Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Mar 29.
Published in final edited form as: Drugs Aging. 2023 Feb 28;40(4):317–334. doi: 10.1007/s40266-023-01014-8

Management of Older Adults with Sickle Cell Disease: Considerations for current and emerging therapies

Charity I Oyedeji 1,2,3, Kimberly L Hodulik 1,4, Marilyn J Telen 1, John J Strouse 1,2,3,5
PMCID: PMC10979738  NIHMSID: NIHMS1965673  PMID: 36853587

Abstract

People with sickle cell disease (SCD) are living longer than ever before, with a median survival increasing from age 14 years in 1973, beyond age 40 in the 1990s, and as high as 61 years in recent cohorts from academic centers. Improvements in survival have been attributed to initiatives such as newborn screening, penicillin prophylaxis, vaccination against encapsulated organisms, better detection and treatment of splenic sequestration, and improved transfusion support. There are an estimated 100,000 people living with SCD in the United States and millions of people with SCD globally. Given that the number of older adults with SCD will likely continue to increase as survival improves, better evidence on how to manage this population is needed. When managing older adults with SCD (defined herein as age ≥ 40 years), healthcare providers should consider the potential pitfalls of extrapolating evidence from existing studies on current and emerging therapies that have typically been conducted with participants at mean ages far below 40. Older adults with SCD have historically had little to no representation in clinical trials; therefore, more guidance is needed on how to use current and emerging therapies in this population. This article summarizes the available evidence for managing older adults with SCD and discusses potential challenges to using approved and emerging drugs in this population.

1. Introduction

Sickle cell disease (SCD) is the most common severe inherited blood disorder in the United States (U.S.). In SCD, a point mutation in the β globin gene causes replacement of glutamic acid by valine at the 6th position of the β globin chain and leads to polymerization of deoxygenated hemoglobin.1 Polymerization of hemoglobin decreases red blood cells (RBCs) deformability and causes RBCs to become sickle-shaped and rigid, this contributes to occlusion of small vessels.2 Multiple other RBC changes, including dehydration, oxidative damage to the cell membrane and contents, and exposure of phosphatidylserine at the cell surface also promote abnormal RBC circulatory behavior and vaso-occlusion. Vaso-occlusion further exacerbates hemoglobin deoxygenation and hemolysis while also causing tissue hypoxia and painful episodes, which are the hallmarks of SCD. SCD causes organ dysfunction by several mechanisms, including recurrent episodes of sickling and vaso-occlusion, inflammation, hemolysis, and increased cell adhesion.3 There is significant phenotypic variability in complications, even among individuals with the same β globin mutation. Individuals may experience retinopathy, cutaneous ulceration, avascular necrosis, infections from encapsulated organisms, acute and chronic cardiopulmonary complications, kidney injury, strokes, and premature death.1 People with sickle cell anemia [homozygous for hemoglobin S (HbSS) or with hemoglobin SBeta0-thalassemia (HbSβ0)], have more hemolysis, are more anemic, and have a shorter life expectancy compared to compound heterozygotes with hemoglobin SC (HbSC) or SBeta+-thalassemia (HbSβ+).3,4

1.1. Comorbidities in the Older Adult with SCD

There are several similarities between conditions acquired by middle age in adults with SCD and in non-SCD geriatric populations (Figure 1).5 Both populations have inflammation, progressive anemia, joint damage, an increased prevalence of vision and hearing loss, and increased risk of hematologic malignancies.5 More data are needed to better distinguish sickle cell-related versus age-related changes that occur in older adults with SCD.

Fig. 1.

Fig. 1

Venn diagram describing characteristics and complications at the intersection of sickle cell disease and geriatrics

a. Individuals with SCD have a shorter life expectancy 4,120, vaso-occlusive pain crises 69,121, avascular necrosis of the bone 122, asplenic/splenectomy 69, increased pulmonary hypertension 123, leg ulcers 69, lower risk of solid tumors 53, more likely to have younger children that are still dependents, and there are few validated frailty and functional assessment tools for this population74.

b. Both geriatric populations and individuals with SCD have functional decline/disability74, worsening anemia9,45, cognitive impairment 124,125, increased susceptibility to infections13,112, silent cerebral ischemia 125,126, vision impairment 127,128, hearing loss 128,129, osteoporosis 122,130, vitamin D deficiency 122,130, joint replacement 69, a higher risk of myeloid malignancies compared to the general population 53,131, may be a caregiver for their partner or parents. renal disease 69,132, high rates of depression 133,134, systemic hypertension, increased venous thromboembolism (VTE) risk 135,136 heart failure 121,123, and chronic pain 69,137.

c. Geriatric populations life expectancy 70-80 years 138, osteoarthritis 139, institutionalization, polypharmacy 140, , coronary artery disease 121, experience falls 141, increased risk of solid tumors 142, have older children (non-dependents), and multiple functional assessment tools 143.

Figure adapted from Oyedeji CI, Hall K, Luciano A, Morey MC, Strouse JJ. Geriatric assessment for older adults with sickle cell disease: protocol for a prospective cohort pilot study. Pilot Feasibility Stud. 2020 Sep 17;6:131. doi: 10.1186/s40814-020-00673-3.

There are unique SCD-related changes that occur as people with SCD advance in age. Serjeant et al. showed that Jamaican adults with SCD aged ≥ 60 years were more likely to be female, have a higher fetal hemoglobin, experience fewer painful crises with age, and have declining hemoglobin and renal function.6 It is important to note that the majority of individuals (51%) in that cohort who were tested for α-thalassemia had at least 1 α-globin gene deletion, which is higher than the ~30% prevalence of alpha thalassemia seen in Blacks in the United States, and may have attenuated the severity of SCD.68 We previously published the largest multicenter study of older adults with SCD.9 We found that older adults (age ≥ 50 years) were more likely to have a lower hemoglobin after controlling for sex, SCD genotype, and hydroxyurea (HU) usage [9.8 g/dL for older adults vs. 10.3 g/dL for younger adults (18-49.99 years); p=0.008], leg ulcers, SCD-related eye complications, avascular necrosis, hip replacement, heart failure, and pulmonary hypertension (defined as tricuspid regurgitant jet velocity ≥ 2.5 meters/second or having evidence of pulmonary HTN on echocardiogram interpretation).9 Older adults had a lower mean eGFR (82 mL/min/1.73 m2 compared to 133 mL/min/1.73 m2 in younger adults; p<0.0001), higher baseline creatinine (mean 1.3 mg/dL versus 0.9 mg/dL; p<0.0001), and increased odds ratio (2.93, p<0.0001) of proteinuria (≥ 1+ on urine dipstick).9 The higher disease burden in older adults with SCD may be partially related to the lack of disease-modifying therapies available during their younger years.

We demonstrated that both younger adults (age 18-49) and older adults with SCD (age ≥ 50 years) also experienced accelerated functional decline, with a physical performance similar to individuals 20-30 years older than their chronological age.10 A review by Idris et al. summarized evidence of accelerated aging in SCD such as shorter telomere lengths associated with more severe disease, dysfunction of the ubiquitin–proteasome system leading to accumulation of low quality proteins, bone disease such as osteoporosis, and earlier onset end-organ dysfunction in people with SCD compared to individuals without SCD.11 This means that providers should consider geriatric conditions and syndromes such as frailty and falls in adults with SCD at an earlier age compared to the general population. Therefore, herein we define older adults as age ≥ 40 years, which is consistent with the definition commonly used in studies of aging in people with SCD.1215

1.2. Potentially Inappropriate Medications and Polypharmacy

As people age, they experience changes in body composition and develop multiple chronic diseases that can lead to changes in pharmacokinetics and pharmacodynamics of medications.16 Older adults in the general population often benefit from efforts to reduce risk of medication-related adverse outcomes, such as avoiding combinations of psychoactive medications that increase risk of falls, deprescribing unnecessary medications to reduce polypharmacy, and renally dosing medications to account for changes in eGFR.17,18 It is unclear at what age older adults with SCD start to experiencing age-related changes also affect drug pharmacodynamics.19,20 In a multicenter study of older adults with SCD, 60.8% of individuals aged ≥ 50 years reported taking opioids daily, which was similar to the proportion of young adults with SCD with daily opioid use.9 This suggests that SCD patients may not change their opioid use patterns as they age, despite the risk associated with taking opioid receptor agonists and sedatives at an older age. Typically, providers are encouraged to avoid or minimize administration of medications that are potentially inappropriate for use in older adults based on the Beers Criteria. The Beers Criteria are guidelines published by the American Geriatrics Society to improve the safety of prescribing medications for adults 65 years and older and include an evidence-based list of potentially inappropriate medications.17 Examples of medications associated with significant adverse events in older patients include anticholinergics, NSAIDS, anti-depressants, benzodiazepines, and opioid receptor agonists.17

Polypharmacy, defined as being on 5 or more regular prescribed medications, also is associated with falls, hospitalizations, and mortality in older adults.2123 The prevalence of polypharmacy in older adults with SCD is unknown. More data are needed on the prevalence and potential associated adverse effects of polypharmacy in older adults with SCD to help providers determine how they should approach modifying and/or deprescribing medications in adults with SCD as they advance in age. For older adults in the general population, every patient interaction is an opportunity to deprescribe medications with an emphasis placed on those who are frail.24 For older adults with SCD, we recommend that after age 40, medications and doses should be reviewed at each clinic visit, during transfers between care settings, and when considering prescribing new medications. We also advise that a formal medication assessment be done annually. The focus of deprescribing is to determine if dose adjustments can be made, if medications that are no longer clinically indicated can be discontinued, and if potentially inappropriate medications can be safely discontinued without compromising patents’ quality of life.24,25

2. Current Therapies

The current management strategy for SCD includes hydroxyurea as the cornerstone of therapy for individuals with HbSS/Sβ0 to reduce complications of SCD. More recently approved SCD therapies—L-glutamine, voxelotor, and crizanlizumab—are usually added to hydroxyurea rather than used as monotherapy. The use of multiagent therapy employing medications with different mechanisms of action is desirable, allowing one to target different pathologic pathways. (Table 1)

Table 1.

Current sickle cell therapies and potential issues in older adults

Therapy Mechanism Clinical trial in adults and age of treatment group Potential issues in older adults
Hydroxyurea26 -Increases fetal hemoglobin, thereby reducing polymerization of HbS
- Reduces leukocytes and number of adherent reticulocytes
- Reduces frequency of vaso-occlusive events and acute chest syndrome
Multicenter Study of Hydroxyurea (MSH)
Mean age 30.5, 10% over age 40
Renal clearance
May cause:
• Reversible, dose-dependent myelosuppression due to age-related decline in hematopoietic stem cells
• Decreased drug clearance as eGFR declines, requiring dose reduction
L-Glutamine27 Leads to production of nicotinamide adenine dinucleotide (NAD+) which reduces erythrocyte oxidative stress and adhesion Trial of L-Glutamine in Sickle Cell Disease
Median 19 (range 5-57)
Hepatic metabolism; metabolized to glutamate and ammonia by the liver
May cause constipation
Voxelotor28 Binds to hemoglobin and shifts O2 dissociation curve toward normal (i.e., leftward), thus reducing O2 tension at which hemoglobin deoxygenates, thereby reducing sickling and hemolysis Hemoglobin Oxygen Affinity Modulation to Inhibit HbS Polymerization
(HOPE) trial
Median age 24 (range 12-59)
Hepatic metabolism
Requires dose adjustment in patients with severe (Child-Pugh C) liver disease
• Limited experience with advanced renal disease
Crizanlizumab29,30 Monoclonal antibody targeting P-selectin to block adhesion of red blood cells and leukocytes to reduce vaso-occlusive events Sickle Cell Disease Patients with Pain Crises (SUSTAIN) trial
Median age 29 (range 16-63)
Reticuloendothelial system clearance
May cause:
• Infusion reaction
• Use with caution in patients with history of frequent severe infections
• Unknown if it is safe in the setting of kidney and/or liver impairment
Opioids31,32 Agonist or mixed agonist/antagonists of Mu-opioid receptor Weight-based vs patient-specific opioid dosing in acute setting
Median age 27.0 (IQR 23.0-32.5)
Hepatic (first pass) and renal clearance
May cause:
• Excess sedation, confusion, respiratory depression
• Opioid-induced constipation
• Increased falls
• Cognitive impairment
• Decreased muscle strength
• Osteoporosis
• Hypogonadism
• Dental Issues
Transfusion Therapy33,34 Increases total hemoglobin and reduces hemoglobin S% The Preoperative Transfusion in Sickle Cell Disease Study
0-9 years 40%
10-19 years 35%
≥ 20 years 25%
Reticuloendothelial system clearance
May cause:
• Volume overload
• Iron overload
• Alloimmunization to minor red blood cell antigens
The Transfusion Alternatives Preoperatively in Sickle Cell Disease (TAPS) study
Median age 13.4 (IQR 6.4-26.5)
Iron Chelation Therapy
Deferoxamine35
Deferiprone36
Deferasirox37
Removal of excess iron from tissues Barriers to adherence to Deferoxamine
Mean age 12.1 (range 5-17)
Deferasirox in Sickle Cell Trial
Median age 15 (range 3-54)
Ferriprox in Patients with Iron Overload in Sickle Cell Disease Trial (FIRST)
Mean age 16.9 ± 9.6 (range 3-59)
Deferoxamine
• Urine and fecal excretion
• May cause hearing loss, visual disturbances, and osteoporosis
Deferiprone
• Urine excretion
• May cause agranulocytosis and arthropathy
Deferasirox
• Fecal excretion
• Can be nephrotoxic so should be avoided when eGFR is < 40 mL/minute/1.73m238
• May cause acute liver injury
Deferoxamine and Deferiprone can be used in patients with decreased renal function; dialyzable

2.1. Hydroxyurea

Hydroxyurea (HU) was the first U.S. Food and Drug Administration (FDA)-approved medication for management of SCD.39 HU increases fetal hemoglobin, which reduces polymerization of HbS, decrease leukocytes, and lowers the number of adherent reticulocytes.40,41 It was initially approved in 1998 for patients with HbSS and frequent severe pain episodes on the basis of the Multicenter Study of Hydroxyurea (MSH). 39,42 This double-blind randomized placebo-controlled trial (RCT) demonstrated that HU reduced the incidence of painful episodes and acute chest syndrome in adults with SCD by almost 50%, leading to termination of the study before the planned end date.26,40 The mean age of participants in the MSH was 30.5 years, with only 10% of the 299 participants in the study aged ≥ 40 years.40,42 A 9-year follow up observational study of MSH patients showed that HU reduced mortality by 40%.41 It is currently the only approved SCD medication shown to improve survival.

There are no studies focused on the efficacy and safety HU in older adults with SCD. In our multicenter study, we found that 31.4% of older adults (aged ≥ 50) years and 42.1% of younger adults were taking HU. There was no significant difference in baseline hemoglobin between younger and older adults taking HU.9,43 Older adults not taking HU did have a lower hemoglobin compared to younger adults not taking HU.9 Older adults on HU also had a lower leukocyte count and platelet count compared to younger adults on HU, which was expected as HU-associated myelosuppression may compound age-associated cell count reduction.9,44,45

The effects of HU on the bone marrow remain unclear, considering that SCD itself has long-term negative effects on bone marrow health.46 Likely mechanisms include bone marrow ischemia from vaso-occlusion, pathologic angiogenesis within the bone marrow microenvironment, and bone marrow hyperplasia stimulated by anemia and tissue hypoxia, leading to increased osteoclasts and bone loss.4648 Pathologic bone marrow changes and the blunted erythropoietin response of older individuals with SCD can impair hematopoiesis.49,50

In addition, there may be an increase in clonal hematopoiesis of indeterminate potential (CHIP) in adults with SCD, as is seen in older adults in the general population; however, the data on CHIP in adults with SCD is limited. In a study using whole-exome sequencing data from individuals with SCD from 5 U.S. and international cohorts compared to African American controls without SCD, CHIP was more prevalent in individuals with SCD and started at a younger age (age 17 in the SCD group vs age 34 in the non-SCD group).51 The study was not powered to determine the impact of HU on CHIP. In another study of CHIP using specimens from the National Heart, Lung, and Blood Institute (NHLBI) Trans-Omics for Precision Medicine (TOPMed) consortium, there was no increase in CHIP among individuals with as compared to without SCD.52 HU also did not have an impact on the prevalence of CHIP.52 However, a population-based study in California showed that there was a 72% increase in risk of hematologic malignancies in individuals with SCD compared to the general California population, but no increase in risk of leukemia after HU was approved in 1998.53 A national retrospective study in England also demonstrated an increase in risk of lymphoma, multiple myeloma, and leukemia in individuals with SCD compared to those without SCD.54 These studies emphasize the need for better data to determine the impact, if any, of HU on the risk of CHIP and hematologic malignancies in SCD.

There are also concerns that HU may induce leg ulcers in 1 to 2% of older adults with myeloproliferative disorders.55 In the general population, hard-to-heal venous leg ulcers are more common in older individuals.56 Cutaneous leg ulcers in SCD typically are caused by vasculopathy from chronic hemolysis, and are associated with vaso-occlusion, lower hemoglobin, and lower fetal hemoglobin57,58 A recent study of HU use in 769 patients with SCD (mean age 38 ± 16) showed that the incidence of leg ulcers in SCD was independent of HU use.59 Although more data are needed on management of older adults on HU with leg ulcers, experts recommend that providers carefully consider the risk and benefit of discontinuing HU, since stopping HU may lead to exacerbation of SCD complications.59

As individuals both with and without SCD age, there is a decline in eGFR which requires reassessment of medication doses.9,60,61 The elimination of HU includes both liver metabolism and renal excretion; therefore, individuals with SCD and renal insufficiency may experience elevated plasma levels.62 In patients with SCD with creatinine clearance < 60 mL/min, the starting dose of HU should be reduced by 50% (7.5 mg/kg/day instead of 15 mg/kg/day).62

Recommendation: HU remains the backbone of therapy for management of SCD. For older adults with SCD it is important to routinely assess their renal and hepatic function and closely monitor for toxicity such as neutropenia, thrombocytopenia, or reticulocytopenia. The NHLBI 2014 Evidence-Based Management of Sickle Cell Disease guidelines, recommend a complete blood count with differential and reticulocyte count every 4 weeks when the HU dose is being adjusted and then every 2-3 months when on a stable dose.63 HU should be held for moderate to severe neutropenia or thrombocytopenia and then restarted after counts recover. When renal function declines, the care provider should consider reducing their HU dose. If cytopenias are severe and do not improve with holding or reducing the dose of HU, further evaluation for nutritional deficiencies, aplastic anemia, or hematologic malignancy may be indicated, since risk of these increases with age.

2.2. L-glutamine

L-glutamine is the second SCD disease-modifying medication approved by the FDA in 2017. L-glutamine is an essential amino acid required to produce nicotinamide adenine dinucleotide (NAD+). The proposed mechanism is by reducing erythrocyte oxidative stress and adhesion. In a phase 3 RCT, L-glutamine treatment for 1 year reduced the median number of vaso-occlusive events (VOEs) to 3 versus 4 in the placebo group (25% reduction) and the median number of hospitalizations to 2 versus 3 in the placebo group, a 33% reduction.27 Two-thirds of participants were also on HU. There were 2 deaths in the phase 3 trial, and both were sudden cardiac death in individuals on L-glutamine in their 40s.27 These 2 occurrences of sudden cardiac death represented 1% of participants in the treatment group. Sudden cardiac death is responsible for ~30% of deaths in adults with SCD.64,65 In addition, patients with renal and hepatic impairment were excluded from the clinical trial. Thus, it is unclear if there is an increased risk of adverse effects from L-glutamine in older adults with SCD.

Recommendation: It is reasonable to consider treating older adults with SCD with L-glutamine in combination with HU, as L-glutamine may be helpful in reducing VOEs and is best used in combination with HU rather than as monotherapy. However, L-glutamine must be used with caution in older adults with liver disease, since L-glutamine is metabolized into glutamate and ammonia by the liver.

2.3. Voxelotor

Voxelotor is an inhibitor of HbS polymerization designed to increase the oxygen affinity of hemoglobin and thereby reduce sickling of RBCs.28 Voxelotor reversibly binds to hemoglobin to stabilize it in an oxygenated state. Voxelotor reduces HbS polymerization and blood viscosity, which improves RBC deformability, reduces hemolysis, and extends RBC survival. Voxelotor was FDA approved in 2019 based on the HOPE Trial, a phase 3, multicenter, double-blind RCT demonstrating an increase in hemoglobin level by at least 1 g/dL in 51% of participants on 1500mg daily compared to 7% in the control group.28 There was also decreased hemolysis with a reduction in indirect bilirubin and reticulocyte count.

Safety, tolerability, and pharmacokinetics of voxelotor were assessed in 8 older adults (mean age 62.5 ± 7.1) with severe renal impairment (eGFR < 30 ml/min/1.73 m2) compared to healthy controls, investigators found no difference in the excretion of a single dose of 900mg.66 The standard dose (1500mg) or multiple doses were not tested in participants with severe renal impairment. It is unclear if it voxelotor is safe in patients with end-stage renal disease.

In the same study, investigators also evaluated the pharmacokinetics of voxelotor in 7 adults (mean age 55.4 ± 6.9) with SCD with hepatic impairment at various severity levels. Investigators found that a single dose of 1500mg was well-tolerated in participants with mild to moderate hepatic impairment.66 The mean concentration of voxelotor was 90% higher in individuals with severe hepatic impairment (Child-Pugh C) and investigators recommended a lower dose of voxelotor at 1000 mg in patients with severe hepatic impairment.66

Recommendation: Voxelotor is a promising treatment for older adults with SCD and moderate to severe anemia. It has the potential to reduce the need for transfusion by increasing the steady-state hemoglobin, thereby reducing the risk of symptomatic anemia due to chronic hemolysis. However, patients on chronic transfusion therapy for stroke prophylaxis likely would not benefit from voxelotor, since they are already increasing their hemoglobin with monthly transfusions providing normal RBCs, whereas voxelotor only increases the number of circulating HbS RBCs. It is also preferred that voxelotor be used in combination with HU, since HU increases HbF, which confers an additional anti-sickling effect, reduces acute complications, and may decrease mortality. Voxelotor should be used with caution in patients with hepatic impairment. More studies are needed on safety of voxelotor in older adults with SCD and to determine if increasing hemoglobin with voxelotor provides physical and cognitive benefits for older adults.

2.4. Crizanlizumab

Crizanlizumab is a monoclonal antibody targeting P-selectin expressed on the surface of the endothelium and platelets to block adhesion of leukocytes and thus reduce VOEs. Crizanlizumab was approved in 2019 for people with SCD age ≥ 16 years and is administered by intravenous infusion every 2 week for two doses and then every 4 weeks.

The SUSTAIN trial, a phase 2 multicenter, double-blind RCT, demonstrated that treatment with crizanlizumab (5 mg per kilogram) resulted in a 45% reduction in rate of VOEs (1.63 crises/year in 5 mg/kg crizanlizumab group compared to 2.98 crises/year in placebo group, p=0.01).29 Crizanlizumab also resulted in a longer median time to the first VOE compared to placebo (4.07 month in the 5 mg/kg crizanlizumab group versus 1.38 months in the placebo group, p=0.001). There also was a longer median time to the second VOE (10.3 vs. 5.1 months, p=0.02) with crizanlizumab therapy. Serious adverse events in the treatment group included pyrexia in 2 participants, pneumonia in 5 participants, and influenza in 3 particpants.29 The effect of renal and hepatic impairment on the pharmacokinetics of crizanlizumab is unknown.67

There currently are ongoing clinical trials to determine the efficacy of crizanlizumab to reduce recurrent priapism and proteinuria in people with SCD.

Recommendation: Crizanlizumab is an effective treatment for adults with SCD with at least 2 VOEs per year. It is unclear if there is a difference in efficacy or safety in older adults, since the effect of renal and hepatic impairment is unknown. There were a few participants who experienced pyrexia and infections as severe adverse events; therefore, crizanlizumab should be used with caution in older adults with a history of frequent infections.

2.5. Opioids

Acute Pain Management

Opioids are a mainstay of therapy for VOEs in individuals with SCD. Oral opioids are commonly prescribed for VOEs that can be managed at home. Parenteral opioids are used for severe VOEs managed in the emergency department (ED), sickle cell day hospital, and inpatient settings.

Multiple studies have shown that older adults with SCD have less healthcare utilization compared to younger adults, but the data is conflicting on whether older adults with SCD experience less pain.6870 Older adults (aged ≥ 37 years) in the Pain in Sickle Cell Epidemiology Study (PiSCES) had higher proportion of pain and pain crisis days, increased mean number of body parts that hurt, and higher proportion of home pain days on opioids compared to the transition group (age 16-25 years). However, older adults had a 50% lower ED reliance for health care compared to the transition group and younger adults aged 26-36 years. All groups had similar pain intensity, proportion of days in the hospital, and fetal hemoglobin levels (%). We also showed a similar proportion of younger and older adults with SCD with daily short-acting and long-acting opioid use, but older adults had fewer hospitalizations.69 These findings of similar pain but less utilization may be due to older adults having better home self-management strategies compared to younger adults. On the contrary, a Jamaican cohort of older adults with SCD age ≥ 60 years reported that pain improved with age and had a higher fetal hemoglobin, which may be explained by survival bias and the higher prevalence of alpha thalassemia in this cohort.6

Older adults in the general population (age ≥ 65 years) experience increased rates of social, medical, and psychological adverse effects related to opioid use.71 Older adults are at higher risk of developing adverse effects such as constipation, falls, sedation, respiratory depression, delirium, and cognitive impairment.71

The American Society of Hematology (ASH) 2020 guidelines for SCD suggest adding non-pharmacologic therapies such as massage, yoga, and audiovisual relaxation for management of acute pain.31

Chronic Pain Management

Chronic pain is a common and debilitating complication in adults with SCD with over half of adults in the United States meeting criteria for chronic pain.72 The etiology of chronic pain can be related to both SCD and non-SCD causes and is often managed with short-acting and/or long-acting opioids.73 Chronic pain, depression, and multiple co-morbidities are risk factors for opioid-related adverse effects in older adults.71 These conditions are common in older adults with SCD; however, more data are needed on whether they have similar risk of adverse effects from opioids compared to older adults in the general population.9,74

Data on chronic opioid therapy in adults with SCD suggest worse symptom burden, poor functional outcomes, and higher healthcare utilization compared to adults with SCD not on chronic opioids.75 Older age and pre-transplant use of long-acting opioids have been associated with persistent pain after curative therapy with hematopoietic stem cell transplant in adults with SCD.76 In a qualitative study, adults with SCD reported an interest in non-opioid options for pain management, but expressed concerns about access and costs of alternative therapies.77 The ASH guidelines suggest including strategies such as non-opiate medications (serotonin and norepinephrine reuptake inhibitor [medications, gabapentinoids, tricyclic antidepressants and non-pharmacologic therapies (cognitive behavior therapy and acupuncture for management of chronic pain).31

Recommendation: Older adults in general are more sensitive to the effects of opioids. As people with SCD age, it is important to review their outpatient and inpatient opioid analgesic regimens to determine if dose reductions are indicated. As their renal function and lean body mass decline with age, they may become more susceptible to adverse events. Data on weaning full agonist opioids and transitioning to buprenorphine has demonstrated safety and efficacy in reducing acute visit in adults with SCD.78 It is advisable to incorporate more non-opioid and non-pharmacologic strategies, such as cognitive behavioral therapy, massage, yoga, and acupuncture to reduce pain and the risk of medication-related side effects in older adults with SCD.31,79 More data are needed to determine the most effective and safe pain management strategies for older adults with SCD.

2.6. Transfusion

Throughout the lifetime of a person with SCD, there are many scenarios where RBC transfusions are indicated. They may experience symptomatic anemia due to an acute hemolytic episode or an aplastic crisis.80 Simple transfusion or red cell exchange (RCE) may also be used to manage moderate or severe acute chest syndrome, treat an acute stroke, or to reduce the risk of complications in the perioperative setting.80,81 Many individuals receive chronic simple RBC transfusions or RCE for primary or secondary stroke prophylaxis, recurrent acute chest syndrome, or cardiopulmonary disease with severe anemia. The SCD-CARRE trial (NCT04084080) is an ongoing multi-center phase 3 RCT to determine the efficacy of RCE vs standard care in high mortality risk adults (age ≥ 18 years with significant cardiopulmonary or renal disease) with SCD to reduce total number of episodes of clinical worsening of SCD requiring acute health care encounters or death over 12 months.

Blood transfusions are common in older adults in the general U.S. population. Older adults are more likely to experience transfusion-related complications such as transfusion-associated circulatory overload (TACO) and alloimmunization.82,83 There is mixed data on the benefit of liberal vs restrictive transfusion strategies in adults in the general population. A systematic review and meta-analysis that included studies in adults aged ≥ 65 years demonstrated a higher mortality rate in those who followed restrictive transfusion strategies compared to a liberal transfusion strategies.84

Since older adults with SCD have a lower hemoglobin, more comorbidities, and are more likely to undergo hip surgery compared to younger adults, they are more likely to encounter scenarios where transfusions are warranted.9 There are few high-quality studies to guide transfusion for adults with SCD undergoing surgery.85 A RCT comparing conservative transfusion (increasing hemoglobin to 10 g/dL with simple transfusion) to aggressive transfusion (RCE with goal HbS < 30%) demonstrated no difference in perioperative complications between the 2 groups.33 The Transfusion Alternatives Preoperatively in Sickle Cell Disease (TAPS) study was another RCT that demonstrated a benefit to preoperative transfusion compared to no transfusion in individuals with HbSS or HbSβ0 undergoing low or moderate risk surgery.34 The transfused group had fewer clinically significant complications compared to the non-transfused group (15% vs 39%, respectively).34 There are no dedicated transfusion guidelines for older adults with SCD. Studies demonstrating the benefits of transfusion for primary and secondary stroke prevention in individuals with SCD were performed in children.86,87 Existing studies on transfusions in SCD have not had adequate representation from older adults (age ≥ 40 years) and people with severe end-organ damage due to conservative inclusion and exclusion criteria that do not reflect real-world experience.

Recommendation: When considering transfusion in older adults with SCD it is important to take an individualized approach. Consider the indications for acute transfusions and chronic transfusion therapy.80,85 Review their medical history for renal failure and cardiopulmonary issues that may put them at risk for TACO. Older adults often require a slower rate of transfusion and may benefit from diuretics or isovolemic RCE. Review their history for prior transfusion reactions, alloantibodies, and iron overload. If they are on chronic transfusion therapy, weigh the risk and benefit of continuing chronic transfusion as they age. New and emerging therapies such as anti-sickling agents and pyruvate kinase activators increase hemoglobin in some individuals with SCD. These agents show promise as a potential way to reduce transfusion in adults with SCD. In addition, the use of erythropoiesis-stimulating agents may also reduce the need for transfusion in adults with SCD, especially in individuals with anemia of CKD.88,89

2.7. Iron chelation

Transfusional iron overload is a common and undertreated condition in adults with SCD. The duration and severity of iron overload increases the risk of iron toxicity and damage to multiple organs, which contributes to liver disease, diabetes, hypogonadism, malignancy, ineffective erythropoiesis, cardiomyopathy, and mortality.90 Iron toxicity can be especially problematic in older adults with SCD who already have organ damage and multiple transfusions over the course of their lives.90 In a study on screening and management of iron overload in 1223 adults with SCD in the Globin Research Network for Data and Discovery (GRNDaD) registry, out of the 69 individuals with a liver iron concentration (LIC) ≥ 5 mg/g dry weight, 20.3% were aged 35-49 years and 17.4% were ≥ 50 years.91

Iron chelation is recommended in patients with iron overload with LIC > 3 mg/g dry weight or serum ferritin > 1000 μg/l.90 The three approved iron chelators are deferoxamine, deferiprone, and deferasirox. Deferoxamine must be administered by continuous subcutaneous or intravenous infusion due to its short half-life and poor oral bioavailability. Deferiprone is administered as a tablet two or three times a day. Deferasirox is available in 2 formulations (dispersible tablet for oral suspension and a film-coated tablet), but the film-coated tablet is better tolerated and has better adherence.92 One of the major concerns about using deferasirox in older adults with SCD is nephrotoxicity.93 As adults with SCD and iron overload develop worsening renal function, they may have to switch to deferiprone or deferoxamine, since these can be used in patients with renal disease. Another concern is that deferoxamine can cause hearing loss and impaired vision, two conditions that are already of concern as individuals with SCD age.5,90

Recommendation: There is limited data on iron chelation therapy in older adults with SCD; therefore, more studies are required to better understand safety in this population. For older adults with SCD and iron overload, we recommend use of iron chelation agents to reduce the risk of organ damage and mortality associated with iron toxicity. When selecting an iron chelator, consider the patient’s comorbidities. Adults with SCD are at higher risk of sensorineural hearing loss compared to the general population.94 When considering starting deferoxamine, refer the patient for audiology and vision exams to establish a baseline.95 Deferasirox should be used with caution in patients with renal impairment and discontinued if their eGFR < 40 mL/minute/1.73m2.38 Older adults with SCD are at higher risk for cytopenias; therefore, patients on deferiprone should have their absolute neutrophil count monitored closely (weekly for 6 months and then every 2 weeks) given the risk of medication-induced agranulocytosis.96

3. Emerging therapies under investigation

There are multiple investigational therapies for the management of SCD (Table 2). However, it is unclear how these new treatments will benefit older adults with SCD since clinical trials are often not powered to assess efficacy in this sub-population.

Table 2.

Emerging therapies for management of sickle cell disease

Medication Mechanism Clinical Trial Information
Pyruvate Kinase Activator Etavopivat RBC pyruvate kinase-R (PKR) activator. Decreases 2,3-DPG in RBCs to increase Hb oxygen affinity.
Increase ATP
Phase 2/3:
NCT04624659 HIBISCUS
Age 12-65 years
Endpoints:
• Increase in hemoglobin
• Rate of VOEs.
Mitapivat RBC pyruvate kinase-R (PKR) activator. Decreases 2,3-DPG in RBCs to increase hemoglobin oxygen affinity.
-Increase ATP
Phase 2 /3
NCT05031780
Age ≥ 16 years
Endpoints:
• % with a hemoglobin response by 12 weeks
• % with treatment-emergent adverse events and treatment-emergent serious adverse events
• % with a hemoglobin response by week 52
• Phase 3: Annual rate of sickle cell pain crises
Anti-sickling GBT021601 Second generation HbS polymerization inhibitor. Binds to hemoglobin and shifts O2 dissociation curve toward normal (i.e., leftward), thus reducing O2 tension at which hemoglobin deoxygenates, thereby reducing sickling Phase 2/3 NCT05431088
6 Months to 65 Years
Endpoints:
• Number of adults with change from baseline in hemoglobin through week 12
• Proportion of participants with an increase from baseline of >1 g/dL in hemoglobin at week 48
• Pharmacokinetics, while observing maximum concentration after a single dose.
• Pharmacokinetics, at minimum concentration and maximum concentration after multiple dose administration
Anti-adhesion therapies Inclacumab IgG4 monoclonal antibody that selectively targets P-selectin Phase 3
NCT04935879
Age ≥ 12
Endpoints:
• Rate of VOEs during the 48-week treatment period
Phase 3
NCT04927247
Age ≥ 12 years
Endpoints:
• Re-admission for a VOE within 90 days of randomization
Antioxidant/Anti-inflammatory Crovalimab C-5 (complement) inhibitor to downregulate complement contribution to acute VOE Phase 2
CROSSWALK-c
(NCT05075824)
Aged 12 - 55 years
Endpoints:
• Annual rate of medical facility VOEs
Rifaximin Oral antibiotic for intestinal microbial modulation (gut decontamination) to reduce translocation of intestinal bacteria and activated neutrophils Phase 2
(NCT03719729)
Median age 29 years (range 24-56).
Endpoints:
• Toxicity profile (incidence of nausea, vomiting, diarrhea, abdominal discomfort, worsening anemia)

DPG, diphosphoglycerate. RBC, red blood cell, ATP, adenosine triphosphate. VOE, vaso-occlusive events

3.1. Pyruvate kinase activators

Etavopivat is an oral RBC pyruvate kinase-R (PKR) activator that decreases in 2,3-diphosphoglycerate (2,3-DPG) in RBCs, which in turn increases hemoglobin oxygen affinity.97 Increasing hemoglobin oxygen affinity then reduces HbS polymerization and RBC sickling.

Etavopivat was shown to decrease 2,3-DPG and increase adenosine triphosphate (ATP) in non-human primates and in healthy subjects.97 In an ex-vivo study of whole blood of individuals with SCD, treatment with etavopivat resulted in increased hemoglobin oxygen affinity and improved sickle RBC function.97 A phase 1 study was recently completed (NCT03815695), showing that most patients treated with etavopivat achieved a total hemoglobin increase of 1 g/dL or more (manuscript in preparation). There is currently a phase 2/3 trial evaluating the safety and efficacy of etavopivat (HIBISCUS; NCT04624659).

Mitapivat is another oral PKR activator under investigation for management of anemia and VOEs in SCD. In a phase 1 dose-escalation clinical trial (NCT04000165), mitapivat was shown to increase hemoglobin by a mean of 1.2 g/dL at the 50 mg dose, and 56.3% (9/16) of participants (mean age 39 years) achieved a hemoglobin response of ≥ 1 g/dL compared to baseline.49 There were also reductions in hemolytic markers. Four VOEs (23.5%) were documented as severe adverse events, 2 of which occurred while the dose was being tapered.49

3.2. Anti-sickling therapies

GBT021601 is a second generation HbS polymerization inhibitor. In SCD mouse models, investigators noted a dose-dependent reduction in p50 (partial pressure at which hemoglobin is 50% saturated), increased hemoglobin and reduced hemolysis.98 GBT021601 is more potent than voxelotor, which may allow for lower dosing and a lower pill burden. Phase 1 studies in 63 healthy volunteers (NCT05036512) and 6 adults with SCD (NCT04983264) demonstrated a higher % hemoglobin occupancy compared to voxelotor in healthy controls, it was well tolerated, and there was no evidence of tissue hypoxia.99 The phase 2/3 trial for GBT021601 in individuals aged 6 months to 65 years is ongoing (NCT05431088).

3.3. Anti-adhesion therapies

Inclacumab is a humanized IgG4 monoclonal antibody that competitively inhibits the interaction between P-selectin with its ligand P-selectin glycoprotein ligand 1 (PSGL-1) to reduce adhesion of sickle RBCs by inhibiting platelet-leukocyte aggregate formation.100,101 It differs from crizanlizumab in that the inhibition with inclacumab is directly in the PSGL-1 binding region on P-selectin rather than at a distant epitope, as with crizanlizumab.102 In a study using blood samples from healthy controls and from people with SCD, inclacumab demonstrated greater maximal inhibition of platelet-leukocyte cell adhesion compared to crizanlizumab.102

Preliminary results of the phase 1 study in 15 healthy adults (median age 42 years (range 22-52) receiving a single dose of inclacumab at 20 mg/kg (n=6) or 40 mg/kg (n=9) demonstrated no treatment-emergent adverse events greater than grade 1.103 Platelet-leukocyte aggregate formation decreased from 33-39% at baseline to 9-14% at 2 hours following end of infusion and was sustained for at least 12 weeks.103

The phase 3 trial of inclacumab in individuals with SCD age ≥ 12 years is ongoing (NCT04935879).

3.4. Antioxidants and anti-inflammatory agents

There are several studies that demonstrate that inflammation and complement activation play important roles in pathologic manifestations of SCD.104,105 A study of the role of C5 in the pathophysiology of SCD demonstrated that C5a and the membrane attack complex (C5b-9) were mediators of ischemia-reperfusion injury and that C5a given to SS mice induced vasoocclusion.106

Crovalimab is a C5 inhibitor evaluated in the phase 1 CROSSWALK clinical trial, in individuals age 12-55 for management of acute uncomplicated VOEs (NCT04912869).107 The current phase 2 trial is to assess the prevention of VOEs in individuals with SCD (NCT05075824). Crovalimab was also previously studied in an open-label phase 1/2 trial for management of paroxysmal nocturnal hemoglobinuria (PNH) using escalating intravenous doses up to 1500mg followed by 170mg - 680mg administered subcutaneously up to every 4 weeks (COMPOSER trial NCT03157635).108 Crovalimab differs from approved C-5 inhibitors (eculizumab and ravilizumab) in that it is highly soluble, allowing subcutaneous self-administration of small volumes.108 It also may have a lower risk of breakthrough symptoms in patients with PNH, where it was shown to decrease hemolytic activity.108 There were no serious treatment-related adverse effects.

Rifaximin is an antibiotic under investigation for use in SCD for its ability to modulate the intestinal microbiome and reduce inflammation.109 Activated and aged neutrophils may increase risk of sickle RBC adherence to the endothelium.109 Individuals with SCD have high levels of activated neutrophils and circulating aged neutrophils. Intestinal microbial composition, such as intestinal bacterial overgrowth, may increase neutrophil activation and VOEs in individuals with SCD. In a Phase 2 clinical trial with 11 adults with HbSS, oral rifaximin 550mg twice daily significantly reduced circulating aged neutrophils by a median 12.7% (range 6.1-40.1] after 2-4 weeks of therapy.110 There were no Clostridium difficile infections or adverse effects.110 In the same trial, treatment with rifaximin in 12 patients reduced median number of VOEs from 2.25 (range 1-6.5) per 6 months to 1 per 6 months (range 0-4) (P =.003).109 The mean self-reported medication adherence was 86% (range 50-100). There was no significant change in white blood cell count, hemoglobin, and hemolysis labs.109

3.5. Novel Development in gene therapy and gene editing

Curative therapies such as hematopoietic stem cell transplantation (HCST), including matched sibling and alternative donors, and gene therapy (gene addition and editing) will likely continue to improve the treatment of people with SCD. However, multiple challenges have limited the availability of these therapies, especially for older adults. Currently, matched sibling HCST is recommended for people with severe SCD as defined by prior stroke or abnormal TCD, frequent pain or recurrent acute chest syndrome despite disease modifying medications. HSCT outcomes (including better disease-free survival and less graft versus host disease) are good for those < 16 years old.111 Availability and experience in adults is more limited. A recent meta-analysis of HSCT for SCD identified 33 studies with 2853 participants. Only 8 studies included participants over 40 years and only 3 studies, enrolling a total of 53 participants, had a median age of over 30 years.112 HCST with reduced intensity conditioning and post-transplant cyclophosphamide permits the use of haploidentical donors, greatly expanding the proportion of people with SCD with donors. This approach is well tolerated in adults with SCD and has lower treatment related mortality compared to myeloablative approaches in older adults leading to increased availability of HSCT.113

Gene therapy is also in clinical trials, and some gene therapy studies include older ages. However, few older adults qualify for the current studies due to rigorous exclusion criteria designed to assure the safety of the required myeloablative preparative regimen generally with high-dose busulfan. The oldest patient in a published report to receive gene therapy is 38 years old.114116

4. Considerations when using combination therapy in older adults

When combining different drugs in older adults with SCD it is important to consider the following:

  1. Hydroxyurea is currently the only approved medication for SCD that has a mortality benefit in SCD; therefore, it should remain an integral part of treatment. Consider the patient’s history of adherence to hydroxyurea and whether it has been optimized by titrating it to the maximum tolerated dose before adding an additional agent.

  2. Consider the patient’s underlying comorbidities, such as CKD or hepatic impairment, especially in patients with severe iron overload. More data are needed to determine the safety of new and emerging therapies in older frail patients and those with end organ damage.

  3. As adults with SCD have new and emerging medications added to their regimen, this is expected to increase the complexity of their medication management and thus increase their risk of medication-related adverse events. Consider the patient’s concerns about polypharmacy, pill burden, issues with swallowing, and visual issues that may make it difficult to read pill bottles. Inquire about their current system for keeping up with taking the right medications at the right time, such as use of a pill-organizer. Also, ask about their social support system at home and determine if they qualify for home health services to help with medication management.

5. Conclusion

There is a paucity of data to guide management of older adults with SCD, requiring extrapolation from SCD data in younger people and data on older adults in the general population. Using guidelines based on clinical trials performed in children and young adults may not be generalizable to older adults who have different physiology and comorbidities. It is also important to note that chronologic age often does not reflect physiologic age, so studies on safety and efficacy of SCD therapies should include frail individuals with advanced physiologic aging in addition to more robust older adults.117

5.1. Future Directions

SCD clinical trials have historically had little or no representation from older adults (age ≥ 40 years) and individuals with severe end organ damage or frailty. There is a need for clinical trials focused on older adults with SCD to determine how to safely prescribe disease-modifying therapies and other commonly used medications in this population. These trials should include the effects of physiologic changes and comorbid conditions that may occur earlier in adults with SCD and can increase the risk of adverse events. Until there is more data on current and emerging therapies in older adults with SCD, we recommend closer monitoring and reporting of adverse events. The most important areas of focus for future studies involving older adults with SCD include: 1) establishing the most clinically relevant and patient-important outcomes for older adults with SCD, such as improving function, and including these outcome measures in clinical trials;74,118 2) non-pharmacologic strategies to improve pain, extend healthspan (which is the length of time a person is healthy), and quality of life;119 3) real-world experience on effectiveness of disease-modifying therapies (monotherapy and combination therapy) in older adults with SCD, since they have historically been underrepresented in clinical trials; 4) pharmacokinetic and pharmacodynamic studies in older adults with SCD to determine optimal doses of medications to minimize risk of adverse events; and 5) transfusion guidelines for older adults with SCD.

Key Points.

Clinical trials for current and emerging therapies for sickle cell disease have historically had little or no representation from older adults (age ≥ 40 years) and individuals with severe end organ damage or frailty; therefore, more real-world data are needed to determine safety and effectiveness of sickle cell therapies in older adults.

Management of older adults with sickle cell disease should focus on both disease and age-related conditions using pharmacologic and non-pharmacologic strategies with special consideration for the impact of physiological changes that occur age, such as a decline in renal and hepatic function and increased sensitivity to psychoactive medications.

Hydroxyurea should remain the cornerstone of disease-modifying therapy for sickle cell disease since it is currently the only approved medication shown to improve survival in sickle cell disease; the addition of other novel therapies with complementary mechanisms of action should be considered in those with inadequate disease control.

Acknowledgments

We would like to acknowledge the contribution of people with SCD who participated in the clinical studies included in this review article and the investigators who led these studies.

Funding

This study was funded by the Duke Center for Research to Advance Healthcare Equity (REACH Equity), which is supported by the National Institute on Minority Health and Health Disparities under award number U54MD012530 and the National Institute on Aging (NIA) Grants for Early Medical/Surgical Specialists’ Transition to Aging Research (GEMSSTAR) award number 1R03AG074054-01 to C.I.O in the preparation of this manuscript. J.J.S received support from the HRSA for the Education and Mentoring to Bring Access to Comprehensive Care (EMBRACE) Network (U1EMC42461-01) for the preparation of this manuscript.

Conflicts of Interest

C.I.O. has received research funds from the NIA (NIH).

K.L.H. declares that they have no conflict of interest.

M.J.T. has received research funds from Doris Duke Charitable Foundation, NIDDK and NHLBI (NIH), FDA, Forma Therapeutics, and CSL Behring; she has also served on an advisory board for Pfizer, Inc., and on a Data Monitoring Committee for Novartis.

J.J.S. receives research funds from NHLBI, CDC, Takeda, and Agios. He has served on a scientific advisory board on gene therapy for sickle cell disease for Aruvant.

Footnotes

Ethics approval

Not applicable

Consent to participate: Not applicable

Consent for publication: Not applicable

Code availability

Not applicable

Availability of data and material

No datasets were generated or analyzed during the current study.

REFERENCES

  • 1.Rees DC, Williams TN, Gladwin MT. Sickle-cell disease. The Lancet. 2010/December/11/2010;376(9757):2018–2031. doi: 10.1016/S0140-6736(10)61029-X [DOI] [PubMed] [Google Scholar]
  • 2.Papageorgiou DP, Abidi SZ, Chang H-Y, et al. Simultaneous polymerization and adhesion under hypoxia in sickle cell disease. Proceedings of the National Academy of Sciences. 2018;115(38):9473–9478. doi:doi: 10.1073/pnas.1807405115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kato GJ, Steinberg MH, Gladwin MT. Intravascular hemolysis and the pathophysiology of sickle cell disease. J Clin Invest. Mar 1 2017;127(3):750–760. doi: 10.1172/jci89741 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Elmariah H, Garrett ME, De Castro LM, et al. Factors associated with survival in a contemporary adult sickle cell disease cohort. Am J Hematol. May 2014;89(5):530–5. doi: 10.1002/ajh.23683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Oyedeji CI, Hall K, Luciano A, Morey MC, Strouse JJ. Geriatric assessment for older adults with sickle cell disease: protocol for a prospective cohort pilot study. Pilot and Feasibility Studies. 2020/09/17 2020;6(1):131. doi: 10.1186/s40814-020-00673-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Serjeant GR, Serjeant BE, Mason KP, Hambleton IR, Fisher C, Higgs DR. The changing face of homozygous sickle cell disease: 102 patients over 60 years. Int J Lab Hematol. Dec 2009;31(6):585–96. doi: 10.1111/j.1751-553X.2008.01089.x [DOI] [PubMed] [Google Scholar]
  • 7.Steinberg MH. Genetic etiologies for phenotypic diversity in sickle cell anemia. ScientificWorldJournal. Jan 18 2009;9:46–67. doi: 10.1100/tsw.2009.10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Steinberg M, Embury S. Alpha-thalassemia in blacks: genetic and clinical aspects and interactions with the sickle hemoglobin gene. Blood. 1986;68(5):985–990. doi: 10.1182/blood.V68.5.985.985 [DOI] [PubMed] [Google Scholar]
  • 9.Oyedeji C, Strouse JJ, Crawford RD, Garrett ME, Ashley-Koch AE, Telen MJ. A multi-institutional comparison of younger and older adults with sickle cell disease. Am J Hematol. 2019;94(4):E115–E117. doi: 10.1002/ajh.25405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Oyedeji CI, Hall K, Luciano A, Morey MC, Strouse JJ. The Sickle Cell Disease Functional Assessment (SCD-FA) tool: a feasibility pilot study. Pilot Feasibility Stud. Mar 4 2022;8(1):53. doi: 10.1186/s40814-022-01005-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Idris IM, Botchwey EA, Hyacinth HI. Sickle cell disease as an accelerated aging syndrome. Exp Biol Med (Maywood). 2022;247(4):368–374. doi: 10.1177/15353702211068522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sandhu MK, Cohen A. Aging in Sickle Cell Disease: Co-morbidities and New Issues in Management. Hemoglobin. 2015;39(4):221–4. doi: 10.3109/03630269.2015.1040493 [DOI] [PubMed] [Google Scholar]
  • 13.Khan AB, Kesse-Adu R, Breen C, et al. A descriptive study of the characteristics of older adults with sickle cell disease. Am J Hematol. Feb 2018;93(2):E38–e40. doi: 10.1002/ajh.24961 [DOI] [PubMed] [Google Scholar]
  • 14.Thein SL, Howard J. How I treat the older adult with sickle cell disease. Blood. 2018;132(17):1750–1760. doi: 10.1182/blood-2018-03-818161 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Oyedeji CI, Faldowski R, Morey M, et al. Characterizing Frailty in Adults with Sickle Cell Disease Using Frailty Phenotype. Blood. 2022;140(Supplement 1):5125–5126. doi: 10.1182/blood-2022-169616 [DOI] [Google Scholar]
  • 16.Shi S, Klotz U. Age-related changes in pharmacokinetics. Curr Drug Metab. Sep 2011;12(7):601–10. doi: 10.2174/138920011796504527 [DOI] [PubMed] [Google Scholar]
  • 17.Panel BtAGSBCUE. American Geriatrics Society 2019 Updated AGS Beers Criteria® for Potentially Inappropriate Medication Use in Older Adults. J Am Geriatr Soc. 2019;67(4):674–694. doi: 10.1111/jgs.15767 [DOI] [PubMed] [Google Scholar]
  • 18.Sergi G, De Rui M, Sarti S, Manzato E. Polypharmacy in the Elderly: Can Comprehensive Geriatric Assessment Reduce Inappropriate Medication Use? Drugs & Aging. Jul 2011 2014-12-07 2011;28(7):509–519. doi: 10.2165/11592010-000000000-00000 [DOI] [PubMed] [Google Scholar]
  • 19.Mangoni AA, Jackson SH. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. Jan 2004;57(1):6–14. doi: 10.1046/j.1365-2125.2003.02007.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bowie MW, Slattum PW. Pharmacodynamics in older adults: a review. Am J Geriatr Pharmacother. Sep 2007;5(3):263–303. doi: 10.1016/j.amjopharm.2007.10.001 [DOI] [PubMed] [Google Scholar]
  • 21.Gnjidic D, Hilmer SN, Blyth FM, et al. Polypharmacy cutoff and outcomes: five or more medicines were used to identify community-dwelling older men at risk of different adverse outcomes. J Clin Epidemiol. Sep 2012;65(9):989–95. doi: 10.1016/j.jclinepi.2012.02.018 [DOI] [PubMed] [Google Scholar]
  • 22.Jyrkkä J, Enlund H, Korhonen MJ, Sulkava R, Hartikainen S. Polypharmacy status as an indicator of mortality in an elderly population. Drugs Aging. 2009;26(12):1039–48. doi: 10.2165/11319530-000000000-00000 [DOI] [PubMed] [Google Scholar]
  • 23.Zaninotto P, Huang YT, Di Gessa G, Abell J, Lassale C, Steptoe A. Polypharmacy is a risk factor for hospital admission due to a fall: evidence from the English Longitudinal Study of Ageing. BMC Public Health. 2020/11/26 2020;20(1):1804. doi: 10.1186/s12889-020-09920-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Frank C, Weir E. Deprescribing for older patients. Cmaj. Dec 9 2014;186(18):1369–76. doi: 10.1503/cmaj.131873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Scott IA, Hilmer SN, Reeve E, et al. Reducing inappropriate polypharmacy: the process of deprescribing. JAMA Intern Med. May 2015;175(5):827–34. doi: 10.1001/jamainternmed.2015.0324 [DOI] [PubMed] [Google Scholar]
  • 26.Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med. May 18 1995;332(20):1317–22. doi: 10.1056/nejm199505183322001 [DOI] [PubMed] [Google Scholar]
  • 27.Niihara Y, Miller ST, Kanter J, et al. A Phase 3 Trial of l-Glutamine in Sickle Cell Disease. New England Journal of Medicine. 2018;379(3):226–235. doi: 10.1056/NEJMoa1715971 [DOI] [PubMed] [Google Scholar]
  • 28.Vichinsky E, Hoppe CC, Ataga KI, et al. A Phase 3 Randomized Trial of Voxelotor in Sickle Cell Disease. New England Journal of Medicine. 2019;381(6):509–519. doi: 10.1056/NEJMoa1903212 [DOI] [PubMed] [Google Scholar]
  • 29.Ataga KI, Kutlar A, Kanter J, et al. Crizanlizumab for the Prevention of Pain Crises in Sickle Cell Disease. New England Journal of Medicine. 2016;376(5):429–439. doi: 10.1056/NEJMoa1611770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kutlar A, Kanter J, Liles DK, et al. Effect of crizanlizumab on pain crises in subgroups of patients with sickle cell disease: A SUSTAIN study analysis. Am J Hematol. Jan 2019;94(1):55–61. doi: 10.1002/ajh.25308 [DOI] [PubMed] [Google Scholar]
  • 31.Brandow AM, Carroll CP, Creary S, et al. American Society of Hematology 2020 guidelines for sickle cell disease: management of acute and chronic pain. Blood Advances. 2020;4(12):2656–2701. doi: 10.1182/bloodadvances.2020001851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tanabe P, Silva S, Bosworth HB, et al. A randomized controlled trial comparing two vaso-occlusive episode (VOE) protocols in sickle cell disease (SCD). Am J Hematol. 2018;93(2):159–168. doi: 10.1002/ajh.24948 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Vichinsky EP, Haberkern CM, Neumayr L, et al. A comparison of conservative and aggressive transfusion regimens in the perioperative management of sickle cell disease. The Preoperative Transfusion in Sickle Cell Disease Study Group. N Engl J Med. Jul 27 1995;333(4):206–13. doi: 10.1056/nejm199507273330402 [DOI] [PubMed] [Google Scholar]
  • 34.Howard J, Malfroy M, Llewelyn C, et al. The Transfusion Alternatives Preoperatively in Sickle Cell Disease (TAPS) study: a randomised, controlled, multicentre clinical trial. Lancet. Mar 16 2013;381(9870):930–8. doi: 10.1016/s0140-6736(12)61726-7 [DOI] [PubMed] [Google Scholar]
  • 35.Treadwell MJ, Law AW, Sung J, et al. Barriers to adherence of deferoxamine usage in sickle cell disease. Pediatr Blood Cancer. May 2005;44(5):500–7. doi: 10.1002/pbc.20290 [DOI] [PubMed] [Google Scholar]
  • 36.Kwiatkowski JL, Hamdy M, El-Beshlawy A, et al. Deferiprone vs deferoxamine for transfusional iron overload in SCD and other anemias: a randomized, open-label noninferiority study. Blood Advances. 2022;6(4):1243–1254. doi: 10.1182/bloodadvances.2021004938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vichinsky E, Onyekwere O, Porter J, et al. A randomised comparison of deferasirox versus deferoxamine for the treatment of transfusional iron overload in sickle cell disease. Br J Haematol. Feb 2007;136(3):501–8. doi: 10.1111/j.1365-2141.2006.06455.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Novartis Pharmaceuticals Corporation. Jadenu (Defirasirox) [package insert]. Novartis website. https://www.hcp.novartis.com/products/jadenu/chronic-iron-overload/dosing--administration/.2021. Accessed September 09, 2022. [Google Scholar]
  • 39.Strouse JJ, Heeney MM. Hydroxyurea for the treatment of sickle cell disease: efficacy, barriers, toxicity, and management in children. Pediatr Blood Cancer. 2012;59(2):365–371. doi: 10.1002/pbc.24178 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Charache S, Barton FB, Moore RD, et al. Hydroxyurea and sickle cell anemia. Clinical utility of a myelosuppressive “switching” agent. The Multicenter Study of Hydroxyurea in Sickle Cell Anemia. Medicine (Baltimore). Nov 1996;75(6):300–26. doi: 10.1097/00005792-199611000-00002 [DOI] [PubMed] [Google Scholar]
  • 41.Steinberg MH, Barton F, Castro O, et al. Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment. Jama. Apr 2 2003;289(13):1645–51. doi: 10.1001/jama.289.13.1645 [DOI] [PubMed] [Google Scholar]
  • 42.Lanzkron S, Strouse JJ, Wilson R, et al. Systematic review: Hydroxyurea for the treatment of adults with sickle cell disease. Ann Intern Med. Jun 17 2008;148(12):939–55. doi: 10.7326/0003-4819-148-12-200806170-00221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Agrawal RK, Patel RK, Shah V, Nainiwal L, Trivedi B. Hydroxyurea in sickle cell disease: drug review. Indian J Hematol Blood Transfus. 2014;30(2):91–96. doi: 10.1007/s12288-013-0261-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Boyette LB, Tuan RS. Adult Stem Cells and Diseases of Aging. J Clin Med. 2014;3(1):88–134. doi: 10.3390/jcm3010088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–1217. doi: 10.1016/j.cell.2013.05.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Xu JZ, Thein SL. Revisiting anemia in sickle cell disease and finding the balance with therapeutic approaches. Blood. 2022;139(20):3030–3039. doi: 10.1182/blood.2021013873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Giordano P, Urbano F, Lassandro G, Faienza MF. Mechanisms of Bone Impairment in Sickle Bone Disease. Int J Environ Res Public Health. Feb 13 2021;18(4)doi: 10.3390/ijerph18041832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Park SY, Matte A, Jung Y, et al. Pathologic angiogenesis in the bone marrow of humanized sickle cell mice is reversed by blood transfusion. Blood. Jun 4 2020;135(23):2071–2084. doi: 10.1182/blood.2019002227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Xu JZ, Conrey AK, Frey IC, et al. A Phase 1 Dose Escalation Study of the Pyruvate Kinase Activator Mitapivat (AG-348) in Sickle Cell Disease. Blood. 2022;doi: 10.1182/blood.2022015403 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sherwood JB, Goldwasser E, Chilcote R, Carmichael LD, Nagel RL. Sickle cell anemia patients have low erythropoietin levels for their degree of anemia. Blood. Jan 1986;67(1):46–9. [PubMed] [Google Scholar]
  • 51.Pincez T, Lee SSK, Ilboudo Y, et al. Clonal hematopoiesis in sickle cell disease. Blood. 2021;138(21):2148–2152. doi: 10.1182/blood.2021011121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Liggett LA, Cato LD, Weinstock JS, et al. Clonal hematopoiesis in sickle cell disease. The Journal of Clinical Investigation. February/16/2022;132(4)doi: 10.1172/JCI156060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Brunson A, Keegan THM, Bang H, Mahajan A, Paulukonis S, Wun T. Increased risk of leukemia among sickle cell disease patients in California. Blood. Sep 28 2017;130(13):1597–1599. doi: 10.1182/blood-2017-05-783233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.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. Aug 2016;109(8):303–9. doi: 10.1177/0141076816651037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ruzzon E, Randi ML, Tezza F, Luzzatto G, Scandellari R, Fabris F. Leg ulcers in elderly on hydroxyurea: a single center experience in Phmyeloproliferative disorders and review of literature. Aging Clin Exp Res. Jun 2006;18(3):187–90. doi: 10.1007/bf03324647 [DOI] [PubMed] [Google Scholar]
  • 56.Hellström A, Nilsson C, Nilsson A, Fagerström C. Leg ulcers in older people: a national study addressing variation in diagnosis, pain and sleep disturbance. BMC Geriatrics. 2016/01/21 2016;16(1):25. doi: 10.1186/s12877-016-0198-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Monfort J-B, Senet P Leg Ulcers in Sickle-Cell Disease: Treatment Update. Adv Wound Care (New Rochelle). 2020;9(6):348–356. doi: 10.1089/wound.2018.0918 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Minniti CP, Eckman J, Sebastiani P, Steinberg MH, Ballas SK. Leg ulcers in sickle cell disease. Am J Hematol. Oct 2010;85(10):831–3. doi: 10.1002/ajh.21838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tolu SS, Crouch A, Choi J, et al. Hydroxyurea and fetal hemoglobin effect on leg ulcers in patients with sickle cell disease. Annals of Hematology. 2022/03/01 2022;101(3):541–548. doi: 10.1007/s00277-021-04635-4 [DOI] [PubMed] [Google Scholar]
  • 60.Weinstein JR, Anderson S. The aging kidney: physiological changes. Adv Chronic Kidney Dis. 2010;17(4):302–307. doi: 10.1053/j.ackd.2010.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Pressiat C, Rakotoson MG, Habibi A, et al. Impact of renal function on hydroxyurea exposure in sickle-cell disease patients. Br J Clin Pharmacol. May 2021;87(5):2274–2285. doi: 10.1111/bcp.14653 [DOI] [PubMed] [Google Scholar]
  • 62.Yan JH, Ataga K, Kaul S, et al. The influence of renal function on hydroxyurea pharmacokinetics in adults with sickle cell disease. J Clin Pharmacol. Apr 2005;45(4):434–45. doi: 10.1177/0091270004273526 [DOI] [PubMed] [Google Scholar]
  • 63.Yawn BP, Buchanan GR, Afenyi-Annan AN, et al. Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. Jama. Sep 10 2014;312(10):1033–48. doi: 10.1001/jama.2014.10517 [DOI] [PubMed] [Google Scholar]
  • 64.Nze C, Fortin B, Freedman R, et al. Sudden death in sickle cell disease: current experience. British Journal of Haematology. 2020;188(4):e43–e45. doi: 10.1111/bjh.16314 [DOI] [PubMed] [Google Scholar]
  • 65.Manci EA, Culberson DE, Yang YM, et al. Causes of death in sickle cell disease: an autopsy study. Br J Haematol. Oct 2003;123(2):359–65. doi: 10.1046/j.1365-2141.2003.04594.x [DOI] [PubMed] [Google Scholar]
  • 66.Preston RA, Marbury T, Balaratnam G, et al. Pharmacokinetics of Voxelotor in Patients With Renal and Hepatic Impairment. The Journal of Clinical Pharmacology. 2021;61(4):493–505. doi: 10.1002/jcph.1757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Stevens DL, Hix M, Gildon BL. Crizanlizumab for the Prevention of Vaso-Occlusive Pain Crises in Sickle Cell Disease. J Pharm Technol. Aug 2021;37(4):209–215. doi: 10.1177/87551225211008460 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Brousseau DC, Owens PL, Mosso AL, Panepinto JA, Steiner CA. Acute Care Utilization and Rehospitalizations for Sickle Cell Disease. JAMA. 2010;303(13):1288–1294. doi: 10.1001/jama.2010.378 [DOI] [PubMed] [Google Scholar]
  • 69.Oyedeji C, Strouse JJ, Crawford RD, Garrett ME, Ashley-Koch AE, Telen MJ. A multi-institutional comparison of younger and older adults with sickle cell disease. Am J Hematol. Apr 2019;94(4):E115–e117. doi: 10.1002/ajh.25405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.McClish DK, Smith WR, Levenson JL, et al. Comorbidity, Pain, Utilization, and Psychosocial Outcomes in Older versus Younger Sickle Cell Adults: The PiSCES Project. Biomed Res Int. 2017;2017:4070547. doi: 10.1155/2017/4070547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dufort A, Samaan Z. Problematic Opioid Use Among Older Adults: Epidemiology, Adverse Outcomes and Treatment Considerations. Drugs & Aging. 2021/12/01 2021;38(12):1043–1053. doi: 10.1007/s40266-021-00893-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Smith WR, Penberthy LT, Bovbjerg VE, et al. Daily assessment of pain in adults with sickle cell disease. Ann Intern Med. Jan 15 2008;148(2):94–101. doi: 10.7326/0003-4819-148-2-200801150-00004 [DOI] [PubMed] [Google Scholar]
  • 73.Osunkwo I, O’Connor HF, Saah E. Optimizing the management of chronic pain in sickle cell disease. Hematology. 2020;2020(1):562–569. doi: 10.1182/hematology.2020000143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Oyedeji CI, Hall K, Luciano A, Morey MC, Strouse JJ. The Sickle Cell Disease Functional Assessment (SCD-FA) tool: a feasibility pilot study. Pilot and Feasibility Studies. 2022/03/04 2022;8(1):53. doi: 10.1186/s40814-022-01005-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Carroll CP, Lanzkron S, Haywood C Jr., et al. Chronic Opioid Therapy and Central Sensitization in Sickle Cell Disease. Am J Prev Med. Jul 2016;51(1 Suppl 1):S69–77. doi: 10.1016/j.amepre.2016.02.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.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 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sinha CB, Bakshi N, Ross D, Krishnamurti L. Management of Chronic Pain in Adults Living With Sickle Cell Disease in the Era of the Opioid Epidemic: A Qualitative Study. JAMA Network Open. 2019;2(5):e194410–e194410. doi: 10.1001/jamanetworkopen.2019.4410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.David MS, Jones J, Lauriello A, et al. Converting adults with sickle cell disease from full agonist opioids to buprenorphine: A reliable method with safety and early evidence of reduced acute care utilization. Am J Hematol. Nov 2022;97(11):1435–1442. doi: 10.1002/ajh.26699 [DOI] [PubMed] [Google Scholar]
  • 79.Williams H, Tanabe P. Sickle Cell Disease: A Review of Nonpharmacological Approaches for Pain. J Pain Symptom Manage. Feb 2016;51(2):163–77. doi: 10.1016/j.jpainsymman.2015.10.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Han H, Hensch L, Tubman VN. Indications for transfusion in the management of sickle cell disease. Hematology. 2021;2021(1):696–703. doi: 10.1182/hematology.2021000307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Oyedeji CI, Welsby IJ. Optimizing management of sickle cell disease in patients undergoing surgery. Hematology Am Soc Hematol Educ Program. Dec 10 2021;2021(1):405–410. doi: 10.1182/hematology.2021000274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Menis M, Anderson SA, Forshee RA, et al. Transfusion-associated circulatory overload (TACO) and potential risk factors among the inpatient US elderly as recorded in Medicare administrative databases during 2011. Vox Sang. Feb 2014;106(2):144–52. doi: 10.1111/vox.12070 [DOI] [PubMed] [Google Scholar]
  • 83.Oud JA, Evers D, de Haas M, et al. Transfusion-induced red blood cell alloimmunisation is unhampered in elderly patients. British Journal of Haematology. n/a(n/a) doi: 10.1111/bjh.18374 [DOI] [PubMed] [Google Scholar]
  • 84.Simon GI, Craswell A, Thom O, Fung YL. Outcomes of restrictive versus liberal transfusion strategies in older adults from nine randomised controlled trials: a systematic review and meta-analysis. Lancet Haematol. Oct 2017;4(10):e465–e474. doi: 10.1016/s2352-3026(17)30141-2 [DOI] [PubMed] [Google Scholar]
  • 85.Chou ST, Alsawas M, Fasano RM, et al. American Society of Hematology 2020 guidelines for sickle cell disease: transfusion support. Blood Advances. 2020;4(2):327–355. doi: 10.1182/bloodadvances.2019001143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lee MT, Piomelli S, Granger S, et al. Stroke Prevention Trial in Sickle Cell Anemia (STOP): extended follow-up and final results. Blood. Aug 1 2006;108(3):847–52. doi: 10.1182/blood-2005-10-009506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ware RE, Helms RW. Stroke With Transfusions Changing to Hydroxyurea (SWiTCH). Blood. Apr 26 2012;119(17):3925–32. doi: 10.1182/blood-2011-11-392340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ferreira FA, Benites BD, Costa FF, Gilli S, Olalla-Saad ST. Recombinant erythropoietin as alternative to red cell transfusion in sickle cell disease. Vox Sanguinis. 2019;114(2):178–181. doi: 10.1111/vox.12750 [DOI] [PubMed] [Google Scholar]
  • 89.Little JA, McGowan VR, Kato GJ, et al. Combination erythropoietin-hydroxyurea therapy in sickle cell disease: experience from the National Institutes of Health and a literature review. Haematologica. Aug 2006;91(8):1076–83. [PMC free article] [PubMed] [Google Scholar]
  • 90.Coates TD, Wood JC. How we manage iron overload in sickle cell patients. Br J Haematol. Jun 2017;177(5):703–716. doi: 10.1111/bjh.14575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wilson SR, Sears M, Williams E, et al. Gaps in the diagnosis and management of iron overload in sickle cell disease: a ‘real-world’ report from the GRNDaD registry. Br J Haematol. Dec 2021;195(5):e157–e160. doi: 10.1111/bjh.17762 [DOI] [PubMed] [Google Scholar]
  • 92.Oyedeji CI, Crawford RD, Shah N. Adherence to Iron Chelation Therapy with Deferasirox Formulations among Patients with Sickle Cell Disease and β-thalassemia. Journal of the National Medical Association. 2021/04/01/2021;113(2):170–176. doi: 10.1016/j.jnma.2020.08.007 [DOI] [PubMed] [Google Scholar]
  • 93.Ribeiro LB, Soares EA, Costa FF, Gilli SCO, Olalla Saad ST, Benites BD. The challenges of handling deferasirox in sickle cell disease patients older than 40 years. Hematology. Dec 2019;24(1):596–600. doi: 10.1080/16078454.2019.1657667 [DOI] [PubMed] [Google Scholar]
  • 94.Kapoor E, Strum D, Shim T, Kim S, Sabetrasekh P, Monfared A. Characterization of Sensorineural Hearing Loss in Adult Patients With Sickle Cell Disease: A Systematic Review and Meta-analysis. Otology & Neurotology. 2021;42(1) [DOI] [PubMed] [Google Scholar]
  • 95.Chen SH, Liang DC, Lin HC, Cheng SY, Chen LJ, Liu HC. Auditory and visual toxicity during deferoxamine therapy in transfusion-dependent patients. J Pediatr Hematol Oncol. Dec 2005;27(12):651–3. doi: 10.1097/01.mph.0000194019.95096.b6 [DOI] [PubMed] [Google Scholar]
  • 96.Chiesi Farmaceutici S p.A. Ferriprox (deferiprone) [package insert]. U.S. Food and Drug Administration website. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/021825lbl.pdf. Revised October 2011. Accessed Sepetember 05, 2022. [Google Scholar]
  • 97.Schroeder P, Fulzele K, Forsyth S, et al. Etavopivat, a Pyruvate Kinase Activator in Red Blood Cells, for the Treatment of Sickle Cell Disease. J Pharmacol Exp Ther. Mar 2022;380(3):210–219. doi: 10.1124/jpet.121.000743 [DOI] [PubMed] [Google Scholar]
  • 98.Dufu K, Alt C, Strutt S, et al. GBT021601 Inhibits HbS Polymerization, Prevents RBC Sickling and Improves the Pathophysiology of Sickle Cell Disease in a Murine Model. Blood. 2020;136(Supplement 1):7–8. doi: 10.1182/blood-2020-140539 [DOI] [Google Scholar]
  • 99.Brown RCC, Redfern A, Lisbon E, Washington C, Agodoa I, Smith-Whitley K. GBT021601, a Next Generation HbS Polymerization Inhibitor: Results of Safety, Tolerability, Pharmacokinetics and Pharmacodynamics in Adults Living with Sickle Cell Disease and Healthy Volunteers. Blood. 2021/11/23/2021;138:3099. doi: 10.1182/blood-2021-152924 [DOI] [Google Scholar]
  • 100.Tarasev M, Herppich A, Gao X, Hines P. S107: P-SELECTIN INHIBITOR INCLACUMAB REDUCES CELL ADHESION IN AN IN-VITRO ASSAYS SHOWING POTENTIAL FOR PREVENTION OF VASO-OCCLUSION EVENTS IN SICKLE CELL DISEASE. HemaSphere. 2022;6 [Google Scholar]
  • 101.Schmitt C, Abt M, Ciorciaro C, et al. First-in-Man Study With Inclacumab, a Human Monoclonal Antibody Against P-selectin. J Cardiovasc Pharmacol. Jun 2015;65(6):611–9. doi: 10.1097/fjc.0000000000000233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Geng X, Mihaila R, Yuan Y, et al. Inclacumab, a Fully Human Anti-P-Selectin Antibody, Directly Binds to PSGL-1 Binding Region and Demonstrates Robust and Durable Inhibition of Cell Adhesion. Blood. 2020;136(Supplement 1):10–11. doi: 10.1182/blood-2020-140530 [DOI] [Google Scholar]
  • 103.Mayer C, Cooper DS, Redfern A, et al. Preliminary Results of a Phase 1 Study in Healthy Subjects Administered Inclacumab, a Fully Human IgG4 Anti-P-Selectin Monoclonal Antibody in Development for Treatment of Sickle Cell Disease. Blood. 2021;138(Supplement 1):977–977. doi: 10.1182/blood-2021-15337034192300 [DOI] [Google Scholar]
  • 104.Conran N, Belcher JD. Inflammation in sickle cell disease. Clin Hemorheol Microcirc. 2018;68(2–3):263–299. doi: 10.3233/ch-189012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Mold C, Tamerius JD, Phillips G Jr. Complement activation during painful crisis in sickle cell anemia. Clin Immunol Immunopathol. Sep 1995;76(3 Pt 1):314–20. doi: 10.1006/clin.1995.1131 [DOI] [PubMed] [Google Scholar]
  • 106.Vercellotti GM, Dalmasso AP, Schaid TR Jr., et al. Critical role of C5a in sickle cell disease. Am J Hematol. Mar 2019;94(3):327–337. doi: 10.1002/ajh.25384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Bartolucci P, Ataga KI, Callaghan MU, et al. Trial in Progress: The Randomized, Double-Blind, Placebo-Controlled Phase Ib CROSSWALK-a Trial Evaluating the Safety of Crovalimab for the Management of Acute Uncomplicated Vaso-Occlusive Episodes (VOEs) in Patients with Sickle Cell Disease (SCD). Blood. 2021;138(Supplement 1):3108–3108. doi: 10.1182/blood-2021-147854 [DOI] [Google Scholar]
  • 108.Röth A, Nishimura JI, Nagy Z, et al. The complement C5 inhibitor crovalimab in paroxysmal nocturnal hemoglobinuria. Blood. Mar 19 2020;135(12):912–920. doi: 10.1182/blood.2019003399 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lim SH, Dutta D, Moore J. Rifaximin for sickle cell disease. Am J Hematol. 2019;94(12):E325–E328. doi: 10.1002/ajh.25637 [DOI] [PubMed] [Google Scholar]
  • 110.Dutta D, Methe BA, Morris A, Lim SH. Effects of rifaximin on circulating aged neutrophils in sickle cell disease. Am J Hematol. 2019;94(6):E175–E176. doi: 10.1002/ajh.25467 [DOI] [PubMed] [Google Scholar]
  • 111.Kanter J, Liem RI, Bernaudin F, et al. American Society of Hematology 2021 guidelines for sickle cell disease: stem cell transplantation. Blood Advances. 2021;5(18):3668–3689. doi: 10.1182/bloodadvances.2021004394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Iqbal M, Reljic T, Corbacioglu S, et al. Systematic Review/Meta-Analysis on Efficacy of Allogeneic Hematopoietic Cell Transplantation in Sickle Cell Disease: An International Effort on Behalf of the Pediatric Diseases Working Party of European Society for Blood and Marrow Transplantation and the Sickle Cell Transplantation International Consortium. Transplant Cell Ther. Feb 2021;27(2):167.e1–167.e12. doi: 10.1016/j.jtct.2020.10.007 [DOI] [PubMed] [Google Scholar]
  • 113.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. Jun 2019;25(6):1197–1209. doi: 10.1016/j.bbmt.2018.11.027 [DOI] [PubMed] [Google Scholar]
  • 114.Kanter J, Walters MC, Krishnamurti L, et al. Biologic and Clinical Efficacy of LentiGlobin for Sickle Cell Disease. N Engl J Med. Feb 17 2022;386(7):617–628. doi: 10.1056/NEJMoa2117175 [DOI] [PubMed] [Google Scholar]
  • 115.Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β-Thalassemia. N Engl J Med. Jan 21 2021;384(3):252–260. doi: 10.1056/NEJMoa2031054 [DOI] [PubMed] [Google Scholar]
  • 116.Esrick EB, Lehmann LE, Biffi A, et al. Post-Transcriptional Genetic Silencing of BCL11A to Treat Sickle Cell Disease. N Engl J Med. Jan 21 2021;384(3):205–215. doi: 10.1056/NEJMoa2029392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Jazwinski SM, Kim S. Examination of the Dimensions of Biological Age. Front Genet. 2019;10:263. doi: 10.3389/fgene.2019.00263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Oyedeji CI, Oyesanya T, Strouse JJ. Living Beyond Life Expectancy: Experience with Aging for Older Adults with Sickle Cell Disease. Blood. 2021;138(Supplement 1):492–492. doi: 10.1182/blood-2021-147288 [DOI] [Google Scholar]
  • 119.Kaeberlein M How healthy is the healthspan concept? Geroscience. Aug 2018;40(4):361–364. doi: 10.1007/s11357-018-0036-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Platt OS, Brambilla DJ, Rosse WF, et al. Mortality In Sickle Cell Disease -- Life Expectancy and Risk Factors for Early Death. N Engl J Med. 1994;330(23):1639–1644. doi: 10.1056/nejm199406093302303 [DOI] [PubMed] [Google Scholar]
  • 121.Paneni F, Diaz Cañestro C, Libby P, Lüscher TF, Camici GG. The Aging Cardiovascular System. Understanding It at the Cellular and Clinical Levels. 2017;69(15):1952–1967. doi: 10.1016/j.jacc.2017.01.064 [DOI] [PubMed] [Google Scholar]
  • 122.Almeida A, Roberts I. Bone involvement in sickle cell disease. Br J Haematol. May 2005;129(4):482–90. doi: 10.1111/j.1365-2141.2005.05476.x [DOI] [PubMed] [Google Scholar]
  • 123.Gladwin MT, Sachdev V. Cardiovascular Abnormalities in Sickle Cell Disease. J Am Coll Cardiol. 2012;59(13):1123–1133. doi: 10.1016/j.jacc.2011.10.900 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Cichowitz C, Carroll PC, Strouse JJ, Haywood C Jr., Lanzkron S Utility of the Montreal Cognitive Assessment as a Screening Test for Neurocognitive Dysfunction in Adults with Sickle Cell Disease. South Med J. Sep 2016;109(9):560–5. doi: 10.14423/smj.0000000000000511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Schmidt WP, Roesler A, Kretzschmar K, Ladwig KH, Junker R, Berger K. Functional and cognitive consequences of silent stroke discovered using brain magnetic resonance imaging in an elderly population. J Am Geriatr Soc. Jul 2004;52(7):1045–50. doi: 10.1111/j.1532-5415.2004.52300.x [DOI] [PubMed] [Google Scholar]
  • 126.Steen RG, Emudianughe T, Hankins GM, et al. Brain imaging findings in pediatric patients with sickle cell disease. Radiology. Jul 2003;228(1):216–25. doi: 10.1148/radiol.2281020943 [DOI] [PubMed] [Google Scholar]
  • 127.Scott AW. Ophthalmic Manifestations of Sickle Cell Disease. South Med J. Sep 2016;109(9):542–8. doi: 10.14423/smj.0000000000000525 [DOI] [PubMed] [Google Scholar]
  • 128.Maharani A, Dawes P, Nazroo J, Tampubolon G, Pendleton N. Visual and hearing impairments are associated with cognitive decline in older people. Age Ageing. Jul 1 2018;47(4):575–581. doi: 10.1093/ageing/afy061 [DOI] [PubMed] [Google Scholar]
  • 129.Onakoya PA, Nwaorgu OG, Shokunbi WA. Sensorineural hearing loss in adults with sickle cell anaemia. Afr J Med Med Sci. Mar 2002;31(1):21–4. [PubMed] [Google Scholar]
  • 130.Jakob F, Seefried L, Schwab M. [Age and osteoporosis. Effects of aging on osteoporosis, the diagnostics and therapy]. Internist (Berl). Jul 2014;55(7):755–61. Alter und Osteoporose. Auswirkungen der Alterung auf die Osteoporose, deren Diagnostik und Therapie. doi: 10.1007/s00108-014-3468-z [DOI] [PubMed] [Google Scholar]
  • 131.Dores GM, Devesa SS, Curtis RE, Linet MS, Morton LM. Acute leukemia incidence and patient survival among children and adults in the United States, 2001–2007. Blood. Jan 5 2012;119(1):34–43. doi: 10.1182/blood-2011-04-347872 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Mallappallil M, Friedman EA, Delano BG, McFarlane SI, Salifu MO. Chronic kidney disease in the elderly: evaluation and management. Clinical practice (London, England). 2014;11(5):525–535. doi: 10.2217/cpr.14.46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Adam SS, Flahiff CM, Kamble S, Telen MJ, Reed SD, De Castro LM. Depression, quality of life, and medical resource utilization in sickle cell disease. Blood advances. 2017;1(23):1983–1992. doi: 10.1182/bloodadvances.2017006940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Byers AL, Yaffe K, Covinsky KE, Friedman MB, Bruce ML. High occurrence of mood and anxiety disorders among older adults: The National Comorbidity Survey Replication. Arch Gen Psychiatry. 2010;67(5):489–496. doi: 10.1001/archgenpsychiatry.2010.35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Naik RP, Streiff MB, Haywood C Jr., Nelson JA, Lanzkron S Venous thromboembolism in adults with sickle cell disease: a serious and under-recognized complication. The American journal of medicine. 2013;126(5):443–449. doi: 10.1016/j.amjmed.2012.12.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Tritschler T, Aujesky D. Venous thromboembolism in the elderly: A narrative review. Thromb Res. Jul 2017;155:140–147. doi: 10.1016/j.thromres.2017.05.015 [DOI] [PubMed] [Google Scholar]
  • 137.Reid MC, Eccleston C, Pillemer K. Management of chronic pain in older adults. BMJ (Clinical research ed). 2015;350:h532–h532. doi: 10.1136/bmj.h532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Zimmer Z, Rubin S. Life Expectancy With and Without Pain in the U.S. Elderly Population. The Journals of Gerontology: Series A. 2016;71(9):1171–1176. doi: 10.1093/gerona/glw028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Loeser RF. Age-related changes in the musculoskeletal system and the development of osteoarthritis. Clin Geriatr Med. 2010;26(3):371–386. doi: 10.1016/j.cger.2010.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Golchin N, Frank SH, Vince A, Isham L, Meropol SB. Polypharmacy in the elderly. J Res Pharm Pract. Apr-Jun 2015;4(2):85–8. doi: 10.4103/2279-042x.155755 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Tinetti ME, Speechley M, Ginter SF. Risk factors for falls among elderly persons living in the community. N Engl J Med. Dec 29 1988;319(26):1701–7. doi: 10.1056/nejm198812293192604 [DOI] [PubMed] [Google Scholar]
  • 142.Pilleron S, Sarfati D, Janssen-Heijnen M, et al. Global cancer incidence in older adults, 2012 and 2035: A population-based study. Int J Cancer. Jan 1 2019;144(1):49–58. doi: 10.1002/ijc.31664 [DOI] [PubMed] [Google Scholar]
  • 143.Parker SG, McCue P, Phelps K, et al. What is Comprehensive Geriatric Assessment (CGA)? An umbrella review. Age Ageing. Jan 1 2018;47(1):149–155. doi: 10.1093/ageing/afx166 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated or analyzed during the current study.

RESOURCES