumstances of death in adult sickle cell disease patients
.
Am J Hematol
.
2006
;
81
(
11
):
858
-
863
.
doi:10.1002/ajh.20685
.
44.
Alzahrani
M
,
Damlaj
M
,
Jeffries
N
, et al
.
Non-myeloablative human leukocyte antigen-matched related donor transplantation in sickle cell disease: outcomes from three independent centres
.
Br J Haematol
.
2021
;
192
(
4
):
761
-
768
.
doi:10.1111/bjh.17311
.
45.
Frangoul
H
,
Altshuler
D
,
Cappellini
MD
, et al
.
CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia
.
N Engl J Med
.
2021
;
384
(
3
):
252
-
260
.
doi:10.1056/NEJMoa2031054
.
46.
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
.
doi:10.1002/ajh.21569
.
47.
Gladwin
MT
,
Barst
RJ
,
Gibbs
JS
, et al
; walk-PHaSST Investigators and Patients.
Risk factors for death in 632 patients with sickle cell disease in the United States and United Kingdom
.
PLoS One
.
2014
;
9
(
7
):
e99489
.
doi:10.1371/journal.pone.0099489
.
48.
Kassim
AA
,
Payne
AB
,
Rodeghier
M
,
Macklin
EA
,
Strunk
RC
,
DeBaun
MR
.
Low forced expiratory volume is associated with earlier death in sickle cell anemia
.
Blood
.
2015
;
126
(
13
):
1544
-
1550
.
doi:10.1182/blood-2015-05-644435
.
49.
Walters
MC
,
Hardy
K
,
Edwards
S
, et al
;
Multicenter Study of Bone Marrow Transplantation for Sickle Cell Disease
.
Pulmonary, gonadal, and central nervous system status after bone marrow transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2010
;
16
(
2
):
263
-
272
.
doi:10.1016/j.bbmt.2009.10.005
.
50.
Friedman
D
,
Dozor
AJ
,
Milner
J
, et al
.
Stable to improved cardiac and pulmonary function in children with high-risk sickle cell disease following haploidentical stem cell transplantation
.
Bone Marrow Transplant
.
2021
;
56
(
9
):
2221
-
2230
.
doi:10.1038/s41409-021-01298-7
.
51.
Sachdev
V
,
Hsieh
M
,
Jeffries
N
, et al
.
Reversal of a rheologic cardiomyopathy following hematopoietic stem cell transplantation for sickle cell disease
.
Blood Adv
.
2019
;
3
(
19
):
2816
-
2824
.
doi:10.1182/bloodadvances.2019000387
.
52.
Farrell
AT
,
Panepinto
J
,
Desai
AA
, et al
.
End points for sickle cell disease clinical trials: renal and cardiopulmonary, cure, and low-resource settings
.
Blood Adv
.
2019
;
3
(
23
):
4002
-
4020
.
doi:10.1182/bloodadvances.2019000883
.
53.
Shenoy
S
,
Gaziev
J
,
Angelucci
E
, et al
.
Late effects screening guidelines after hematopoietic cell transplantation (HCT) for hemoglobinopathy: consensus statement from the second pediatric blood and marrow transplant Consortium International Conference on Late Effects after Pediatric HCT
.
Biol Blood Marrow Transplant
.
2018
;
24
(
7
):
1313
-
1321
.
doi:10.1016/j.bbmt.2018.04.002
.