Abstract

An increasing number of allogeneic transplant and autologous gene modification transplant therapies seek to eradicate sickle hemoglobin and the consequent hemolysis, vasculopathy, functional compromise, morbidity, and mortality. Because these modalities are used in parallel, it is important to be able to define the spectrum and stability of correction, long-term effects, and the pros and cons of each modality. A comparison between interventions that will be sought by providers and patients undergoing intervention requires uniform assessments that evaluate disease- and intervention-related effects for informed decision-making. This expert summary outlines a pathway to functional evaluations with timing recommendations, provides broad management guidelines, and touches upon ongoing research efforts in the field. The road map for long-term follow-up can help clinicians and researchers choose assessments and time them in comparable fashion between the various transformative therapy efforts.

Knowledge regarding genetics, pathophysiology, and natural history of sickle cell disease (SCD) has evolved significantly, resulting in improved understanding of complications, morbidity, and mortality and expansion of treatment choices. Curative therapies have expanded in parallel, ranging from HLA-matched sibling transplantation to alternate donor transplantation (allogeneic hematopoietic cell transplant [allo-HCT]) and autologous gene-modified cell transplantation (GT).1-7 Although results are positive across therapies, outcome analyses are of short duration (restricted by trial guidelines) and highly variable between trials. With increasing pathways to disease eradication, it is important to define treatment efficacy in relation to age, disease, and modality as uniformly as possible to allow for just comparison of advantages and risks. This can be achieved with a combination of clinical and laboratory investigation–based end points. The American Society of Hematology, in collaboration with the US Food and Drug Administration, defines outcomes as a reflection of patient desires integrated with objective measurements to assess disease status, amenable to easy measurement at low cost, providing relevant, interpretable, and comparable information and facilitating complete data collection.8 Data collected using comparable methods across therapies at similar time points will help inform families and providers define expectations, provide insights into interventions after therapy, and help choose the best approach of therapy for individual patients. Additional progress is achieved with further research for better end points that are easier or superior. Such research, initially restricted to smaller focused trials, is then adopted for general application if useful. We summarize practical guidelines for long-term follow-up based on existing data and discuss opportunities for further research.

Comparison between treatment approaches is based on objective measures that define positive and negative outcomes. Curative end points should measure the restoration of physical and psychosocial quality of life to levels similar to unaffected peers, correct laboratory parameters, and abrogate medical needs connected to the disease or intervention. The benefit should be stable over the long term, spanning a normal life span. Currently, there are few validated measures, and high variability exists in the description of outcomes between curative therapy trials and in comparisons with conservative SCD management protocols. Thus, the recommendations included here are provided by experts in the field as a working summary of long-term follow-up assessments and associated parameters, along with timing guidelines whenever possible, with a goal to practice harmony. The summary is organized to include individual organ functions, graft function, health-related quality of life (HRQOL), pain perception, patient-related outcomes, and transition of care from childhood to adulthood to encompass lifetime care and monitoring for individuals with SCD treated with curative intention and provide timelines for monitoring to allow for comparison.

Brain

The risk of overt cerebrovascular disease is often a reason to seek allo-HCT/GT in SCD. It is important to prevent new pathology and stabilize established injury. By age 30 years, 50% of adults with hemoglobin SS and Sβ0 will have a silent cerebral infarct,9 and without transcranial Doppler velocity (TCD) screening and transfusion, 5% to 10% will suffer an overt stroke.10 Cognitive deficits are common with and without visible cerebrovascular pathology on imaging. The etiology is multifactorial, but medical risk factors include hypoxia,11 covert cerebrovascular disease,12,13 reduced arterial oxygen content,14 and a compensatory increase in cerebral blood flow.15,16 Allo-HCT/GT results in higher recipient hemoglobin levels and decreased sickling parameters, which should result in improved oxygen-carrying capacity. In keeping with this, allo-HCT survivors have demonstrated reduced cerebral blood flow17,18 and improved oxygen extraction18-21 after intervention. The risk of progressive cerebrovascular injury visible by magnetic resonance imaging (MRI) is lower after HCT22-27 than after chronic transfusion therapy. In parallel with the above-mentioned observations, stabilization in intelligence quotient and some fluid neurocognitive domains such as processing speed are described.22,23,28-31 The effects of GT on cerebrovascular outcomes are unknown because only a small number of patients with stroke have undergone the procedure. As more patients with SCD undergo allo-HCT/GT, systematic assessments of cerebrovascular outcomes are essential for comparison and determination of stability as described in Table 1. Modalities of assessment include MRI and TCD measurements. TCD, however, requires bone windows (limited to <16 years) and is operator expertise dependent.

Table 1.

Recommended assessment of brain function

BrainAssessmentInterval
Clinical MRI and MRA of the brain
TCD if <16 years (if expertise available) 
Before intervention
Then yearly ×2
At 5 years after intervention
Repeat every 5 years or earlier as needed in the event of symptoms 
 Cognitive assessment 
Global measures such as Bayley-III (0-3.5 years), intelligence quotient (WPPSI-IV and WISC-V). Domain-specific measures: executive function working memory, attention, and processing speed 
Before intervention
Then yearly ×2
At 5 years after intervention
Repeat every 5 years or earlier as needed in the event of intervention 
Research Cerebral hemodynamics: cerebral blood flow, oxygen extraction fraction, cerebrovascular reactivity, and MR velocity Before intervention
Then yearly ×2
Repeat at 5 years and continue beyond if impaired 
BrainAssessmentInterval
Clinical MRI and MRA of the brain
TCD if <16 years (if expertise available) 
Before intervention
Then yearly ×2
At 5 years after intervention
Repeat every 5 years or earlier as needed in the event of symptoms 
 Cognitive assessment 
Global measures such as Bayley-III (0-3.5 years), intelligence quotient (WPPSI-IV and WISC-V). Domain-specific measures: executive function working memory, attention, and processing speed 
Before intervention
Then yearly ×2
At 5 years after intervention
Repeat every 5 years or earlier as needed in the event of intervention 
Research Cerebral hemodynamics: cerebral blood flow, oxygen extraction fraction, cerebrovascular reactivity, and MR velocity Before intervention
Then yearly ×2
Repeat at 5 years and continue beyond if impaired 

MRI, Magnetic Resonance Imaging; MRA, Magnetic Resonance Angiography; TCD, Transcranial Doppler velocity; WISC-V, Wechsler Intelligence Scale for Children-Fifth edition; WPPSI-IV, Wechsler Preschool and Primary Scale of Intelligence-Fourth edition; MR, Magnetic Resonance.

For specific measures, refer to Tables 5 and 6 in end points for clinical trials in SCD.8 

Cardiopulmonary

Cardiopulmonary complications are a significant cause of morbidity32 and early mortality33,34 and manifest as acute chest syndrome, pulmonary hypertension, and cardiopulmonary hypofunction, all predictors of mortality.35-39 Cardiopulmonary pathology is influenced negatively by advancing age, disease progression, allergens and asthma, and external pathogens such as infections and toxins. Along with symptom improvement (such as abrogation of acute chest syndrome), stabilization with or without improvement of pulmonary functions (forced expiratory volume in 1 second and forced vital capacity) has been demonstrated after HCT.27,40-48 Improvement in assessments such as diffusing capacity of the lungs for carbon monoxide, although largely limited to children, has also shown stabilization in some adults.27,41,44,46,47 Descriptions of cardiac function are traditionally limited to tricuspid regurgitant jet (TRJ) velocity in adolescence and adulthood.27,42,43,45,47,49,50 Decrease in TRJ velocity has been demonstrated after HCT, in even higher magnitude in adults than in children. In contrast, recently, an asymptomatic drop in shortening fraction and ejection fractions (EFs) have been detected after HCT, drawing attention to potential cardiotoxicity of conditioning agents, especially after disease-related cardiac involvement.27,42,43,47 A cardiopulmonary review and meta-analysis51 that examined pulmonary (forced expiratory volume in 1 second, forced vital capacity, and diffusing capacity of the lungs for carbon monoxide) and cardiac (TRJ velocity, shortening fraction, and EF) functions described stability after HCT on all parameters except for a significant decrease in EF. These standard measures have been supplanted by measures of cardiac stress (high brain natriuretic peptide and cardiac MRI changes) as more sensitive measures of change. A decrease in myocardial fibrosis after HCT can be detected by cardiac MRI.52,53 Such myocardial fibrosis may not be preventable with supportive care and supplants evaluation in adolescent/young adults with evidence of left heart dilation or prominent trabeculations on echocardiogram.39 Recommended assessments are listed in Table 2.

Table 2.

Recommended assessment of cardiac, pulmonary, renal, and gonadal functions

AssessmentInterval
Lungs   
Clinical PFTs: FEV1, FVC, TLC, and DLCO Before intervention, 6 months, 12 months, yearly thereafter for 5 years, and beyond 5 years if abnormality detected/persists 
PFT changes in the presence of cGVHD or infections  
Research PFT changes in the presence of external variables/environmental pathogens such as smoking, vaping, and reactive airway disease Will require assessment at more frequent intervals 
Heart   
Clinical Echocardiogram including LVEF, SF, and TRJV Before intervention, 6 months, 12 months, yearly thereafter for 5 years, and beyond 5 years if abnormality detected/persists 
Research 6-minute walk test Before intervention, yearly for 2 years, every alternate year thereafter. 
NT-proBNP Continue beyond 5 years if abnormality detected/persists 
CMRi  
Kidneys   
Clinical BUN, creatinine, electrolytes, GFR, or 24-hour creatinine clearance Before intervention 
Urine analysis for blood/protein Annually until nephrotoxic medications discontinued. 
Microalbuminuria (albumin-to-creatinine ratio) Continue to monitor every 2 years and beyond due to chronic nature of disease 
Blood Pressure  
Research Renal injury biomarkers such as urinary KIM-1, NGAL, MCP-1, and urine elafin Before and after intervention; and at least every 1-2 years after HCT or GT 
Ovaries   
Clinical AMH, LH, estradiol, and FSH Before intervention and then yearly, based on age 
Pubertal changes Intervention: estrogen supplementation  
Premature ovarian insufficiency   
Oocyte cryopreservation  Per patient preference 
Ovarian tissue cryopreservation for implantation  Per patient preference 
Testicles   
Clinical FSH, LH, and testosterone Before intervention and then yearly, based on age 
Pubertal changes Semen analysis if applicable Per patient preference 
Sperm cryopreservation Intervention such as testosterone supplementation  
Research Testicular tissue implantation Per patient preference 
Testicular tissue cryopreservation   
AssessmentInterval
Lungs   
Clinical PFTs: FEV1, FVC, TLC, and DLCO Before intervention, 6 months, 12 months, yearly thereafter for 5 years, and beyond 5 years if abnormality detected/persists 
PFT changes in the presence of cGVHD or infections  
Research PFT changes in the presence of external variables/environmental pathogens such as smoking, vaping, and reactive airway disease Will require assessment at more frequent intervals 
Heart   
Clinical Echocardiogram including LVEF, SF, and TRJV Before intervention, 6 months, 12 months, yearly thereafter for 5 years, and beyond 5 years if abnormality detected/persists 
Research 6-minute walk test Before intervention, yearly for 2 years, every alternate year thereafter. 
NT-proBNP Continue beyond 5 years if abnormality detected/persists 
CMRi  
Kidneys   
Clinical BUN, creatinine, electrolytes, GFR, or 24-hour creatinine clearance Before intervention 
Urine analysis for blood/protein Annually until nephrotoxic medications discontinued. 
Microalbuminuria (albumin-to-creatinine ratio) Continue to monitor every 2 years and beyond due to chronic nature of disease 
Blood Pressure  
Research Renal injury biomarkers such as urinary KIM-1, NGAL, MCP-1, and urine elafin Before and after intervention; and at least every 1-2 years after HCT or GT 
Ovaries   
Clinical AMH, LH, estradiol, and FSH Before intervention and then yearly, based on age 
Pubertal changes Intervention: estrogen supplementation  
Premature ovarian insufficiency   
Oocyte cryopreservation  Per patient preference 
Ovarian tissue cryopreservation for implantation  Per patient preference 
Testicles   
Clinical FSH, LH, and testosterone Before intervention and then yearly, based on age 
Pubertal changes Semen analysis if applicable Per patient preference 
Sperm cryopreservation Intervention such as testosterone supplementation  
Research Testicular tissue implantation Per patient preference 
Testicular tissue cryopreservation   

AMH, anti-Müllerian hormone; BUN, blood urea nitrogen; cGVHD, chronic GVHD; CMRi, cardiac MRI; DLCO, diffusion capacity of the lungs for carbon monoxide; FEV1, forced expiratory volume in 1 second; FSH, follicle-stimulating hormone; FVC, forced vital capacity; GFR, glomerular filtration rate; GT, gene therapy; KIM-1, Kidney Injury Molecule-1; LH, luteinizing hormone; LVEF, left ventricular ejection fraction; MCP-1, monocyte chemoattractant protein-1; NGAL, neutrophil gelatinase-associated lipocalin; NT-proBNP, N-terminal pro B-type natriuretic peptide; PFT, pulmonary function tests; SF, shortening fraction; TLC, total lung capacity; TRJV, TRJ velocity.

Kidneys

Chronic kidney disease manifests as defective urine concentration, glomerular hyperfiltration (increased renal perfusion), electrolyte imbalance (tubular dysfunction), and albuminuria.54 Hyperfiltration is detected as early as age 2.5 years. As glomerular damage progresses, the glomerular filtration rate declines into adulthood, signaling worsening nephropathy. Microinfarcts and ischemic injury upregulate endothelin-1 and reactive oxygen species, resulting in medullary hypoxia and papillary necrosis. Free hemoglobin, caused by ongoing hemolysis, is toxic to the tubules. Decreased hemolysis (such as coinherited α-thalassemia) can be protective to this effect. Microalbuminuria worsens with age, affecting >60% of patients aged >45 years. Microalbuminuria >100 mg/g before allo-HCT/GT may not be reversible.55 Risk factors for acute kidney injury in SCD include anti-inflammatory medications, heart failure, dehydration, sepsis, thrombosis, and rhabdomyolysis.56 

Allo-HCT/GT can affect already compromised kidneys via medications used for conditioning, graft-versus-host disease (GVHD) prophylaxis, hypertension, infections, and thrombotic microangiopathy.56 Serial monitoring and supportive care are essential to maintain renal function and combat hypertension to promote healing. In children, diminished glomerular filtration rate (assessment formulas include CKiD U25 for children and young adults) has been described after myeloablative chemotherapy; correction of hyperfiltration and normalization of renal volume and structure have been described after nonmyeloablative transplants. Long-term renal outcomes after allo-HCT/GT in adults with established chronic kidney disease is unknown.

Functional parameters of renal function should be tracked after allo-HCT/GT. Pathologic albuminuria (albumin-to-creatinine ratio of >30 mg/g) can be exacerbated by complications such as hypertension, infection, and GVHD. Proteinuria (albumin-to-creatinine ratio of >300 mg/g) is predictive of increased mortality at 1 year after HCT.55 Novel biomarkers of renal dysfunction, such as Kidney Injury Molecule 1 (KIM-1), Neutrophil Gelatinase–Associated Lipocalin, Monocyte Chemoattractant Protein 1, and urine elafin, are under investigation for early detection of renal injury and sensitivity of the rate of change.57,58 Recommended assessments are listed in Table 2.

Reproduction

At baseline, SCD complications and medications may result in low ovarian reserve, reduced sperm counts, and infertility.59,60 These risks are compounded by gonadotoxic agents and irradiation used in allo-HCT/GT protocols. Gonadal outcomes should be discussed before embarking on intervention. Fertility preservation is indicated for all individuals who have not completed their families and wish to do so.34 Ensuring safe35 and accessible care,36 including assistance with collection of sperm, ova or gonadal tissue (needs planning around hydroxyurea discontinuation for 2-3 months if applicable), and storage cost planning, is essential. Ovarian tissue cryopreservation is considered standard of care before considering gonadotoxic therapy. Testicular tissue cryopreservation is undertaken under research protocols when sperm collection is not feasible due to age. Access to in vitro fertilization after intervention is an area of active advocacy.37 

Although limited fertility preservation outcomes are described, anecdotal pregnancy outcomes are described and expected to increase with accessibility.61-63 Follow-up care for reproductive organs after intervention also require additional multispecialty involvement. The best follow-up care is provided by synergy between endocrinologists, urologists, reproductive endocrinology-infertility physicians, and maternal-fetal medicine specialists.

After allo-HCT/GT, patients require gonadal function monitoring.59 For people with ovaries, annual testing with anti-Müllerian hormone, luteinizing hormone, estradiol, and follicle-stimulating hormone is indicated. Adolescent and adult females should be screened for clinical symptoms of stalled puberty and premature ovarian insufficiency.59 Hormone induction or replacement therapy may be required. For people with testicles, annual screening also includes testosterone level; semen analysis should be offered when appropriate (Table 2). Although oocyte recovery does not significantly improve after HCT, sperm production can.

During follow-up, individuals should be queried for pregnancy intention.60 Individuals attempting unassisted conception for over 6 months warrant referral for infertility care. Individuals who have cryopreserved gonadal tissue, gametes, or embryos and wish to pursue pregnancy with assistance also need referral to reproductive endocrinology and infertility specialists. Conversely, posttherapy patients of reproductive age require counseling regarding limitations of fertility testing and the possibility of unassisted pregnancy. Individuals undergoing allo-HCT/GT should be counseled that their gametes contain a hemoglobinopathy trait that is heritable. A discussion of in vitro fertilization with preimplantation testing may be applicable to some.64 

Additional organs

Growth parameters and thyroid and adrenal function monitoring are essential after allo-HCT/GT because all interventions currently use chemotherapy ± radiation to achieve desired goals. Exposure can invoke toxicities based on age, physiology, and disease status. Toxicities compound preexisting pathology or commence de novo. GVHD is a risk with allo-HCT. The risk of disorders of glucose regulation, lipid metabolism, and metabolic syndromes, which are genetic/environmental, may exacerbate after allo-HCT/GT and consequent lifestyle change during the recovery period. Comprehensive care also includes bone care, dental and visual health, and management of iron overload if present (Table 3).

Table 3.

Recommended assessment of additional organ functions

OrganAssessmentInterval
Growth Height, weight, and body mass index Yearly ×2 
Hormone levels if short stature (IGF-1 and IGFBP-3) Subsequently yearly if abnormal and within growth period 
Bone age  
Thyroid function Thyroid-stimulating hormone At 6 months, 1 year, and then annually 
Free thyroxine level  
Glucose metabolism Fasting blood sugar Annual 
Glucose tolerance test As needed 
Lipid metabolism Fasting lipid panel Annual 
Bone health Vitamin D3 level Annual 
Bone mineral density 2 years and subsequently if intervention planned 
Dental health Dental evaluation Annual 
Visual function Vision test Annual 
Glaucoma screening  
Retinal vessel assessment  
Iron accumulation Serum ferritin and transferrin saturation Before intervention 
Cardiac and liver MRI 6 months, 1 year, and yearly until normal with therapy/phlebotomy 
OrganAssessmentInterval
Growth Height, weight, and body mass index Yearly ×2 
Hormone levels if short stature (IGF-1 and IGFBP-3) Subsequently yearly if abnormal and within growth period 
Bone age  
Thyroid function Thyroid-stimulating hormone At 6 months, 1 year, and then annually 
Free thyroxine level  
Glucose metabolism Fasting blood sugar Annual 
Glucose tolerance test As needed 
Lipid metabolism Fasting lipid panel Annual 
Bone health Vitamin D3 level Annual 
Bone mineral density 2 years and subsequently if intervention planned 
Dental health Dental evaluation Annual 
Visual function Vision test Annual 
Glaucoma screening  
Retinal vessel assessment  
Iron accumulation Serum ferritin and transferrin saturation Before intervention 
Cardiac and liver MRI 6 months, 1 year, and yearly until normal with therapy/phlebotomy 

IGF-1, insulin-like growth factor 1; IGFBP-3, insulin-like growth factor binding protein-3.

Infection is a major cause of morbidity and mortality in SCD, contributing to 20% to 50% of deaths.65 Vulnerability to infection results from hyposplenism or asplenia.66 Additional genetic etiologies such as defective complement activation,67 micronutrient zinc deficiency,68 HLA-II subtype polymorphism,69 dysfunctional T/B cells and Fc receptors, and transforming growth factor β/bone morphogenetic protein pathways have been associated with an increased risk of bacteremia.70 Impairment in alternative complement pathways67 and abnormal disease macro and microenvironments contribute to this predisposition.71 

Allo-HCT–mediated immunosuppression prevails until immune reconstitution (IR).72 IR is inversely proportional to age and influenced by graft source, HLA match, conditioning, engraftment kinetics, and GVHD prophylaxis/treatment.73,74 Infection risks are highest before cellular IR (before day +100) and then recede unless systemic immunosuppression is intensified.75 Within this framework, variability is noted due to variations in transplant parameters, such as donor source, age, and disease severity.1-5,24,76,77 

T-cell recovery is most efficient in young recipients with preserved thymic function undergoing sibling HCT. Function is more impaired after cord blood transplant, immunoablative conditioning, advanced age, mismatched HCT, and prolonged GVHD-related immunosuppression.78,79 Initial T-cell recovery occurs with peripheral expansion of donor memory CD8+ T cells and subsequently with thymus-generated donor T cells from hematopoietic precursors.80,81 Thymic involution (age related) impairs CD4+ T-cell repertoire recovery.82 B-cell counts recover by 9 months after HCT; antibody repertoire recovery is aided by CD4+ T cells.82 Haploidentical HCT using posttransplant cyclophosphamide GVHD prophylaxis results in preferential accumulation of regulatory T cells; de novo donor T-cell recovery takes a year, rendering patients susceptible to viral infections.75,83-86 In GT patients, IR should commence with the recovery of the cellular immune system after myeloablation and is expected within 3 to 6 months of product reinfusion.

Assessment of IR includes absolute lymphocyte counts, lymphocyte subsets (CD4+ and CD8+ T cells, natural killer cells, and B cells), and antibody titers. Lymphocyte recovery, notably the CD4+ T-cell compartment, correlates with survival and decreased opportunistic infections.87-91 Functional assays, including polyfunctional T cells and virus-specific responses (such as cytomegalovirus and Epstein-Barr virus) can provide important information on immune recovery.92 Molecular assessments including T-cell receptor repertoire, B-cell receptor diversity, T-cell receptor excision circles, and kappa-deleting recombination excision circles can assess thymopoiesis better to evaluate opportunistic infection risks.93,94 Tests for IR are listed in Table 4.

Table 4.

Recommended assessment of IR

IRAssessmentInterval
Clinical Absolute numbers of NK cells, CD4+ and CD8+ T cells, B cells, and T-regs 3, 6, 9, 12, 18, and 24 months after HSCT; subsequently at regular intervals until normal 
Antibody titers (to immunization antigens such as tetanus, diphtheria, and hepatitis) Assess antibody titers 4-6 weeks after reimmunization 
Research Functional T-cell assays At regular intervals after 6 months until normalized to define pace of reconstitution 
Virus specific responses  
TREC and KREC evaluation  
T-cell receptor repertoire  
B-cell receptor diversity  
IRAssessmentInterval
Clinical Absolute numbers of NK cells, CD4+ and CD8+ T cells, B cells, and T-regs 3, 6, 9, 12, 18, and 24 months after HSCT; subsequently at regular intervals until normal 
Antibody titers (to immunization antigens such as tetanus, diphtheria, and hepatitis) Assess antibody titers 4-6 weeks after reimmunization 
Research Functional T-cell assays At regular intervals after 6 months until normalized to define pace of reconstitution 
Virus specific responses  
TREC and KREC evaluation  
T-cell receptor repertoire  
B-cell receptor diversity  

KRECs, Kappa-deleting recombination excision circles; NK cells, natural killer cells; TRECs, T-cell receptor excision circles; T-regs, regulatory T cells.

Delayed IR adds to mortality risk even after correction of SCD pathology.80,95 Allo-HCT/GT is undertaken in the context of functional or surgical asplenia and increased susceptibility to bacterial infections.65 After allo-HCT, B-cell counts typically recover between 3 and 12 months and CD4+ T cells between 6 and 9 months in children and later in adults, impairing vaccine response. Without revaccination, antibody titers to vaccine-preventable diseases decline during the first decade after HCT. Hence, revaccination after allo-HCT/GT should be commenced at partial recovery. Patients exposed to nonlymphodepleting busulfan may retain vaccine memory, but long-term data on persistence of titers are lacking.96 Inactivated or cell-free vaccination can be resumed at 3 to 6 months; live-attenuated vaccines are administered 24 months after HCT (supplemental Table 1) and are avoided in patients on systemic immunosuppression for GVHD control.97 Postexposure prophylaxis can prevent morbidity after potential or documented exposure in immunocompromised patients (supplemental Table 2). Recently, the US Food and Drug Administration approved pemivibart, an anti-COVID monoclonal antibody, for immunocompromised patients as preexposure prophylaxis.

Psychologic changes follow physical manifestations in SCD, increase with age and disease progression, and affect activities of daily living and consequently HRQOL.98 HRQOL is a multidimensional concept representing the general perception of physical, psychological, and social aspects of life.99,100 It is a subjective rating that allows for a general assessment of well-being.98 

HRQOL assessments after allo-HCT/GT are important tools for assessing treatment benefit in disorders that are not imminently life threatening.101 These assessments can quantify the impacts of a disease-free state while considering transplant complications, such as infections, organ damage, or chronic GVHD. HRQL may represent the most clinically relevant measure of treatment toxicity.102 

Studies evaluating HRQOL after allo-HCT have shown variable results. Pediatric studies have shown an initial decline in the peri-transplant period, followed by an improvement in HRQOL by 1 year after HCT.102,103 In adults, after nonablative conditioning, there was no decline in HRQOL at day +30.104 This variability may be due to effects of donor source, conditioning regimens, and transplant complications. More pediatric patients are now choosing curative treatment before significant complications have affected HRQOL.

Numerous HRQOL evaluations now provide extensive insight into SCD and GVHD variables. Although these are informative, they are time consuming. Less-specific but easier tools such as the Child Health Questionnaire or the Pediatric Quality of Life Inventory are more feasible for completion, allowing for comparisons across clinical trials.102,105,106 The PedsQL SCD module focuses specifically on the complications of the disease.107 A pediatric evaluation should include both child and parent reports in an age-dependent fashion.108 Other HRQOL measures include the Patient Reported Outcomes Measures Information System, a set of person-centered measures that evaluates physical, mental, and social health in adults and children.109 A comparison of HRQOL parameters with age-matched external controls helps identify the rate of change over time, where patients function as their own internal controls.

Assessing HRQOL before intervention, 6 months, 12 months, 2 years, and yearly thereafter if continued morbidity persists provides a longitudinal assessment that helps compare benefits from a patient perspective and informs decision-making.110 

Vaso-occlusive pain episodes are the hallmark of SCD, contributing to impaired HRQOL and predicting premature mortality. Vaso-occlusive pain episodes are, hence, an indication for allo-HCT/GT. Pain symptoms (except avascular necrosis or structural bone damage) resolve over time in most cases after the establishment of nonsickle erythropoiesis.6,7,49,111-114 Acute or chronic pain persistence even after successful curative interventions in a minority remains a vexing paradox. Graft failure, older age, history of recurrent pain crisis in the 2 years before intervention, and chronic opioid use are predictors of residual pain. Patients with pain episodes after HCT were more likely to have had chronic pain without SCD complications, opioid-induced hyperalgesia, central sensitization, or genetic predisposition.114,115 The persistent pain is of complex neurobiology, in which SCD pain complexes with prolonged opioid exposure and other mechanisms, especially with age progression.116 A higher rate of pain crises in older patients with more disease complications should inform pain-focused rehabilitation after intervention. Patients are typically not screened in detail for chronic pain before intervention; pain data are not captured by transplant registries such as the Center for International Blood and Marrow Transplant Research (CIBMTR).113 

Patient Reported Outcomes Measures Information System measures with pain-focused segments capture intensity, interference, mobility, and quality of pain depending on patient age. The maintenance of a pain diary records patient-experienced pain. Patients must be screened for chronic pain and pain-related domains before intervention and subsequently tracked.117 Pain domains can be tracked qualitatively and quantitatively serially with the PhenX Toolkit (phenxtoolkit.org). This will enhance multidisciplinary interventions for ameliorating chronic pain. Ideally, pain-mitigating interventions are best instituted before allo-HCT/GT and continued afterward until the pain pathway is subdued. This requires recognition of this entity by medical and family caregivers and the patient.

The absence of acute pain episodes over a consecutive 12-month period after intervention has been considered pain-free success in clinical trials.118 An issue with comparing pain parameters after allo-HCT/GT are the variations in defining and measuring pain that does not allow for intertrial comparison. Validated measures of pain burden are likely to elicit a more accurate picture but add to the burden of data collection. However, understanding pain domains and documenting measures of pain burden, including emergency room visits, hospitalizations, opioid use, pain dairies, and pain-related HRQOL questionnaires, are important objective ways of tracking pain (Table 5) and allow for intervention and counseling.

Table 5.

Longitudinal measurement of pain-related disease burden after HCT 

DomainMeasureAssessment
Health care utilization Emergency room visits/hospitalizations for pain Document yearly until return to normal 
Opioid use Monthly morphine equivalents  
Patient-reported pain intensity PROMIS 25 profile in children, PROMIS 29 profile in adults  
Patient-reported pain interference PROMIS 25 profile in children, PROMIS 29 profile in adults  
Identification of high-impact chronic pain Graded chronic pain scale revised  
DomainMeasureAssessment
Health care utilization Emergency room visits/hospitalizations for pain Document yearly until return to normal 
Opioid use Monthly morphine equivalents  
Patient-reported pain intensity PROMIS 25 profile in children, PROMIS 29 profile in adults  
Patient-reported pain interference PROMIS 25 profile in children, PROMIS 29 profile in adults  
Identification of high-impact chronic pain Graded chronic pain scale revised  

PROMIS 25, Patient Reported Outcomes Measurement System 25.

For further details on pain-related measurements.119 

Although it is ideal to have full donor chimerism, mixed chimerism after allo-HCT if stable can be adequate to control SCD manifestations and may even offset immunologic complications, such as GVHD. Mixed donor/recipient chimerism allows for reduced conditioning intensity and opens future possibilities for using nonchemoradiation-based agents such as depleting antibodies. Red blood cell modeling studies based on red cell survival as well as patient-derived mixed chimerism data have indicated that myeloid engraftment levels of 20% to 25% are necessary to achieve normal hemoglobin levels and eradicate hemolysis and pain symptoms.120,121 Similarly, tracking genetic edits (transduction efficiency, vector copy numbers, double-stranded DNA edits, and base edits) after GT has demonstrated that mixed engraftment with a threshold level of genetically modified cells (especially those with efficient gene correction) can correct anemia and control SCD symptoms, such as pain, adequately.

It is important to evaluate the stability and effect of donor myeloid chimerism or percent gene-modified cells and organ functions long term to determine the adequacy of reversal of vascular pathophysiology. These, combined with measures of hemoglobin S levels and hemolysis, determine the adequacy of engraftment. Studying more sensitive measures of hemolysis and targeted red cell engraftment add to the standard measures of chimerism determination.45,122-124 Rheologic assessments of red blood cells can help quantify the difference between red cells with sufficient antisickling hemoglobin and those with insufficient modification.125 Novel biomarkers may help predict engraftment.126 Increased early myeloid-derived suppressor cells at day 60 showed positive correlation with myeloid donor cell engraftment after haploidentical HSCT for SCD.127 A similar observation was made with immunoregulatory cytokines and chemokines, such as interleukin-17A, interleukin-10, and VEGF (vascular endothelial growth factor), in predicting engraftment/rejection. Table 6 lists tracking methods for assessing the adequacy of chimerism or the presence of gene-modified cells.

Table 6.

Recommended assessment of chimerism

ChimerismAssessmentInterval
Clinical STR analysis on bone marrow or sorted peripheral blood cells (myeloid and T cells) 1, 3, 6, 12, and 24 months 
Repeat if continued mixed chimerism 
Repeat in the event of return of abnormal hematologic/hemolysis parameters 
Autologous cell engraftment Percent of edited cells (gene editing) All GT patients should be tracked for at least 15 y by mandate 
Vector copy number (average of viral vector per stem cell)  
Hemoglobin analysis for Hb S level Every 3 months × 1 year 
Every 6 months in year 2 
Yearly × 5 years 
As needed subsequently 
Hemolysis parameters, including hemoglobin, absolute reticulocyte count, lactate dehydrogenase, and indirect bilirubin Every 3 months × 1 year 
Every 6 months in year 2 
Yearly × 5 years 
As needed subsequently 
Research Glycophorin A + CD71+ red cell precursor genomic analysis for engraftment128,129  
Functional biomarkers 
Red cell viscosity, RBC elongation index by ektacytometry, and RBC adhesion index 
Specialized cell subtypes, cytokines, and chemokines 
ChimerismAssessmentInterval
Clinical STR analysis on bone marrow or sorted peripheral blood cells (myeloid and T cells) 1, 3, 6, 12, and 24 months 
Repeat if continued mixed chimerism 
Repeat in the event of return of abnormal hematologic/hemolysis parameters 
Autologous cell engraftment Percent of edited cells (gene editing) All GT patients should be tracked for at least 15 y by mandate 
Vector copy number (average of viral vector per stem cell)  
Hemoglobin analysis for Hb S level Every 3 months × 1 year 
Every 6 months in year 2 
Yearly × 5 years 
As needed subsequently 
Hemolysis parameters, including hemoglobin, absolute reticulocyte count, lactate dehydrogenase, and indirect bilirubin Every 3 months × 1 year 
Every 6 months in year 2 
Yearly × 5 years 
As needed subsequently 
Research Glycophorin A + CD71+ red cell precursor genomic analysis for engraftment128,129  
Functional biomarkers 
Red cell viscosity, RBC elongation index by ektacytometry, and RBC adhesion index 
Specialized cell subtypes, cytokines, and chemokines 

Hb S, hemoglobin S; RBC, red blood cell; STR, short tandem repeat.

Two large studies have reported an increased (3.6× to 11× higher) prevalence of acute myeloid leukemia (AML) in individuals with SCD.130,131 However, with >27 years of follow-up, 6 patients with SCD were diagnosed with AML vs 1.67 expected cases controlled for age, race, ethnicity, and sex.131 Thus, the absolute risk of leukemia is low for the general population of individuals with SCD.

Myelodysplastic syndrome (MDS) and AML have been reported after allo-HCT/GT interventions.132 An increased risk of MDS and AML in adults with SCD after conditioning with alemtuzumab and low-dose radiation is described in the setting of graft failure and mixed chimerism.133 The incidence of AML was similarly increased in the first group of adults who received myeloablative busulfan and GT with Lovo-cel (Lovotibeglogene Autotemcel). This group received cells with a lower vector copy number and a lower dose of hematopoietic progenitor cells, and subsequent optimization of manufacture, intensity of myeloablation, and infusion of higher cell doses have resulted in successful engraftment with no malignant evolution to date.

The reasons for susceptibility to MDS and AML remain unclear. Four of 5 patients who developed MDS or AML after allo-HCT and graft failure had different pathogenic TP53 mutations, with variant allele frequencies (VAFs) ranging from 2.9% to 78.8% in the peripheral blood on next-generation sequencing.134 The same mutations were present with lower VAFs (0.06%-0.36%) before allo-HCT in 3 patients. No mutation was detected until 1.5 years after HCT in the fourth. The mutation was identified 4 years after HCT (VAF, 0.06%) and increased to 0.34% at 5 years and 78.8% at 5.5 years when AML was diagnosed. There were no TP53 mutations identified in 15 other patients (7 with graft failure), with no MDS or AML 4 to 12 years after HSCT.

The prevalence of somatic mutations identified with high sensitivity (VAF <1%) in patients with SCD is unknown. When whole-genome sequencing data were analyzed (VAF 5%-10%), there was no increased incidence noted.135 When whole-exome sequencing data were evaluated (VAF 2.5%), the prevalence of clonal hematopoiesis (CH) was higher in patients with SCD.136 Low-level CH may be precipitated by inflammation and erythropoietic stress.137 Exposure to genotoxic chemoradiation may promote further proliferation of autologous cells, leading to MDS or AML.132,138 These events underscore the need for tracking blood for CH before and after intervention to determine risk, with efforts ongoing. Sample retention before and after allo-HCT/GT to analyze retrospectively in the event of MDS or AML development is worth a consideration.

Successfully transitioning from pediatric to adult care is necessary after allo-HCT/GT. Transition involves a comprehensive approach to ensure sustained health and well-being after therapy and includes monitoring for GVHD and associated complications, infection prevention, and management of residual SCD-related complications. The kinetics of change in individual organs affected by SCD (improvement, stabilization, and progression) are unknown, especially after newer therapies. Tracking patients as described will be crucial for understanding the disease course and other effects/complications. Pediatric SCD/transplant teams will need to identify adult care providers who can continue to provide relevant aspects of care. Putting the onus of the follow-up on the young patient is likely to limit the success of the transition. It is also important that an adult SCD provider follows the patient long term to recognize disease-related events if any. This effort in individuals who do not have immediate pressing medical needs of established SCD will need additional effort to accomplish.

Coordination between pediatric and adult care providers, a transition document that provides relevant details, and follow-through on care necessities ensure a smooth transition. In addition, psychosocial care is vital. Young adults who are navigating significant life changes at the time of transition tend to neglect their health, especially after undergoing curative therapy with discernible change in the clinical course of their disease. Although clinically desired hematologic parameters are achieved, residual issues such as established vascular endothelial damage, chronic pain, and late effects of the intervention may continue and require specific medical, educational, and psychosocial adaptations during schooling or vocational decision-making. A well-structured transition plan that includes HCT-related follow-up,139 education on medication adherence, symptom monitoring, organ function tracking, and lifestyle adjustments can improve long-term outcomes and enhance quality of life after allo-HCT/GT (Table 7).

Table 7.

Long-term management considerations after allogeneic HCT and GT

Management areaSpecific needs
Transplant related Regular follow-ups for GVHD and infection screening 
Engraftment and chimerism monitoring 
SCD Annual eye assessments for retinopathy screening 
Ongoing assessment of organ function especially in areas of previous damage (kidney, lung, etc) 
Chronic pain management Assessment for any evidence of ongoing pain due to previous bone/muscular damage or caused by opioid-induced hyperalgesia 
Long-term pain plan discussion including goals of weaning off opioids or switching to buprenorphine-based pain management 
Psychosocial monitoring Evaluation for depression, anxiety, and posttraumatic stress disorder 
Counseling for emotional and psychological adjustment 
Social determinants of health assessment 
Neurocognitive monitoring Neurocognitive testing to assist people with entering school or workforce 
Referral for neurocognitive rehabilitation of needed 
Evaluation for any progression of previous infarcts or CNS-vascular damage 
Lifestyle adjustment Ensure individual understands the needing for continuous health care coverage 
Guidance on healthy diet and physical activity 
Education on navigating and understanding the adult health care systems 
Identification of other health providers (PCP and GYN) needed for the adult 
Management areaSpecific needs
Transplant related Regular follow-ups for GVHD and infection screening 
Engraftment and chimerism monitoring 
SCD Annual eye assessments for retinopathy screening 
Ongoing assessment of organ function especially in areas of previous damage (kidney, lung, etc) 
Chronic pain management Assessment for any evidence of ongoing pain due to previous bone/muscular damage or caused by opioid-induced hyperalgesia 
Long-term pain plan discussion including goals of weaning off opioids or switching to buprenorphine-based pain management 
Psychosocial monitoring Evaluation for depression, anxiety, and posttraumatic stress disorder 
Counseling for emotional and psychological adjustment 
Social determinants of health assessment 
Neurocognitive monitoring Neurocognitive testing to assist people with entering school or workforce 
Referral for neurocognitive rehabilitation of needed 
Evaluation for any progression of previous infarcts or CNS-vascular damage 
Lifestyle adjustment Ensure individual understands the needing for continuous health care coverage 
Guidance on healthy diet and physical activity 
Education on navigating and understanding the adult health care systems 
Identification of other health providers (PCP and GYN) needed for the adult 

CNS, central nervous system; GYN, Gynaecologist; PCP, Primary Care Physician.

Planned efforts at collecting measured outcome details are underway within specific research groups. Some recently developed networks work alongside standard measures collected by the CIBMTR after HCT or GT; for example, the Globin Research Network for Data and Discovery maintained registry that has been adopted by the National Alliance of Sickle Cell Centers to track intervention efficacy, engraftment, transduction assessments, and fertility outcomes,140 the Cooperative Assessment of Late Effects for SCD Curative Therapies evaluates long-term effects on the heart, lungs, and kidneys, Project Sickle Cure tracks outcomes on patients who received HCT from matched sibling donors. The Sickle Cell Clinical Research and Intervention Program141 and the Sickle Cell Disease Implementation Consortium142 evaluate longitudinal clinical, neurocognitive, geospatial, psychosocial, and health outcome data across patient lifetimes, assess transition to adulthood, and maintain specimen banks.

The expansion of HCT- and GT-derived hematopoietic manipulations in SCD bodes well for an increasing number of patients with SCD. The functional ability of these interventions will depend on the efficacy of the modality used, disease status at intervention, age, comorbidities, longevity of correction, and the side effects encountered. This makes it important to monitor outcomes uniformly between treatment modalities to determine the scale of advantage gained, delineate disadvantages to combat them better, and predict the duration over which to expect change. It also helps identify which treatment modality could serve individual patients better. This insight is applicable to medical personnel as well as patients and families and is critical for joint decision-making. Comparable outcome analyses maintained with registries can also help comparison with advancing conservative therapies, anticipating pitfalls to combat, and promote additional research effort. HCT and GT registries, such as the CIBMTR, and smaller collaborative efforts from institutions active in providing HCT/GT interventions focusing on in-depth monitoring and documentation can enhance knowledge regarding treatment efforts and provide comparison. A world with manageable SCD care with longevity that parallels the unaffected is work in progress, medically, socially, and economically.143 

Contribution: S.S. coordinated this invited review and wrote the manuscript; J.K., A.K., C.F., E.S., M.B., L.P., L.K., and A.A.K. wrote parts of the manuscript and edited the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Shalini Shenoy, Pediatric Hematology/Oncology, Washington University School of Medicine, 660 S Euclid Ave, Box 8116, St Louis, MO 63110; email: shalinishenoy@wustl.edu.

1.
Abraham
A
,
Cluster
A
,
Jacobsohn
D
, et al
.
Unrelated umbilical cord blood transplantation for sickle cell disease following reduced-intensity conditioning: results of a phase I trial
.
Biol Blood Marrow Transplant
.
2017
;
23
(
9
):
1587
-
1592
.
2.
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
.
3.
Bolanos-Meade
J
,
Fuchs
EJ
,
Luznik
L
, et al
.
HLA-haploidentical bone marrow transplantation with posttransplant cyclophosphamide expands the donor pool for patients with sickle cell disease
.
Blood
.
2012
;
120
(
22
):
4285
-
4291
.
4.
Kassim
AA
,
de la Fuente
J
,
Nur
E
, et al
.
An international learning collaborative phase 2 trial for haploidentical bone marrow transplant in sickle cell disease
.
Blood
.
2024
;
143
(
25
):
2654
-
2665
.
5.
Foell
J
,
Kleinschmidt
K
,
Jakob
M
,
Troeger
A
,
Corbacioglu
S
.
Alternative donor: αß/CD19 T-cell-depleted haploidentical hematopoietic stem cell transplantation for sickle cell disease
.
Hematol Oncol Stem Cell Ther
.
2020
;
13
(
2
):
98
-
105
.
6.
Frangoul
H
,
Locatelli
F
,
Sharma
A
, et al
.
Exagamglogene autotemcel for severe sickle cell disease
.
N Engl J Med
.
2024
;
390
(
18
):
1649
-
1662
.
7.
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
.
8.
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
.
9.
Kassim
AA
,
Pruthi
S
,
Day
M
, et al
.
Silent cerebral infarcts and cerebral aneurysms are prevalent in adults with sickle cell anemia
.
Blood
.
2016
;
127
(
16
):
2038
-
2040
.
10.
Adams
RJ
,
McKie
VC
,
Hsu
L
, et al
.
Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography
.
N Engl J Med
.
1998
;
339
(
1
):
5
-
11
.
11.
DeBaun
MR
,
Sarnaik
SA
,
Rodeghier
MJ
, et al
.
Associated risk factors for silent cerebral infarcts in sickle cell anemia: low baseline hemoglobin, sex, and relative high systolic blood pressure
.
Blood
.
2012
;
119
(
16
):
3684
-
3690
.
12.
DeBaun
MR
,
Armstrong
FD
,
McKinstry
RC
,
Ware
RE
,
Vichinsky
E
,
Kirkham
FJ
.
Silent cerebral infarcts: a review on a prevalent and progressive cause of neurologic injury in sickle cell anemia
.
Blood
.
2012
;
119
(
20
):
4587
-
4596
.
13.
DeBaun
MR
,
Gordon
M
,
McKinstry
RC
, et al
.
Controlled trial of transfusions for silent cerebral infarcts in sickle cell anemia
.
N Engl J Med
.
2014
;
371
(
8
):
699
-
710
.
14.
Fields
ME
,
Guilliams
KP
,
Ragan
DK
, et al
.
Regional oxygen extraction predicts border zone vulnerability to stroke in sickle cell disease
.
Neurology
.
2018
;
90
(
13
):
e1134
-
e1142
.
15.
Jones
RS
,
Donahue
MJ
,
Davis
LT
, et al
.
Silent infarction in sickle cell disease is associated with brain volume loss in excess of infarct volume
.
Front Neurol
.
2023
;
14
:
1112865
.
16.
Wang
Y
,
Fellah
S
,
Fields
ME
, et al
.
Cerebral oxygen metabolic stress, microstructural injury, and infarction in adults with sickle cell disease
.
Neurology
.
2021
;
97
(
9
):
e902
-
e912
.
17.
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
.
18.
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
.
19.
Hulbert
ML
,
Fields
ME
,
Guilliams
KP
, et al
.
Normalization of cerebral hemodynamics after hematopoietic stem cell transplant in children with sickle cell disease
.
Blood
.
2023
;
141
(
4
):
335
-
344
.
20.
Aumann
MA
,
Richerson
W
,
Song
AK
, et al
.
Cerebral hemodynamic changes after haploidentical hematopoietic stem cell transplant in adults with sickle cell disease
.
Blood Adv
.
2024
;
8
(
3
):
608
-
619
.
21.
Afzali-Hashemi
L
,
Dovern
E
,
Baas
KPA
, et al
.
Cerebral hemodynamics and oxygenation in adult patients with sickle cell disease after stem cell transplantation
.
Am J Hematol
.
2024
;
99
(
2
):
163
-
171
.
22.
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
.
23.
Green
NS
,
Bhatia
M
,
Griffith
EY
, et al
.
Enhanced long-term brain magnetic resonance imaging evaluation of children with sickle cell disease after hematopoietic cell transplantation
.
Biol Blood Marrow Transplant
.
2017
;
23
(
4
):
670
-
676
.
24.
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
(
7
):
2749
-
2756
.
25.
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
.
26.
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
.
27.
Stenger
E
,
Xiang
Y
,
Wetzel
M
, et al
.
Long-term organ function after HCT for SCD: a report from the sickle cell transplant advocacy and research alliance
.
Transplant Cell Ther
.
2023
;
29
(
1
):
47.e1
-
47.e10
.
28.
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
.
29.
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
.
30.
Braniecki
S
,
Vichinsky
E
,
Walters
MC
, et al
.
Neurocognitive outcome in children with sickle cell disease after myeloimmunoablative conditioning and haploidentical hematopoietic stem cell transplantation: a non-randomized clinical trial
.
Front Neurol
.
2024
;
15
:
1263373
.
31.
Carlson
EJ
,
Al Ghriwati
N
,
Wolters
P
, et al
.
Longitudinal neurocognitive effects of nonmyeloablative hematopoietic stem cell transplant among older adolescents and adults with sickle cell disease: a description and comparison with sibling donors
.
Neuropsychol Rehabil
.
2024
;
34
(
7
):
899
-
918
.
32.
MacLean
JE
,
Atenafu
E
,
Kirby-Allen
M
, et al
.
Longitudinal decline in lung volume in a population of children with sickle cell disease
.
Am J Respir Crit Care Med
.
2008
;
178
(
10
):
1055
-
1059
.
33.
Hamideh
D
,
Alvarez
O
.
Sickle cell disease related mortality in the United States (1999-2009)
.
Pediatr Blood Cancer
.
2013
;
60
(
9
):
1482
-
1486
.
34.
Payne
AB
,
Mehal
JM
,
Chapman
C
, et al
.
Trends in sickle cell disease-related mortality in the United States, 1979 to 2017
.
Ann Emerg Med
.
2020
;
76
(
3S
):
S28
-
S36
.
35.
Gladwin
MT
,
Barst
RJ
,
Gibbs
JSR
, et al
.
Risk factors for death in 632 patients with sickle cell disease in the United States and United Kingdom
.
PLoS One
.
2014
;
9
(
7
):
e99489
.
36.
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
.
37.
Nouraie
M
,
Darbari
DS
,
Rana
S
, et al
.
Tricuspid regurgitation velocity and other biomarkers of mortality in children, adolescents and young adults with sickle cell disease in the United States: the PUSH study
.
Am J Hematol
.
2020
;
95
(
7
):
766
-
774
.
38.
Sachdev
V
,
Tian
X
,
Gu
Y
, et al
.
A phenotypic risk score for predicting mortality in sickle cell disease
.
Br J Haematol
.
2021
;
192
(
5
):
932
-
941
.
39.
Morin
CE
,
Sharma
A
,
Selukar
S
, et al
.
Diffuse myocardial fibrosis occurs in young patients with sickle cell anemia despite early disease-modifying therapy
.
Blood
.
2023
;
141
(
11
):
1358
-
1362
.
40.
Ruhl
AP
,
Shalhoub
R
,
Jeffries
N
, et al
.
Pulmonary function after non-myeloablative hematopoietic cell transplant for sickle cell disease
.
Ann Am Thorac Soc
.
2024
;
21
(
10
):
1398
-
1406
.
41.
Dallas
MH
,
Triplett
B
,
Shook
DR
, et al
.
Long-term outcome and evaluation of organ function in pediatric patients undergoing haploidentical and matched related hematopoietic cell transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2013
;
19
(
5
):
820
-
830
.
42.
Sachdev
V
,
Limerick
E
,
Nguyen
ML
, et al
.
Cardiac effects 2 years after successful non-myeloablative human leukocyte antigen-matched related donor hematopoietic cell transplants in sickle cell disease
.
Am J Hematol
.
2023
;
98
(
8
):
E219
-
E221
.
43.
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
.
44.
Walters
MC
,
Hardy
K
,
Edwards
S
, et al
.
Pulmonary, gonadal, and central nervous system status after bone marrow transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2010
;
16
(
2
):
263
-
272
.
45.
Bhatia
M
,
Jin
Z
,
Baker
C
, et al
.
Reduced toxicity, myeloablative conditioning with BU, fludarabine, alemtuzumab and SCT from sibling donors in children with sickle cell disease
.
Bone Marrow Transplant
.
2014
;
49
(
7
):
913
-
920
.
46.
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
.
47.
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
.
48.
Monagel
DA
,
Guilcher
GMT
,
Nettel-Aguirre
A
,
Bendiak
GN
.
Pulmonary function in children and adolescents with sickle cell disease after nonmyeloablative hematopoietic cell transplantation
.
Pediatr Blood Cancer
.
2022
;
69
(
10
):
e29927
.
49.
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
-
56
.
50.
Covi
S
,
Ravindranath
Y
,
Farooqi
A
,
Savasan
S
,
Chu
R
,
Aggarwal
S
.
Changes in Bi-ventricular function after hematopoietic stem cell transplant as assessed by speckle tracking echocardiography
.
Pediatr Cardiol
.
2018
;
39
(
2
):
365
-
374
.
51.
Dovern
E
,
Aydin
M
,
DeBaun
MR
,
Alizade
K
,
Biemond
BJ
,
Nur
E
.
Effect of allogeneic hematopoietic stem cell transplantation on sickle cell disease-related organ complications: a systematic review and meta-analysis
.
Am J Hematol
.
2024
;
99
(
6
):
1129
-
1141
.
52.
Sharma
A
,
Selukar
S
,
Bi
Y
, et al
.
Impact of hematopoietic cell transplantation on myocardial fibrosis in young patients with sickle cell disease
.
Blood
.
2024
;
144
(
6
):
672
-
675
.
53.
Limerick
E
,
Shmukler
J
,
Sirajuddin
A
, et al
.
Improvement in cardiac morphology demonstrated by cardiac magnetic resonance imaging and echocardiography after haploidentical hematopoietic cell transplantation in adults with sickle cell disease
.
Transplant Cell Ther
.
2024
;
30
(
2
):
231.e1
-
231.e9
.
54.
Amarapurkar
P
,
Roberts
L
,
Navarrete
J
,
El Rassi
F
.
Sickle cell disease and kidney
.
Adv Chronic Kidney Dis
.
2022
;
29
(
2
):
141
-
148.e1
.
55.
Hingorani
SR
,
Seidel
K
,
Lindner
A
,
Aneja
T
,
Schoch
G
,
McDonald
G
.
Albuminuria in hematopoietic cell transplantation patients: prevalence, clinical associations, and impact on survival
.
Biol Blood Marrow Transplant
.
2008
;
14
(
12
):
1365
-
1372
.
56.
Fitzhugh
CD
,
Volanakis
EJ
,
Idassi
O
,
Duberman
JA
,
DeBaun
MR
,
Friedman
DL
.
Long-term health effects of curative therapies on heart, lungs, and kidneys for individuals with sickle cell disease compared to those with hematologic malignancies
.
J Clin Med
.
2022
;
11
(
11
):
3118
.
57.
Hingorani
S
,
Finn
LS
,
Pao
E
, et al
.
Urinary elafin and kidney injury in hematopoietic cell transplant recipients
.
Clin J Am Soc Nephrol
.
2015
;
10
(
1
):
12
-
20
.
58.
Laurentino
MR
,
Parente Filho
SLA
,
Parente
LLC
,
da Silva Júnior
GB
,
Daher
EDF
,
Lemes
RPG
.
Non-invasive urinary biomarkers of renal function in sickle cell disease: an overview
.
Ann Hematol
.
2019
;
98
(
12
):
2653
-
2660
.
59.
Nickel
RS
,
Maher
JY
,
Hsieh
MH
,
Davis
MF
,
Hsieh
MM
,
Pecker
LH
.
Fertility after curative therapy for sickle cell disease: a comprehensive review to guide care
.
J Clin Med
.
2022
;
11
(
9
):
2318
.
60.
Pecker
LH
,
Kuo
KHM
.
Go the distance: reproductive health care for people with sickle cell disease
.
Hematol Oncol Clin North Am
.
2022
;
36
(
6
):
1255
-
1270
.
61.
Pecker
LH
,
Maher
JY
,
Law
JY
,
Beach
MC
,
Lanzkron
S
,
Christianson
MS
.
Risks associated with fertility preservation for women with sickle cell anemia
.
Fertil Steril
.
2018
;
110
(
4
):
720
-
731
.
62.
Missontsa
MM
,
Bernaudin
F
,
Fortin
A
, et al
.
Ovarian tissue cryopreservation for fertility preservation before hematopoietic stem cell transplantation in patients with sickle cell disease: safety, ovarian function follow-up, and results of ovarian tissue transplantation
.
J Assist Reprod Genet
.
2024
;
41
(
4
):
1027
-
1034
.
63.
Roux
C
,
Amiot
C
,
Agnani
G
,
Aubard
Y
,
Rohrlich
PS
,
Piver
P
.
Live birth after ovarian tissue autograft in a patient with sickle cell disease treated by allogeneic bone marrow transplantation
.
Fertil Steril
.
2010
;
93
(
7
):
2413.e15
. 2413.e9.
64.
Early
ML
,
Strodel
RJ
,
Lake
IV
, et al
.
Acceptable, hopeful, and useful: development and mixed-method evaluation of an educational tool about reproductive options for people with sickle cell disease or trait
.
J Assist Reprod Genet
.
2022
;
39
(
1
):
183
-
193
.
65.
Booth
C
,
Inusa
B
,
Obaro
SK
.
Infection in sickle cell disease: a review
.
Int J Infect Dis
.
2010
;
14
(
1
):
e2
-
e12
.
66.
William
BM
,
Thawani
N
,
Sae-Tia
S
,
Corazza
GR
.
Hyposplenism: a comprehensive review. Part II: clinical manifestations, diagnosis, and management
.
Hematology
.
2007
;
12
(
2
):
89
-
98
.
67.
Larcher
VF
,
Wyke
RJ
,
Davis
LR
,
Stroud
CE
,
Williams
R
.
Defective yeast opsonisation and functional deficiency of complement in sickle cell disease
.
Arch Dis Child
.
1982
;
57
(
5
):
343
-
346
.
68.
Prasad
AS
,
Beck
FW
,
Kaplan
J
, et al
.
Effect of zinc supplementation on incidence of infections and hospital admissions in sickle cell disease (SCD)
.
Am J Hematol
.
1999
;
61
(
3
):
194
-
202
.
69.
Tamouza
R
,
Neonato
MG
,
Busson
M
, et al
.
Infectious complications in sickle cell disease are influenced by HLA class II alleles
.
Hum Immunol
.
2002
;
63
(
3
):
194
-
199
.
70.
Adewoye
AH
,
Nolan
VG
,
Ma
Q
, et al
.
Association of polymorphisms of IGF1R and genes in the transforming growth factor- beta /bone morphogenetic protein pathway with bacteremia in sickle cell anemia
.
Clin Infect Dis
.
2006
;
43
(
5
):
593
-
598
.
71.
Zarrouk
V
,
Habibi
A
,
Zahar
JR
, et al
.
Bloodstream infection in adults with sickle cell disease: association with venous catheters, Staphylococcus aureus, and bone-joint infections
.
Medicine (Baltim)
.
2006
;
85
(
1
):
43
-
48
.
72.
Velardi
E
,
Clave
E
,
Arruda
LCM
,
Benini
F
,
Locatelli
F
,
Toubert
A
.
The role of the thymus in allogeneic bone marrow transplantation and the recovery of the peripheral T-cell compartment
.
Semin Immunopathol
.
2021
;
43
(
1
):
101
-
117
.
73.
de Koning
C
,
Nierkens
S
,
Boelens
JJ
.
Strategies before, during, and after hematopoietic cell transplantation to improve T-cell immune reconstitution
.
Blood
.
2016
;
128
(
23
):
2607
-
2615
.
74.
Velardi
E
,
Tsai
JJ
,
van den Brink
MRM
.
T cell regeneration after immunological injury
.
Nat Rev Immunol
.
2021
;
21
(
5
):
277
-
291
.
75.
Patel
DA
,
Dhedin
N
,
Chen
H
, et al
.
Early viral reactivation despite excellent immune reconstitution following haploidentical Bone marrow transplant with post-transplant cytoxan for sickle cell disease
.
Transpl Infect Dis
.
2020
;
22
(
1
):
e13222
.
76.
Hsieh
MM
,
Kang
EM
,
Fitzhugh
CD
, et al
.
Allogeneic hematopoietic stem-cell transplantation for sickle cell disease
.
N Engl J Med
.
2009
;
361
(
24
):
2309
-
2317
.
77.
Cairo
MS
,
Talano
JA
,
Moore
TB
, et al
.
Familial haploidentical stem cell transplant in children and adolescents with high-risk sickle cell disease: a phase 2 clinical trial
.
JAMA Pediatr
.
2020
;
174
(
2
):
195
-
197
.
78.
van den Brink
MR
,
Velardi
E
,
Perales
MA
.
Immune reconstitution following stem cell transplantation
.
Hematology Am Soc Hematol Educ Program
.
2015
;
2015
:
215
-
219
.
79.
Storek
J
,
Geddes
M
,
Khan
F
, et al
.
Reconstitution of the immune system after hematopoietic stem cell transplantation in humans
.
Semin Immunopathol
.
2008
;
30
(
4
):
425
-
437
.
80.
Mackall
CL
.
T-cell immunodeficiency following cytotoxic antineoplastic therapy: a review
.
Stem Cell
.
2000
;
18
(
1
):
10
-
18
.
81.
Fagnoni
FF
,
Lozza
L
,
Zibera
C
, et al
.
T-cell dynamics after high-dose chemotherapy in adults: elucidation of the elusive CD8+ subset reveals multiple homeostatic T-cell compartments with distinct implications for immune competence
.
Immunology
.
2002
;
106
(
1
):
27
-
37
.
82.
Storek
J
,
Wells
D
,
Dawson
MA
,
Storer
B
,
Maloney
DG
.
Factors influencing B lymphopoiesis after allogeneic hematopoietic cell transplantation
.
Blood
.
2001
;
98
(
2
):
489
-
491
.
83.
Kanakry
CG
,
Fuchs
EJ
,
Luznik
L
.
Modern approaches to HLA-haploidentical blood or marrow transplantation
.
Nat Rev Clin Oncol
.
2016
;
13
(
2
):
132
.
84.
Luo
XH
,
Chang
YJ
,
Huang
XJ
.
Improving cytomegalovirus-specific T cell reconstitution after haploidentical stem cell transplantation
.
J Immunol Res
.
2014
;
2014
:
631951
.
85.
Crocchiolo
R
,
Bramanti
S
,
Vai
A
, et al
.
Infections after T-replete haploidentical transplantation and high-dose cyclophosphamide as graft-versus-host disease prophylaxis
.
Transpl Infect Dis
.
2015
;
17
(
2
):
242
-
249
.
86.
Chang
YJ
,
Zhao
XY
,
Huang
XJ
.
Immune reconstitution after haploidentical hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2014
;
20
(
4
):
440
-
449
.
87.
Le Blanc
K
,
Barrett
AJ
,
Schaffer
M
, et al
.
Lymphocyte recovery is a major determinant of outcome after matched unrelated myeloablative transplantation for myelogenous malignancies
.
Biol Blood Marrow Transplant
.
2009
;
15
(
9
):
1108
-
1115
.
88.
Kim
HT
,
Armand
P
,
Frederick
D
, et al
.
Absolute lymphocyte count recovery after allogeneic hematopoietic stem cell transplantation predicts clinical outcome
.
Biol Blood Marrow Transplant
.
2015
;
21
(
5
):
873
-
880
.
89.
Kim
DH
,
Sohn
SK
,
Won
DI
,
Lee
NY
,
Suh
JS
,
Lee
KB
.
Rapid helper T-cell recovery above 200 × 106/L at 3 months correlates to successful transplant outcomes after allogeneic stem cell transplantation
.
Bone Marrow Transplant
.
2006
;
37
(
12
):
1119
-
1128
.
90.
Kanda
J
,
Chiou
LW
,
Szabolcs
P
, et al
.
Immune recovery in adult patients after myeloablative dual umbilical cord blood, matched sibling, and matched unrelated donor hematopoietic cell transplantation
.
Biol Blood Marrow Transplant
.
2012
;
18
(
11
):
1664
-
1676.e1
.
91.
Bhatt
ST
,
Bednarski
JJ
,
Berg
J
, et al
.
Immune reconstitution and infection patterns after early alemtuzumab and reduced intensity transplantation for nonmalignant disorders in pediatric patients
.
Biol Blood Marrow Transplant
.
2019
;
25
(
3
):
556
-
561
.
92.
Small
TN
,
Papadopoulos
EB
,
Boulad
F
, et al
.
Comparison of immune reconstitution after unrelated and related T-cell-depleted bone marrow transplantation: effect of patient age and donor leukocyte infusions
.
Blood
.
1999
;
93
(
2
):
467
-
480
.
93.
Lewin
SR
,
Heller
G
,
Zhang
L
, et al
.
Direct evidence for new T-cell generation by patients after either T-cell-depleted or unmodified allogeneic hematopoietic stem cell transplantations
.
Blood
.
2002
;
100
(
6
):
2235
-
2242
.
94.
Krell
PF
,
Reuther
S
,
Fischer
U
, et al
.
Next-generation-sequencing-spectratyping reveals public T-cell receptor repertoires in pediatric very severe aplastic anemia and identifies a β chain CDR3 sequence associated with hepatitis-induced pathogenesis
.
Haematologica
.
2013
;
98
(
9
):
1388
-
1396
.
95.
Mackall
CL
,
Fleisher
TA
,
Brown
MR
, et al
.
Lymphocyte depletion during treatment with intensive chemotherapy for cancer
.
Blood
.
1994
;
84
(
7
):
2221
-
2228
.
96.
John
TD
,
Maron
G
,
Abraham
A
, et al
.
Strategic infection prevention after genetically modified hematopoietic stem cell therapies: recommendations from the International Society for Cell & Gene Therapy Stem Cell Engineering Committee
.
Cytotherapy
.
2024
;
26
(
7
):
660
-
671
.
97.
Ljungman
P
,
Engelhard
D
,
de la Cámara
R
, et al
.
Vaccination of stem cell transplant recipients: recommendations of the Infectious Diseases Working Party of the EBMT
.
Bone Marrow Transplant
.
2005
;
35
(
8
):
737
-
746
.
98.
Stokoe
M
,
Zwicker
HM
,
Forbes
C
, et al
.
Health related quality of life in children with sickle cell disease: a systematic review and meta-analysis
.
Blood Rev
.
2022
;
56
:
100982
.
99.
Panepinto
JA
,
Bonner
M
.
Health-related quality of life in sickle cell disease: past, present, and future
.
Pediatr Blood Cancer
.
2012
;
59
(
2
):
377
-
385
.
100.
Panepinto
JA
,
Paul Scott
J
,
Badaki-Makun
O
, et al
.
Determining the longitudinal validity and meaningful differences in HRQL of the PedsQL Sickle Cell Disease Module
.
Health Qual Life Outcomes
.
2017
;
15
(
1
):
124
.
101.
Shenoy
S
,
Angelucci
E
,
Arnold
SD
, et al
.
Current results and future research priorities in late effects after hematopoietic stem cell transplantation for children with sickle cell disease and thalassemia: a consensus statement from the Second Pediatric Blood and Marrow Transplant Consortium International Conference on Late Effects after Pediatric Hematopoietic Stem Cell Transplantation
.
Biol Blood Marrow Transplant
.
2017
;
23
(
4
):
552
-
561
.
102.
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
.
103.
Kelly
MJ
,
Pennarola
BW
,
Rodday
AM
,
Parsons
SK
;
Journeys to Recovery Study, HSCT-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
.
104.
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
.
105.
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
.
106.
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
.
107.
Panepinto
JA
,
Torres
S
,
Bendo
CB
, et al
.
PedsQL sickle cell disease module: feasibility, reliability, and validity
.
Pediatr Blood Cancer
.
2013
;
60
(
8
):
1338
-
1344
.
108.
Alberts
NM
,
Kang
G
,
Li
C
, et al
.
Pain in youth with sickle cell disease: a report from the Sickle Cell Clinical Research and Intervention Program
.
Clin J Pain
.
2021
;
37
(
1
):
43
-
50
.
109.
Irwin
DE
,
Gross
HE
,
Stucky
BD
, et al
.
Development of six PROMIS pediatrics proxy-report item banks
.
Health Qual Life Outcomes
.
2012
;
10
:
22
.
110.
Badawy
SM
,
Beg
U
,
Liem
RI
,
Chaudhury
S
,
Thompson
AA
.
A systematic review of quality of life in sickle cell disease and thalassemia after stem cell transplant or gene therapy
.
Blood Adv
.
2021
;
5
(
2
):
570
-
583
.
111.
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
.
112.
Leonard
A
,
Furstenau
D
,
Abraham
A
, et al
.
Reduction in vaso-occlusive events following stem cell transplantation in patients with sickle cell disease
.
Blood Adv
.
2023
;
7
(
2
):
227
-
234
.
113.
Krishnamurti
L
,
Liang
J
,
He
Z
, et al
.
Incidence and risk factors of pain crisis after hematopoietic cell transplantation for sickle cell disease
.
Blood Adv
.
2024
;
8
(
8
):
1908
-
1919
.
114.
Campbell
CM
,
Moscou-Jackson
G
,
Carroll
CP
, et al
.
An evaluation of central sensitization in patients with sickle cell disease
.
J Pain
.
2016
;
17
(
5
):
617
-
627
.
115.
Carroll
CP
,
Lanzkron
S
,
Haywood
C
, et al
.
Chronic opioid therapy and central sensitization in sickle cell disease
.
Am J Prev Med
.
2016
;
51
(
1 suppl 1
):
S69
-
S77
.
116.
Darbari
DS
,
Ballas
SK
,
Clauw
DJ
.
Thinking beyond sickling to better understand pain in sickle cell disease
.
Eur J Haematol
.
2014
;
93
(
2
):
89
-
95
.
117.
Jagtiani
A
,
Chou
E
,
Gillespie
SE
, et al
.
High-impact chronic pain in sickle cell disease: insights from the pain in sickle cell epidemiology study (PiSCES)
.
Pain
.
2024
;
165
(
10
):
2364
-
2369
.
118.
Locatelli
F
,
Corbacioglu
S
,
Hobbs
W
,
Frangoul
H
,
Walters
MC
.
Defining curative endpoints for sickle cell disease in the era of gene therapy and gene editing
.
Am J Hematol
.
2024
;
99
(
3
):
430
-
438
.
119.
Von Korff
M
,
DeBar
LL
,
Krebs
EE
,
Kerns
RD
,
Deyo
RA
,
Keefe
FJ
.
Graded chronic pain scale revised: mild, bothersome, and high-impact chronic pain
.
Pain
.
2020
;
161
(
3
):
651
-
661
.
120.
Abraham
A
,
Hsieh
M
,
Eapen
M
, et al
.
Relationship between mixed donor-recipient chimerism and disease recurrence after hematopoietic cell transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2017
;
23
(
12
):
2178
-
2183
.
121.
Fitzhugh
CD
,
Cordes
S
,
Taylor
T
, et al
.
At least 20% donor myeloid chimerism is necessary to reverse the sickle phenotype after allogeneic HSCT
.
Blood
.
2017
;
130
(
17
):
1946
-
1948
.
122.
Lewis
J
,
Greenway
SC
,
Khan
F
,
Singh
G
,
Bhatia
M
,
Guilcher
GMT
.
Assessment of donor cell engraftment after hematopoietic stem cell transplantation for sickle cell disease: a review of current and future methods
.
Am J Hematol
.
2022
;
97
(
10
):
1359
-
1371
.
123.
Wu
CJ
,
Hochberg
EP
,
Rogers
SA
, et al
.
Molecular assessment of erythroid lineage chimerism following nonmyeloablative allogeneic stem cell transplantation
.
Exp Hematol
.
2003
;
31
(
10
):
924
-
933
.
124.
Lu
M
,
Kanne
CK
,
Reddington
RC
,
Lezzar
DL
,
Sheehan
VA
,
Shevkoplyas
SS
.
Concurrent assessment of deformability and adhesiveness of sickle red blood cells by measuring perfusion of an adhesive artificial microvascular network
.
Front Physiol
.
2021
;
12
:
633080
.
125.
Saah
E
,
Fadaei
P
,
Gurkan
UA
,
Sheehan
V
.
Sickle cell disease pathophysiology and related molecular and biophysical biomarkers
.
Hematol Oncol Clin North Am
.
2022
;
36
(
6
):
1077
-
1095
.
126.
Shaikh
A
,
Olkhanud
PB
,
Gangaplara
A
, et al
.
Thrombospondin-1, platelet factor 4, and galectin-1 are associated with engraftment in patients with sickle cell disease who underwent haploidentical hematopoietic stem cell transplantation
.
Transplant Cell Ther
.
2022
;
28
(
5
):
249.e1
-
249.e13
.
127.
Bhat
DK
,
Olkhanud
PB
,
Gangaplara
A
, et al
.
Early myeloid derived suppressor cells (eMDSCs) are associated with high donor myeloid chimerism following haploidentical HSCT for sickle cell disease
.
Front Immunol
.
2021
;
12
:
757279
.
128.
Lewis
J
,
Greenway
SC
,
Khan
F
,
Singh
G
,
Bhatia
M
,
Guilcher
GMT
.
Assessment of donor cell engraftment after hematopoietic stem cell transplantation for sickle cell disease: A review of current and future methods
.
Am J Hematol
.
2022
;
97
(
10
):
1359
-
1371
.
129.
Wu
C.J.
,
Krishnamurti
L.
,
Kutok
J.L.
, et al
.
Evidence for ineffective erythropoiesis in severe sickle cell disease
.
Blood
.
2005
;
106
(
10
):
3639
-
3645
.
130.
Seminog
OO
,
Ogunlaja
OI
,
Yeates
D
,
Goldacre
MJ
.
Risk of individual malignant neoplasms in patients with sickle cell disease: English national record linkage study
.
J R Soc Med
.
2016
;
109
(
8
):
303
-
309
.
131.
Brunson
A
,
Keegan
THM
,
Bang
H
,
Mahajan
A
,
Paulukonis
S
,
Wun
T
.
Increased risk of leukemia among sickle cell disease patients in California
.
Blood
.
2017
;
130
(
13
):
1597
-
1599
.
132.
Fitzhugh
CD
.
Knowledge to date on secondary malignancy following hematopoietic cell transplantation for sickle cell disease
.
Hematology Am Soc Hematol Educ Program
.
2022
;
2022
(
1
):
266
-
271
.
133.
Lawal
RA
,
Mukherjee
D
,
Limerick
EM
, et al
.
Increased incidence of hematologic malignancies in SCD after HCT in adults with graft failure and mixed chimerism
.
Blood
.
2022
;
140
(
23
):
2514
-
2518
.
134.
Mukherjee
D
,
Lawal
RA
,
Fitzhugh
CD
,
Hourigan
CS
,
Dillon
LW
.
TP53 mutation screening for patients at risk of myeloid malignancy
.
Leukemia
.
2024
;
38
(
7
):
1604
-
1608
.
135.
Liggett
LA
,
Cato
LD
,
Weinstock
JS
, et al
.
Clonal hematopoiesis in sickle cell disease
.
J Clin Investig
.
2022
;
132
(
4
):
e156060
.
136.
Pincez
T
,
Lee
SSK
,
Ilboudo
Y
, et al
.
Clonal hematopoiesis in sickle cell disease
.
Blood
.
2021
;
138
(
21
):
2148
-
2152
.
137.
Gondek
LP
,
Sheehan
VA
,
Fitzhugh
CD
.
Clonal hematopoiesis and the risk of hematologic malignancies after curative therapies for sickle cell disease
.
J Clin Med
.
2022
;
11
(
11
):
3160
.
138.
Jones
RJ
,
DeBaun
MR
.
Leukemia after gene therapy for sickle cell disease: insertional mutagenesis, busulfan, both, or neither
.
Blood
.
2021
;
138
(
11
):
942
-
947
.
139.
Bhatia
S
,
Armenian
SH
,
Landier
W
.
How I monitor long-term and late effects after blood or marrow transplantation
.
Blood
.
2017
;
130
(
11
):
1302
-
1314
.
140.
Lanzkron
S
,
Manwani
D
,
Desai
P
,
Kanter
J
,
Little
J
.
GRNDaD: big data and sickle cell disease
.
Blood Adv
.
2022
;
6
(
3
):
1088
.
141.
Hankins
JS
,
Estepp
JH
,
Hodges
JR
, et al
.
Sickle Cell Clinical Research and Intervention Program (SCCRIP): a lifespan cohort study for sickle cell disease progression from the pediatric stage into adulthood
.
Pediatr Blood Cancer
.
2018
;
65
(
9
):
e27228
.
142.
DiMartino
LD
,
Baumann
AA
,
Hsu
LL
, et al
.
The Sickle Cell Disease Implementation Consortium: translating evidence-based guidelines into practice for sickle cell disease
.
Am J Hematol
.
2018
;
93
(
12
):
E391
-
e395
.
143.
Sharma
A
,
John
TD
.
Dismantling cost and infrastructure barriers to equitable access to gene therapies for sickle cell disease
.
Lancet Haematol
.
2024
;
11
(
8
):
e556
-
e559
.

Author notes

This article contains follow-up guidelines and has no data to access/share.

The full-text version of this article contains a data supplement.

Supplemental data