• Open access
  • Published: 03 March 2022

Advances in the diagnosis and treatment of sickle cell disease

  • A. M. Brandow 1 &
  • R. I. Liem   ORCID: orcid.org/0000-0003-2057-3749 2  

Journal of Hematology & Oncology volume  15 , Article number:  20 ( 2022 ) Cite this article

38k Accesses

65 Citations

2 Altmetric

Metrics details

Sickle cell disease (SCD), which affects approximately 100,000 individuals in the USA and more than 3 million worldwide, is caused by mutations in the βb globin gene that result in sickle hemoglobin production. Sickle hemoglobin polymerization leads to red blood cell sickling, chronic hemolysis and vaso-occlusion. Acute and chronic pain as well as end-organ damage occur throughout the lifespan of individuals living with SCD resulting in significant disease morbidity and a median life expectancy of 43 years in the USA. In this review, we discuss advances in the diagnosis and management of four major complications: acute and chronic pain, cardiopulmonary disease, central nervous system disease and kidney disease. We also discuss advances in disease-modifying and curative therapeutic options for SCD. The recent availability of l -glutamine, crizanlizumab and voxelotor provides an alternative or supplement to hydroxyurea, which remains the mainstay for disease-modifying therapy. Five-year event-free and overall survival rates remain high for individuals with SCD undergoing allogeneic hematopoietic stem cell transplant using matched sibling donors. However, newer approaches to graft-versus-host (GVHD) prophylaxis and the incorporation of post-transplant cyclophosphamide have improved engraftment rates, reduced GVHD and have allowed for alternative donors for individuals without an HLA-matched sibling. Despite progress in the field, additional longitudinal studies, clinical trials as well as dissemination and implementation studies are needed to optimize outcomes in SCD.

Introduction

Sickle cell disease (SCD), a group of inherited hemoglobinopathies characterized by mutations that affect the β-globin chain of hemoglobin, affects approximately 100,000 people in the USA and more than 3 million people worldwide [ 1 , 2 ]. SCD is characterized by chronic hemolytic anemia, severe acute and chronic pain as well as end-organ damage that occurs across the lifespan. SCD is associated with premature mortality with a median age of death of 43 years (IQR 31.5–55 years) [ 3 ]. Treatment requires early diagnosis, prevention of complications and management of end-organ damage. In this review, we discuss recent advances in the diagnosis and management of four major complications in SCD: acute and chronic pain, cardiopulmonary disease, central nervous system disease and kidney disease. Updates in disease-modifying and curative therapies for SCD are also discussed.

Molecular basis and pathophysiology

Hemoglobin S (HbS) results from the replacement of glutamic acid by valine in the sixth position of the β-globin chain of hemoglobin (Fig.  1 ). Severe forms of SCD include hemoglobin SS due to homozygous inheritance of HbS and S/β 0 thalassemia due to co-inheritance of HbS with the β 0 thalassemia mutation. Other forms include co-inheritance of HbS with other β-globin gene mutations such as hemoglobin C, hemoglobin D-Los Angeles/Punjab or β + thalassemia. Hb S has reduced solubility and increased polymerization, which cause red blood cell sickling, hemolysis and vaso-occlusion (Table 1 ) that subsequently lead to pain episodes and end-organ damage such as cardiopulmonary, cerebrovascular and kidney disease (Table 2 ).

figure 1

Genetic and molecular basis of sickle cell disease. SCD is caused by mutations in the β globin gene, located on the β globin locus found on the short arm of chromosome 11. The homozygous inheritance of Hb S or co-inheritance of Hb S with the β 0 thalassemia mutation results in the most common forms of severe SCD. Co-inheritance of Hb S with other variants such as Hb C, Hb D-Los Angeles/Punjab, Hb O-Arab or β + thalassemia also leads to clinically significant sickling syndromes (LCR, locus control region; HS, hypersensitivity site)

Acute and chronic pain

Severe intermittent acute pain is the most common SCD complication and accounts for over 70% of acute care visits for individuals with SCD [ 4 ]. Chronic daily pain increases with older age, occurring in 30–40% of adolescents and adults with SCD [ 5 , 6 ]. Acute pain is largely related to vaso-occlusion of sickled red blood cells with ischemia–reperfusion injury and tissue infarction and presents in one isolated anatomic location (e.g., arm, leg, back) or multiple locations. Chronic pain can be caused by sensitization of the central and/or peripheral nervous system and is often diffuse with neuropathic pain features [ 7 , 8 ]. A consensus definition for chronic pain includes “Reports of ongoing pain on most days over the past 6 months either in a single location or multiple locations” [ 9 ]. Disease complications such as avascular necrosis (hip, shoulder) and leg ulcers also cause chronic pain [ 9 ].

Diagnosis of acute and chronic pain

The gold standard for pain assessment and diagnosis is patient self-report. There are no reliable diagnostic tests to confirm the presence of acute or chronic pain in individuals with SCD except when there are identifiable causes like avascular necrosis on imaging or leg ulcers on exam. The effects of pain on individuals’ function are assessed using patient-reported outcome measures (PROs) that determine to what extent pain interferes with individuals’ daily function. Tools shown to be valid, reliable and responsive can be used in clinical practice to track patients’ pain-related function over time to determine additional treatment needs and to compare to population norms [ 10 ]. There are currently no plasma pain biomarkers that improve assessment and management of SCD acute or chronic pain.

Depression and anxiety as co-morbid conditions in SCD can contribute to increased pain, more pain-related distress/interference and poor coping [ 11 ]. The prevalence of depression and anxiety range from 26–33% and 6.5–36%, respectively, in adults with SCD [ 11 , 12 , 13 ]. Adults with SCD have an 11% higher prevalence of depression compared to Black American adults without SCD [ 14 ]. Depression and anxiety can be assessed using self-reported validated screening tools (e.g., Depression: Patient Health Questionnaire (PHQ-9) [ 15 ] for adults, Center for Epidemiologic Studies Depression Scale for Children (CES-DC) [ 16 ], PROMIS assessments for adults and children; Anxiety: Generalized Anxiety Disorder 7-item (GAD-7) scale for adults, State-Trait Anxiety Inventory for Children (STAIC) [ 17 ], PROMIS assessments for adults and children). Individuals who screen positive using these tools should be referred for evaluation by a psychologist/psychiatrist.

Management of acute and chronic pain

The goal of acute pain management is to provide sufficient analgesia to return patients to their usual function, which may mean complete resolution of pain for some or return to baseline chronic pain for others. The goal of chronic pain management is to optimize individuals’ function, which may not mean being pain free. When there is an identifiable cause of chronic pain, treatment of the underlying issue (e.g., joint replacement for avascular necrosis, leg ulcer treatment) is important. Opioids, oral for outpatient management and intravenous for inpatient management, are first line therapy for acute SCD pain. In the acute care setting, analgesics should be initiated within 30–60 min of triage [ 18 ]. Ketamine, a non-opioid analgesic, can be prescribed at sub-anesthetic (analgesic) intravenous doses (0.1–0.3 mg/kg per h, maximum 1 mg/kg per h) as adjuvant treatment for acute SCD pain refractory to opioids [ 18 , 19 ]. In an uncontrolled observational study of 85 patients with SCD receiving ketamine infusions for acute pain, ketamine was associated with a decrease in mean opioid consumption by oral morphine equivalents (3.1 vs. 2.2 mg/kg/day, p  < 0.001) and reductions in mean pain scores (0–10 scale) from baseline until discontinuation of the infusion (7.81 vs. 5.44, p  < 0.001) [ 20 ]. Nonsteroidal anti-inflammatory drugs (NSAIDs) are routinely used as adjuvant therapy for acute pain treatment [ 18 ]. In a RCT ( n  = 20) of hospitalized patients with acute pain, ketorolac was associated with lower total dose of meperidine required (1866.7 ± 12.4 vs. 2804.5 ± 795.1 mg, p  < 0.05) and shorter hospitalization (median 3.3 vs. 7.2 days, p  = 0.027) [ 21 ]. In a case series of children treated for 70 acute pain events in the ED, 53% of events resolved with ketorolac and hydration alone with reduction in 100 mm visual analog scale (VAS) pain score from 60 to 13 ( p  < 0.001) [ 22 ]. Patients at risk for NSAID toxicity (e.g., renal impairment, on anticoagulation) should be identified.

Despite paucity of data, chronic opioid therapy (COT) can be considered after assessing benefits versus harms [ 23 ] and the functional status of patients with SCD who have chronic pain. Harms of COT seen in patient populations other than SCD are dose dependent and include myocardial infarction, bone fracture, increased risk of motor vehicle collisions, sexual dysfunction and mortality [ 23 ]. There are few published studies investigating non-opioid analgesics for chronic SCD pain [ 24 , 25 , 26 ]. In a randomized trial of 39 participants, those who received Vitamin D experienced a range of 6–10 pain days over 24 weeks while those who received placebo experienced 10–16 pain days, which was not significantly different [ 26 ]. In a phase 1, uncontrolled trial of 18 participants taking trifluoperazine, an antipsychotic drug, 8 participants showed a 50% reduction in the VAS (10 cm horizontal line) pain score from baseline on at least 3 assessments over 24 h without severe sedation or supplemental opioid analgesics, 7 participants showed pain reduction on 1 assessment, and the remaining 3 participants showed no reduction [ 24 ]. Although published data are not available for serotonin and norepinephrine reuptake inhibitors (SNRIs), gabapentinoids and tricyclic antidepressants (TCAs) in individuals with SCD, evidence supports their use in fibromyalgia, a chronic pain condition similar to SCD chronic pain in mechanism. A Cochrane Review that included 10 RCTs ( n  = 6038) showed that the SNRIs milnacipran and duloxetine, compared to placebo, were associated with a reduction in pain [ 27 ]. A systematic review and meta-analysis of 9 studies ( n  = 520) showed the TCA amitriptyline improved pain intensity and function [ 28 ]. Finally, a meta-analysis of 5 RCTs ( n  = 1874) of the gabapentinoid pregabalin showed a reduction in pain intensity [ 29 ]. Collectively, the indirect evidence from fibromyalgia supports the conditional recommendation in current SCD practice guidelines to consider these 3 drug classes for chronic SCD pain treatment [ 18 ]. Standard formulary dosing recommendations should be followed and reported adverse effects considered.

Non-pharmacologic therapies (e.g., integrative, psychological-based therapies) are important components of SCD pain treatment. In a case–control study of 101 children with SCD and chronic pain referred for cognitive behavioral therapy (CBT) (57 CBT, 44 no CBT) [ 30 ], CBT was associated with more rapid decrease in pain hospitalizations (estimate − 0.63, p  < 0.05) and faster reduction in hospital days over time (estimate − 5.50, p  < 0.05). Among 18 children who received CBT and completed PROs pre- and 12 months posttreatment, improvements were seen in mean pain intensity (5.47 vs. 3.76, p  = 0.009; 0–10 numeric rating pain scale), functional disability (26.24 vs. 15.18, p  < 0.001; 0–60 score range) and pain coping (8.00 vs. 9.65, p  = 0.03; 3–15 score range) post treatment [ 30 ]. In 2 uncontrolled clinical trials, acupuncture was associated with a significant reduction in pain scores by 2.1 points (0–10 numeric pain scale) in 24 participants immediately after treatment [ 31 ] or a significant mean difference in pre-post pain scores of 0.9333 (0–10 numeric pain scale) ( p  < 0.000) after 33 acupuncture sessions [ 32 ].

Cardiopulmonary disease

Cardiopulmonary disease is associated with increased morbidity and mortality in individuals with SCD. Pulmonary hypertension (PH), most commonly pulmonary arterial hypertension (PAH), is present based on right-heart catheterization in up to 10% of adults with SCD [ 33 ]. Chronic intravascular hemolysis represents the biggest risk factor for development of PAH in SCD and leads to pulmonary arteriole vasoconstriction and smooth muscle proliferation. Based on pulmonary function testing (PFT), obstructive lung disease may be observed in 16% of children and 8% of adults with SCD, while restrictive lung disease may be seen in up to 28% of adults and only 7% of children with SCD [ 34 , 35 ]. Sleep-disordered breathing, which can manifest as obstructive sleep apnea or nocturnal hypoxemia, occurs in up to 42% of children and 46% of adults with SCD [ 36 , 37 ]. Cardiopulmonary disease, including PH or restrictive lung disease, presents with dyspnea with or without exertion, chest pain, hypoxemia or exercise intolerance that is unexplained or increased from baseline. Obstructive lung disease can also present with wheezing.

Diagnosis of cardiopulmonary disease

The confirmation of PH in patients with SCD requires right-heart catheterization. Recently, the mean pulmonary artery pressure threshold used to define PH in the general population was lowered from ≥ 25 to ≥ 20 mm Hg [ 38 ]. Elevated peak tricuspid regurgitant jet velocity (TRJV) ≥ 2.5 m/s on Doppler echocardiogram (ECHO) is associated with early mortality in adults with SCD and may suggest elevated pulmonary artery pressures, especially when other signs of PH (e.g., right-heart strain, septal flattening) or left ventricular diastolic dysfunction, which may contribute to PH, are present [ 39 ]. However, the positive predictive value (PPV) of peak TRJV alone for identifying PH in adults with SCD is only 25% [ 40 ]. Increasing the peak TRJV threshold to at least 2.9 m/s has been shown to increase the PPV to 64%. For a peak TRJV of 2.5–2.8 m/s, an increased N-terminal pro-brain natriuretic peptide (NT-proBNP) ≥ 164.5 pg/mL or a reduced 6-min walk distance (6MWD) < 333 m can also improve the PPV to 62% with a false negative rate of 7% [ 33 , 40 , 41 ].

PFT, which includes spirometry and measurement of lung volumes and diffusion capacity, is standard for diagnosing obstructive and restrictive lung disease in patients with SCD. Emerging modalities include impulse oscillometry, a non-invasive method using forced sound waves to detect changes in lower airway mechanics in individuals unable to perform spirometry [ 42 ], and airway provocation studies using cold air or methacholine to reveal latent airway hyperreactivity [ 43 ]. Formal in-lab, sleep study/polysomnography remains the gold standard to evaluate for sleep-disordered breathing, which may include nocturnal hypoxemia, apnea/hypopnea events and other causes of sleep disruption. Nocturnal hypoxemia may increase red blood cell sickling, cellular adhesion and endothelial dysfunction. In 47 children with SCD, mean overnight oxygen saturation was higher in those with grade 0 compared to grade 2 or 3 cerebral arteriopathy (97 ± 1.6 vs. 93.9 ± 3.7 vs. 93.5 ± 3.0%, p  < 0.01) on magnetic resonance angiography and lower overnight oxygen saturation was independently associated with mild, moderate or severe cerebral arteriopathy after adjusting for reticulocytosis (OR 0.50, 95% CI 0.26–0.96, p  < 0.05) [ 44 ].

Management of cardiopulmonary disease

Patients with SCD who have symptoms suggestive of cardiopulmonary disease, such as worsening dyspnea, hypoxemia or reduced exercise tolerance, should be evaluated with a diagnostic ECHO and PFT. The presence of snoring, witnessed apnea, respiratory pauses or hypoxemia during sleep, daytime somnolence or nocturnal enuresis in older children and adults is sufficient for a diagnostic sleep study.

Without treatment, the mortality rate in SCD patients with PH is high compared to those without (5-year, all-cause mortality rate of 32 vs. 16%, p  < 0.001) [ 33 ]. PAH-targeted therapies should be considered for SCD patients with PAH confirmed by right-heart catheterization. However, the only RCT ( n  = 6) in individuals with SCD and PAH confirmed by right-heart catheterization (bosentan versus placebo) was stopped early for poor accrual with no efficacy endpoints analyzed [ 45 ]. In SCD patients with elevated peak TRJV, a randomized controlled trial ( n  = 74) of sildenafil, a phosphodiesterase-5 inhibitor, was discontinued early due to increased pain events in the sildenafil versus placebo arm (35 vs. 14%, p  = 0.029) with no treatment benefit [ 46 ]. Despite absence of clinical trial data, patients with SCD and confirmed PH should be considered for hydroxyurea or monthly red blood cell transfusions given their disease-modifying benefits. In a retrospective analysis of 13 adults with SCD and PAH, 77% of patients starting at a New York Heart Association (NYHA) functional capacity class III or IV achieved class I/II after a median of 4 exchange transfusions with improvement in median pulmonary vascular resistance (3.7 vs. 2.8 Wood units, p  = 0.01) [ 47 ].

Approximately 28% of children with SCD have asthma, which is associated with increased pain episodes that may result from impaired oxygenation leading to sickling and vaso-occlusion as well as with acute chest syndrome and higher mortality [ 48 , 49 , 50 ]. First line therapies include standard beta-adrenergic bronchodilators and supplemental oxygen as needed. When corticosteroids are indicated, courses should be tapered over several days given the risk of rebound SCD pain from abrupt discontinuation. Inhaled corticosteroids such as fluticasone proprionate or beclomethasone diproprionate are reserved for patients with recurrent asthma exacerbations, but their anti-inflammatory effects and impact on preventing pain episodes in patients with SCD who do not have asthma is under investigation [ 51 ]. Finally, management of sleep-disordered breathing is tailored to findings on formal sleep study in consultation with a sleep/pulmonary specialist.

Central nervous system (CNS) complications

CNS complications, such as overt and silent cerebral infarcts, cause significant morbidity in individuals with SCD. Eleven percent of patients with HbSS disease by age 20 years and 24% by age 45 years will have had an overt stroke [ 52 ]. Silent cerebral infarcts occur in 39% by 18 years and in > 50% by 30 years [ 53 , 54 ]. Patients with either type of stroke are at increased risk of recurrent stroke [ 55 ]. Overt stroke involves large-arteries, including middle cerebral arteries and intracranial internal carotid arteries, while silent cerebral infarcts involve penetrating arteries. The pathophysiology of overt stroke includes vasculopathy, increased sickled red blood cell adherence, and hemolysis-induced endothelial activation and altered vasomotor tone [ 56 ]. Overt strokes present as weakness or paresis, dysarthria or aphasia, seizures, sensory deficits, headache or altered level of consciousness, while silent cerebral infarcts are associated with cognitive deficits, including lower IQ and impaired academic performance.

Diagnosis of CNS complications in SCD

Overt stroke is diagnosed by evidence of acute infarct on brain MRI diffusion-weighted imaging and focal deficit on neurologic exam. A silent cerebral infarct is defined by a brain “MRI signal abnormality at least 3 mm in one dimension and visible in 2 planes on fluid-attenuated inversion recovery (FLAIR) T2-weighted images” and no deficit on neurologic exam [ 57 ]. Since silent cerebral infarcts cannot be detected clinically, a screening baseline brain MRI is recommended in school-aged children with SCD [ 58 ]. Recent SCD clinical practice guidelines also suggest a screening brain MRI in adults with SCD to facilitate rehabilitation services, patient and family understanding of cognitive deficits and further needs assessment [ 58 ]. An MRA should be added to screening/diagnostic MRIs to evaluate for cerebral vasculopathy (e.g., moyamoya), which may increase risk for recurrent stroke or hemorrhage [ 59 ].

Annual screening for increased stroke risk by transcranial doppler (TCD) ultrasound is recommended by the American Society of Hematology for children 2–16 years old with HbSS or HbS/β° thalassemia [ 58 ]. Increased stroke risk on non-imaging TCD is indicated by abnormally elevated cerebral blood flow velocity, defined as ≥ 200 cm/s (time-averaged mean of the maximum velocity) on 2 occasions or a single velocity of > 220 cm/s in the distal internal carotid or proximal middle cerebral artery [ 60 ]. Many centers rely on imaging TCD, which results in velocities 10–15% lower than values obtained by non-imaging protocols and therefore, require adjustments to cut-offs for abnormal velocities. Data supporting stroke risk assessment using TCD are lacking for adults with SCD and standard recommendations do not exist.

Neurocognitive deficits occur in over 30% of children and adults with severe SCD [ 61 , 62 ]. These occur as a result of overt and/or silent cerebral infarcts but in some patients, the etiology is unknown. The Bright Futures Guidelines for Health Supervision of Infants, Children and Adolescents or the Cognitive Assessment Toolkit for adults are commonly used tools to screen for developmental delays or neurocognitive impairment [ 58 ]. Abnormal results should prompt referral for formal neuropsychological evaluation, which directs the need for brain imaging to evaluate for silent cerebral infarcts and facilitate educational/vocational accommodations.

Management of CNS complications

Monthly chronic red blood cell transfusions to suppress HbS < 30% are standard of care for primary stroke prevention in children with an abnormal TCD. In an RCT of 130 children, chronic transfusions, compared to no transfusions, were associated with a difference in stroke risk of 92% (1 vs. 10 strokes, p  < 0.001) [ 60 ]. However, children with abnormal TCD and no MRI/MRA evidence of cerebral vasculopathy can safely transition to hydroxyurea after 1 year of transfusions [ 63 ]. Lifelong transfusions to maintain HbS < 30% remain standard of care for secondary stroke prevention in individuals with overt stroke [ 64 ]. Chronic monthly red blood cell transfusions should also be considered for children with silent cerebral infarct [ 58 ]. In a randomized controlled trial ( n  = 196), monthly transfusions, compared to observation without hydroxyurea, reduced risk of overt stroke, new silent cerebral infarct or enlarging silent cerebral infarct in children with HbSS or HbS/β 0 thalassemia and an existing silent cerebral infarct (2 vs. 4.8 events, incidence rate ratio of 0.41, 95% CI 0.12–0.99, p  = 0.04) [ 57 ].

Acute stroke treatment requires transfusion therapy to increase cerebral oxygen delivery. Red blood cell exchange transfusion, defined as replacement of patients’ red blood cells with donor red blood cells, to rapidly reduce HbS to < 30% is the recommended treatment as simple transfusion alone is shown to have a fivefold greater relative risk (57 vs. 21% with recurrent stroke, RR = 5.0; 95% CI 1.3–18.6) of subsequent stroke compared to exchange transfusion [ 65 ]. However, a simple transfusion is often given urgently while preparing for exchange transfusion [ 58 ]. Tissue plasminogen activator (tPA) is not recommended for children with SCD who have an acute stroke since the pathophysiology of SCD stroke is less likely to be thromboembolic in origin and there is risk for harm. Since the benefits and risks of tPA in adults with SCD and overt stroke are not clear, its use depends on co-morbidities, risk factors and stroke protocols but should not delay or replace prompt transfusion therapy.

Data guiding treatment of SCD cerebral vasculopathy (e.g., moyamoya) are limited, and only nonrandomized, low-quality evidence exists for neurosurgical interventions (e.g., encephaloduroarteriosynangiosis) [ 66 ]. Consultation with a neurosurgeon to discuss surgical options in patients with moyamoya and history of stroke or transient ischemic attack should be considered [ 58 ].

Kidney disease

Glomerulopathy, characterized by hyperfiltration leading to albuminuria, is an early asymptomatic manifestation of SCD nephropathy and worsens with age. Hyperfiltration, defined by an absolute increase in glomerular filtration rate, may be seen in 43% of children with SCD [ 67 ]. Albuminuria, defined by the presence of urine albumin ≥ 30 mg/g over 24 h, has been observed in 32% of adults with SCD [ 68 ]. Glomerulopathy results from intravascular hemolysis and endothelial dysfunction in the renal cortex. Medullary hypoperfusion and ischemia also contribute to kidney disease in SCD, causing hematuria, urine concentrating defects and distal tubular dysfunction [ 69 ]. Approximately 20–40% of adults with SCD develop chronic kidney disease (CKD) and are at risk of developing end-stage renal disease (ESRD), with rapid declines in estimated glomerular filtration rate (eGFR) > 3 mL/min/1.73 m 2 associated with increased mortality (HR 2.4, 95% CI 1.31–4.42, p  = 0.005) [ 68 ].

Diagnosis of kidney disease in SCD

The diagnosis of sickle cell nephropathy is made by detecting abnormalities such as albuminuria, hematuria or CKD rather than by distinct diagnostic criteria in SCD, which have not been developed. Traditional markers of kidney function such as serum creatinine and eGFR should be interpreted with caution in individuals with SCD because renal hyperfiltration affects their accuracy by increasing both. Practical considerations preclude directly measuring GFR by urine or plasma clearance techniques, which achieves the most accurate results. The accuracy of eGFR, however, may be improved by equations that incorporate serum cystatin C [ 70 ].

Since microalbuminuria/proteinuria precedes CKD in SCD, annual screening for urine microalbumin/protein is recommended beginning at age 10 years [ 71 ]. When evaluating urine for microalbumin concentration, samples from first morning rather than random voids are preferable to exclude orthostatic proteinuria. Recent studies suggest HMOX1 and APOL1 gene variants may be associated with CKD in individuals with SCD [ 72 ]. Potential novel predictors of acute kidney injury in individuals with SCD include urine biomarkers kidney injury molecule 1 (KIM-1) [ 73 ], monocyte chemotactic protein 1 (MCP-1) [ 74 ] and neutrophil gelatinase-associated lipocalin (NGAL) [ 75 ]. Their contribution to chronic kidney disease and interaction with other causes of kidney injury in SCD (e.g., inflammation, hemolysis) are not clear.

Management of kidney disease

Managing kidney complications in SCD should focus on mitigating risk factors for acute and chronic kidney injury such as medication toxicity, reduced kidney perfusion from hypotension and dehydration, and general disease progression, as well as early screening and treatment of microalbuminuria/proteinuria. Acute kidney injury, either an increase in serum creatinine ≥ 0.3 mg/dL or a 50% increase in serum creatinine from baseline, is associated with ketorolac use in children with SCD hospitalized for pain [ 76 ]. Increasing intravenous fluids to maintain urine output > 0.5 to 1 mL/kg/h and limiting NSAIDs and antibiotics associated with nephrotoxicity in this setting are important. Despite absence of controlled clinical trials, hydroxyurea may be associated with improvements in glomerular hyperfiltration and urine concentrating ability in children with SCD [ 77 , 78 ]. Hydroxyurea is also associated with a lower prevalence (34.7 vs. 55.4%, p  = 0.01) and likelihood of albuminuria (OR 0.28, 95% CI 0.11–0.75, p  = 0.01) in adults with SCD after adjusting for age, angiotensin-converting enzyme inhibitor (ACE-I)/angiotensin receptor blockade (ARB) use and major disease risk factors [ 79 ].

ACE-I or ARB therapy reduces microalbuminuria in patients with SCD. In a phase 2 trial of 36 children and adults, a ≥ 25% reduction in urine albumin-to-creatinine ratio was observed in 83% ( p  < 0.0001) and 58% ( p  < 0.0001) of patients with macroalbuminuria (> 300 mg/g creatinine) and microalbuminuria (30–300 mg/g creatinine), respectively, after 6 months of treatment with losartan at a dose of 0.7 mg/kg/day (max of 50 mg) in children and 50 mg daily in adults [ 80 ]. However, ACE-I or ARB therapy has not been shown to improve kidney function or prevent CKD. Hemodialysis is associated with a 1-year mortality rate of 26.3% after starting hemodialysis and an increase risk of death in SCD patients with ESRD compared to non-SCD patients with ESRD (44.6 vs. 34.5% deaths, mortality hazard ratio of 2.8, 95% CI 2.31–3.38) [ 81 ]. Renal transplant should be considered for individuals with SCD and ESRD because of recent improvements in renal graft survival and post-transplant mortality [ 82 ].

Disease-modifying therapies in SCD

Since publication of its landmark trial in 1995, hydroxyurea continues to represent a mainstay of disease-modifying therapy for SCD. Hydroxyurea induces fetal hemoglobin production through stress erythropoiesis, reduces inflammation, increases nitric oxide and decreases cell adhesion. The FDA approved hydroxyurea in 1998 for adults with SCD. Subsequently, hydroxyurea was FDA approved for children in 2017 to reduce the frequency pain events and need for blood transfusions in children ≥ 2 years of age [ 63 ]. The landscape of disease-modifying therapies, however, has improved with the recent FDA approval of 3 other treatments— l -glutamine and crizanlizumab for reducing acute complications (e.g., pain), and voxelotor for improving anemia (Table 3 ) [ 83 , 84 , 85 ]. Other therapies in current development focus on inducing fetal hemoglobin, reducing anti-sickling or cellular adhesion, or activating pyruvate kinase-R.

l -glutamine

Glutamine is required for the synthesis of glutathione, nicotinamide adenine dinucleotide and arginine. The essential amino acid protects red blood cells against oxidative damage, which forms the basis for its proposed utility in SCD. The exact mechanism of benefit in SCD, however, remains unclear. In a phase 3 RCT of 230 participants (hemoglobin SS or S/β 0 thalassemia), l -glutamine compared to placebo was associated with fewer pain events (median 3 vs. 4, p  = 0.005) and hospitalizations for pain (median 2 vs. 3, p  = 0.005) over the 48-week treatment period [ 84 ]. The percentage of patients who had at least 1 episode of acute chest syndrome, defined as presence of chest wall pain with fever and a new pulmonary infiltrate, was lower in the l -glutamine group (8.6 vs. 23.1%, p  = 0.003). There were no significant between-group differences in hemoglobin, hematocrit or reticulocyte count. Common side effects of l -glutamine include GI upset (constipation, nausea, vomiting and abdominal pain) and headaches.

Crizanlizumab

P-selectin expression, triggered by inflammation, promotes adhesion of neutrophils, activated platelets and sickle red blood cells to the endothelial surface and to each other, which promotes vaso-occlusion in SCD. Crizanlizumab, given as a monthly intravenous infusion, is a humanized monoclonal antibody that binds P-selectin and blocks the adhesion molecule’s interaction with its ligand, P-selectin glycoprotein ligand 1. FDA approval for crizanlizumab was based on a phase 2 RCT ( n  = 198, all genotypes), in which the median rate of pain events (primary endpoint) was lower (1.63 vs. 2.68, p  = 0.01) and time to first pain event (secondary endpoint) was longer (4.07 vs. 1.38 months, p  = 0.001) for patients on high-dose crizanlizumab (5 mg/kg/dose) compared to placebo treated for 52 weeks (14 doses total) [ 83 ]. In this trial, patients with SCD on chronic transfusion therapy were excluded, but those on stable hydroxyurea dosing were not. Adverse events were uncommon but included headache, back pain, nausea, arthralgia and pain in the extremity.

Polymerization of Hb S in the deoxygenated state represents the initial step in red blood cell sickling, which leads to reduced red blood cell deformability and increased hemolysis. Voxelotor is a first-in-class allosteric modifier of Hb S that increases oxygen affinity. The primary endpoint for the phase 3 RCT of voxelotor ( n  = 274, all genotypes) that led to FDA approval was an increase in hemoglobin of at least 1 g/dL after 24 weeks of treatment [ 85 ]. More participants receiving 1500 mg daily of oral voxelotor versus placebo had a hemoglobin response of at least 1 g/dL (51%, 95% CI 41–61 vs. 7%, 95% CI 1–12, p < 0.001). Approximately 2/3 of the participants in these trials were on hydroxyurea, with treatment benefits observed regardless of hydroxyurea status. Despite improvements associated with voxelotor in biomarkers of hemolysis (reticulocyte count, indirect bilirubin and lactate dehydrogenase), annualized incidence rate of vaso-occlusive crisis was not significantly different among treatment groups. Adverse events included headaches, GI symptoms, arthralgia, fatigue and rash.

Curative therapies in SCD

For individuals with SCD undergoing hematopoietic stem cell transplantation (HSCT) using HLA-matched sibling donors and either myeloablative or reduced-intensity conditioning regimens, the five-year event-free and overall survival is high at 91% and 93%, respectively [ 86 ]. Limited availability of HLA-matched sibling donors in this population requires alternative donors or the promise of autologous strategies such as gene-based therapies (i.e. gene addition, transfer or editing) (Table 4 ). Matched unrelated donors have not been used routinely due to increased risk of graft-versus-host disease (GVHD) as high as 19% (95% CI 12–28) in the first 100 days for acute GVHD and 29% (95% CI 21–38) over 3 years for chronic GVHD [ 87 ]. Haplo-identical HSCT, using biological parents or siblings as donors, that incorporate post-transplant cyclophosphamide demonstrates acceptable engraftment rates, transplant-related morbidity and overall mortality [ 88 ]. Regardless of allogeneic HSCT type, older age is associated with lower event-free (102/418 vs. 72/491 events, HR 1.74, 95% CI 1.24–2.45) and overall survival (54/418 vs. 22/491 events, HR 3.15, 95% CI 1.86–5.34) in patients ≥ 13 years old compared to < 12 years old undergoing HSCT [ 87 ].

Advancing research in SCD

Despite progress to date, additional high-quality, longitudinal data are needed to better understand the natural history of the disease and to inform optimal screening for SCD-related complications. In the era of multiple FDA-approved therapies with disease-modifying potential, clinical trials to evaluate additional indications and test them in combination with or compared to each other are needed. Dissemination and implementation studies are also needed to identify barriers and facilitators related to treatment in everyday life, which can be incorporated into decision aids and treatment algorithms for patients and their providers [ 89 ]. Lastly, continued efforts should acknowledge social determinants of health and other factors that affect access and disease-related outcomes such as the role of third-party payers, provider and patient education, health literacy and patient trust. Establishing evidence-derived quality of care metrics can also drive public policy changes required to ensure care optimization for this population.

Conclusions

SCD is associated with complications that include acute and chronic pain as well as end-organ damage such as cardiopulmonary, cerebrovascular and kidney disease that result in increased morbidity and mortality. Several well-designed clinical trials have resulted in key advances in management of SCD in the past decade. Data from these trials have led to FDA approval of 3 new drugs, l -glutamine, crizanlizumab and voxelotor, which prevent acute pain and improve chronic anemia. Moderate to high-quality data support recommendations for managing SCD cerebrovascular disease and early kidney disease. However, further research is needed to determine the best treatment for chronic pain and cardiopulmonary disease in SCD. Comparative effectiveness research, dissemination and implementation studies and a continued focus on social determinants of health are also essential.

Availability of data and materials

Not applicable.

Abbreviations

Six-minute walk distance

Angiotensin-converting enzyme inhibitor

Angiotensin receptor blockade

Cognitive behavioral therapy

Chronic kidney disease

Chronic opioid therapy

Echocardiogram

End stage renal disease

Fluid-attenuated inversion recovery

Glomerular filtration rate

Graft-versus-host disease

Hemoglobin S

Hematopoietic stem cell transplant

Nonsteroidal anti-inflammatory drugs

N-terminal pro-brain natriuretic peptide

New York Heart Association

Pulmonary arterial hypertension

Pulmonary function test

Pulmonary hypertension

Positive predictive value

Patient-reported outcomes

Randomized controlled trial

  • Sickle cell disease

Serotonin and norepinephrine reuptake inhibitors

Tricyclic antidepressants

Transcranial Doppler

Tissue plasminogen activator

Tricuspid regurgitant jet velocity

Visual Analog Scale

Piel FB, Patil AP, Howes RE, Nyangiri OA, Gething PW, Dewi M, et al. Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model-based map and population estimates. Lancet. 2013;381(9861):142–51.

Article   PubMed   PubMed Central   Google Scholar  

Brousseau DC, Panepinto JA, Nimmer M, Hoffmann RG. The number of people with sickle-cell disease in the United States: national and state estimates. Am J Hematol. 2010;85(1):77–8.

PubMed   Google Scholar  

Payne AB, Mehal JM, Chapman C, Haberling DL, Richardson LC, Bean CJ, et al. Trends in sickle cell disease-related mortality in the United States, 1979 to 2017. Ann Emerg Med. 2020;76(3S):S28–36.

Article   PubMed   Google Scholar  

Brousseau DC, Owens PL, Mosso AL, Panepinto JA, Steiner CA. Acute care utilization and rehospitalizations for sickle cell disease. JAMA. 2010;303(13):1288–94.

Article   CAS   PubMed   Google Scholar  

Sil S, Cohen LL, Dampier C. Psychosocial and functional outcomes in youth with chronic sickle cell pain. Clin J Pain. 2016;32(6):527–33.

Smith WR, Penberthy LT, Bovbjerg VE, McClish DK, Roberts JD, Dahman B, et al. Daily assessment of pain in adults with sickle cell disease. Ann Intern Med. 2008;148(2):94–101.

Ballas SK, Darbari DS. Neuropathy, neuropathic pain, and sickle cell disease. Am J Hematol. 2013;88(11):927–9.

Sharma D, Brandow AM. Neuropathic pain in individuals with sickle cell disease. Neurosci Lett. 2020;714:134445.

Dampier C, Palermo TM, Darbari DS, Hassell K, Smith W, Zempsky W. AAPT diagnostic criteria for chronic sickle cell disease pain. J Pain. 2017;18(5):490–8.

Darbari DS, Hampson JP, Ichesco E, Kadom N, Vezina G, Evangelou I, et al. Frequency of hospitalizations for pain and association with altered brain network connectivity in sickle cell disease. J Pain. 2015;16(11):1077–86.

Levenson JL, Mcclish DK, Dahman BA, Bovbjerg VE, Penberthy LT, et al. Depression and anxiety in adults with sickle cell disease: the PiSCES project. Psychosom Med. 2008;70(2):192–6.

Treadwell MJBB, Kaur K, Gildengorin G. Emotional distress, barriers to care, and health-related quality of life in sickle cell disease. J Clin Outcomes Manag. 2015;22:10.

Google Scholar  

Jonassaint CR, Jones VL, Leong S, Frierson GM. A systematic review of the association between depression and health care utilization in children and adults with sickle cell disease. Br J Haematol. 2016;174(1):136–47.

Laurence B, George D, Woods D. Association between elevated depressive symptoms and clinical disease severity in African-American adults with sickle cell disease. J Natl Med Assoc. 2006;98(3):365–9.

PubMed   PubMed Central   Google Scholar  

Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. 2001;16(9):606–13.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Faulstich ME, Carey MP, Ruggiero L, Enyart P, Gresham F. Assessment of depression in childhood and adolescence: an evaluation of the Center for Epidemiological Studies Depression Scale for Children (CES-DC). Am J Psychiatry. 1986;143(8):1024–7.

Spielberger CD, Gorsuch RL, Lushene R, Vagg PR, Jacobs GA. Manual for the state-trait anxiety inventory. Palo Alto: Consulting Psychologists Press; 1983.

Brandow AM, Carroll CP, Creary S, Edwards-Elliott R, Glassberg J, Hurley RW, et al. American Society of Hematology 2020 guidelines for sickle cell disease: management of acute and chronic pain. Blood Adv. 2020;4(12):2656–701.

Lubega FA, DeSilva MS, Munube D, Nkwine R, Tumukunde J, Agaba PK, et al. Low dose ketamine versus morphine for acute severe vaso occlusive pain in children: a randomized controlled trial. Scand J Pain. 2018;18(1):19–27.

Nobrega R, Sheehy KA, Lippold C, Rice AL, Finkel JC, Quezado ZMN. Patient characteristics affect the response to ketamine and opioids during the treatment of vaso-occlusive episode-related pain in sickle cell disease. Pediatr Res. 2018;83(2):445–54.

Perlin E, Finke H, Castro O, Rana S, Pittman J, Burt R, et al. Enhancement of pain control with ketorolac tromethamine in patients with sickle cell vaso-occlusive crisis. Am J Hematol. 1994;46(1):43–7.

Beiter JL Jr, Simon HK, Chambliss CR, Adamkiewicz T, Sullivan K. Intravenous ketorolac in the emergency department management of sickle cell pain and predictors of its effectiveness. Arch Pediatr Adolesc Med. 2001;155(4):496–500.

Chou R, Turner JA, Devine EB, Hansen RN, Sullivan SD, Blazina I, et al. The effectiveness and risks of long-term opioid therapy for chronic pain: a systematic review for a National Institutes of Health Pathways to Prevention Workshop. Ann Intern Med. 2015;162(4):276–86.

Molokie RE, Wilkie DJ, Wittert H, Suarez ML, Yao Y, Zhao Z, et al. Mechanism-driven phase I translational study of trifluoperazine in adults with sickle cell disease. Eur J Pharmacol. 2014;723:419–24.

Schlaeger JM, Molokie RE, Yao Y, Suarez ML, Golembiewski J, Wilkie DJ, et al. Management of sickle cell pain using pregabalin: a pilot study. Pain Manag Nurs. 2017;18(6):391–400.

Osunkwo I, Ziegler TR, Alvarez J, McCracken C, Cherry K, Osunkwo CE, et al. High dose vitamin D therapy for chronic pain in children and adolescents with sickle cell disease: results of a randomized double blind pilot study. Br J Haematol. 2012;159(2):211–5.

Hauser W, Urrutia G, Tort S, Uceyler N, Walitt B. Serotonin and noradrenaline reuptake inhibitors (SNRIs) for fibromyalgia syndrome. Cochrane Database Syst Rev. 2013;1:CD010292.

Hauser W, Wolfe F, Tolle T, Uceyler N, Sommer C. The role of antidepressants in the management of fibromyalgia syndrome: a systematic review and meta-analysis. CNS Drugs. 2012;26(4):297–307.

Derry S, Cording M, Wiffen PJ, Law S, Phillips T, Moore RA. Pregabalin for pain in fibromyalgia in adults. Cochrane Database Syst Rev. 2016;9:CD011790.

Sil S, Lai K, Lee JL, Gilleland Marchak J, Thompson B, Cohen L, et al. Preliminary evaluation of the clinical implementation of cognitive-behavioral therapy for chronic pain management in pediatric sickle cell disease. Complement Ther Med. 2020;49:102348.

Lu K, Cheng MC, Ge X, Berger A, Xu D, Kato GJ, et al. A retrospective review of acupuncture use for the treatment of pain in sickle cell disease patients: descriptive analysis from a single institution. Clin J Pain. 2014;30(9):825–30.

Mahmood LA, Reece-Stremtan S, Idiokitas R, Martin B, Margulies S, Hardy SJ, et al. Acupuncture for pain management in children with sickle cell disease. Complement Ther Med. 2020;49:102287.

Mehari A, Alam S, Tian X, Cuttica MJ, Barnett CF, Miles G, et al. Hemodynamic predictors of mortality in adults with sickle cell disease. Am J Respir Crit Care Med. 2013;187(8):840–7.

Cohen RT, Strunk RC, Rodeghier M, Rosen CL, Kirkham FJ, Kirkby J, et al. Pattern of lung function is not associated with prior or future morbidity in children with sickle cell anemia. Ann Am Thorac Soc. 2016;13(8):1314–23.

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):1544–50.

Sharma S, Efird JT, Knupp C, Kadali R, Liles D, Shiue K, et al. Sleep disorders in adult sickle cell patients. J Clin Sleep Med. 2015;11(3):219–23.

Rosen CL, Debaun MR, Strunk RC, Redline S, Seicean S, Craven DI, et al. Obstructive sleep apnea and sickle cell anemia. Pediatrics. 2014;134(2):273–81.

Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, et al. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J. 2019;53(1).

Chaturvedi S, Labib Ghafuri D, Kassim A, Rodeghier M, DeBaun MR. Elevated tricuspid regurgitant jet velocity, reduced forced expiratory volume in 1 second, and mortality in adults with sickle cell disease. Am J Hematol. 2017;92(2):125–30.

Parent F, Bachir D, Inamo J, Lionnet F, Driss F, Loko G, et al. A hemodynamic study of pulmonary hypertension in sickle cell disease. N Engl J Med. 2011;365(1):44–53.

Machado RF, Anthi A, Steinberg MH, Bonds D, Sachdev V, Kato GJ, et al. N-terminal pro-brain natriuretic peptide levels and risk of death in sickle cell disease. JAMA. 2006;296(3):310–8.

Mondal P, Yirinec A, Midya V, Sankoorikal BJ, Smink G, Khokhar A, et al. Diagnostic value of spirometry vs impulse oscillometry: a comparative study in children with sickle cell disease. Pediatr Pulmonol. 2019;54(9):1422–30.

Field JJ, Stocks J, Kirkham FJ, Rosen CL, Dietzen DJ, Semon T, et al. Airway hyperresponsiveness in children with sickle cell anemia. Chest. 2011;139(3):563–8.

Dlamini N, Saunders DE, Bynevelt M, Trompeter S, Cox TC, Bucks RS, et al. Nocturnal oxyhemoglobin desaturation and arteriopathy in a pediatric sickle cell disease cohort. Neurology. 2017;89(24):2406–12.

Barst RJ, Mubarak KK, Machado RF, Ataga KI, Benza RL, Castro O, et al. Exercise capacity and haemodynamics in patients with sickle cell disease with pulmonary hypertension treated with bosentan: results of the ASSET studies. Br J Haematol. 2010;149(3):426–35.

Machado RF, Barst RJ, Yovetich NA, Hassell KL, Kato GJ, Gordeuk VR, et al. Hospitalization for pain in patients with sickle cell disease treated with sildenafil for elevated TRV and low exercise capacity. Blood. 2011;118(4):855–64.

Turpin M, Chantalat-Auger C, Parent F, Driss F, Lionnet F, Habibi A, et al. Chronic blood exchange transfusions in the management of pre-capillary pulmonary hypertension complicating sickle cell disease. European Respiratory Journal. 2018;52(4).

Knight-Madden JM, Barton-Gooden A, Weaver SR, Reid M, Greenough A. Mortality, asthma, smoking and acute chest syndrome in young adults with sickle cell disease. Lung. 2013;191(1):95–100.

Strunk RC, Cohen RT, Cooper BP, Rodeghier M, Kirkham FJ, Warner JO, et al. Wheezing symptoms and parental asthma are associated with a physician diagnosis of asthma in children with sickle cell anemia. J Pediatr. 2014;164(4):821-6e1.

Article   Google Scholar  

Takahashi T, Okubo Y, Handa A. Acute chest syndrome among children hospitalized with vaso-occlusive crisis: a nationwide study in the United States. Pediatr Blood Cancer. 2018;65(3):e26885.

Glassberg J, Minnitti C, Cromwell C, Cytryn L, Kraus T, Skloot GS, et al. Inhaled steroids reduce pain and sVCAM levels in individuals with sickle cell disease: a triple-blind, randomized trial. Am J Hematol. 2017;92(7):622–31.

Ohene-Frempong K, Weiner SJ, Sleeper LA, Miller ST, Embury S, Moohr JW, et al. Cerebrovascular accidents in sickle cell disease: rates and risk factors. Blood. 1998;91(1):288–94.

CAS   PubMed   Google Scholar  

Bernaudin F, Verlhac S, Arnaud C, Kamdem A, Vasile M, Kasbi F, et al. Chronic and acute anemia and extracranial internal carotid stenosis are risk factors for silent cerebral infarcts in sickle cell anemia. Blood. 2015;125(10):1653–61.

Kassim AA, Pruthi S, Day M, Rodeghier M, Gindville MC, Brodsky MA, et al. Silent cerebral infarcts and cerebral aneurysms are prevalent in adults with sickle cell anemia. Blood. 2016;127(16):2038–40.

Powars D, Wilson B, Imbus C, Pegelow C, Allen J. The natural history of stroke in sickle cell disease. Am J Med. 1978;65(3):461–71.

Switzer JA, Hess DC, Nichols FT, Adams RJ. Pathophysiology and treatment of stroke in sickle-cell disease: present and future. Lancet Neurol. 2006;5(6):501–12.

DeBaun MR, Gordon M, McKinstry RC, Noetzel MJ, White DA, Sarnaik SA, et al. Controlled trial of transfusions for silent cerebral infarcts in sickle cell anemia. N Engl J Med. 2014;371(8):699–710.

DeBaun MR, Jordan LC, King AA, Schatz J, Vichinsky E, Fox CK, et al. American Society of Hematology 2020 guidelines for sickle cell disease: prevention, diagnosis, and treatment of cerebrovascular disease in children and adults. Blood Adv. 2020;4(8):1554–88.

Dobson SR, Holden KR, Nietert PJ, Cure JK, Laver JH, Disco D, et al. Moyamoya syndrome in childhood sickle cell disease: a predictive factor for recurrent cerebrovascular events. Blood. 2002;99(9):3144–50.

Adams RJ, McKie VC, Hsu L, Files B, Vichinsky E, Pegelow C, 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.

Vichinsky EP, Neumayr LD, Gold JI, Weiner MW, Rule RR, Truran D, et al. Neuropsychological dysfunction and neuroimaging abnormalities in neurologically intact adults with sickle cell anemia. JAMA. 2010;303(18):1823–31.

Hijmans CT, Fijnvandraat K, Grootenhuis MA, van Geloven N, Heijboer H, Peters M, et al. Neurocognitive deficits in children with sickle cell disease: a comprehensive profile. Pediatr Blood Cancer. 2011;56(5):783–8.

Ware RE, Davis BR, Schultz WH, Brown RC, Aygun B, Sarnaik S, et al. Hydroxycarbamide versus chronic transfusion for maintenance of transcranial doppler flow velocities in children with sickle cell anaemia-TCD With Transfusions Changing to Hydroxyurea (TWiTCH): a multicentre, open-label, phase 3, non-inferiority trial. Lancet. 2016;387(10019):661–70.

Scothorn DJ, Price C, Schwartz D, Terrill C, Buchanan GR, Shurney W, et al. Risk of recurrent stroke in children with sickle cell disease receiving blood transfusion therapy for at least five years after initial stroke. J Pediatr. 2002;140(3):348–54.

Hulbert ML, Scothorn DJ, Panepinto JA, Scott JP, Buchanan GR, Sarnaik S, et al. Exchange blood transfusion compared with simple transfusion for first overt stroke is associated with a lower risk of subsequent stroke: a retrospective cohort study of 137 children with sickle cell anemia. J Pediatr. 2006;149(5):710–2.

Hall EM, Leonard J, Smith JL, Guilliams KP, Binkley M, Fallon RJ, et al. Reduction in overt and silent stroke recurrence rate following cerebral revascularization surgery in children with sickle cell disease and severe cerebral vasculopathy. Pediatr Blood Cancer. 2016;63(8):1431–7.

Lebensburger JD, Aban I, Pernell B, Kasztan M, Feig DI, Hilliard LM, et al. Hyperfiltration during early childhood precedes albuminuria in pediatric sickle cell nephropathy. Am J Hematol. 2019;94(4):417–23.

Niss O, Lane A, Asnani MR, Yee ME, Raj A, Creary S, et al. Progression of albuminuria in patients with sickle cell anemia: a multicenter, longitudinal study. Blood Adv. 2020;4(7):1501–11.

Cazenave M, Audard V, Bertocchio JP, Habibi A, Baron S, Prot-Bertoye C, et al. Tubular acidification defect in adults with sickle cell disease. Clin J Am Soc Nephrol. 2020;15(1):16–24.

Yee MEM, Lane PA, Archer DR, Joiner CH, Eckman JR, Guasch A. Estimation of glomerular filtration rate using serum cystatin C and creatinine in adults with sickle cell anemia. Am J Hematol. 2017;92(10):E598–9.

Yawn BP, Buchanan GR, Afenyi-Annan AN, Ballas SK, Hassell KL, James AH, et al. Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. JAMA. 2014;312(10):1033–48.

Saraf SL, Zhang X, Shah B, Kanias T, Gudehithlu KP, Kittles R, et al. Genetic variants and cell-free hemoglobin processing in sickle cell nephropathy. Haematologica. 2015;100(10):1275–84.

Hamideh D, Raj V, Harrington T, Li H, Margolles E, Amole F, et al. Albuminuria correlates with hemolysis and NAG and KIM-1 in patients with sickle cell anemia. Pediatr Nephrol. 2014;29(10):1997–2003.

dos Santos TE, Goncalves RP, Barbosa MC, da Silva GB, Jr., Daher Ede F. Monocyte chemoatractant protein-1: a potential biomarker of renal lesion and its relation with oxidative status in sickle cell disease. Blood Cells Mol Dis. 2015;54(3):297–301.

Audard V, Moutereau S, Vandemelebrouck G, Habibi A, Khellaf M, Grimbert P, et al. First evidence of subclinical renal tubular injury during sickle-cell crisis. Orphanet J Rare Dis. 2014;9:67.

Baddam S, Aban I, Hilliard L, Howard T, Askenazi D, Lebensburger JD. Acute kidney injury during a pediatric sickle cell vaso-occlusive pain crisis. Pediatr Nephrol. 2017;32(8):1451–6.

Zahr RS, Hankins JS, Kang G, Li C, Wang WC, Lebensburger J, et al. Hydroxyurea prevents onset and progression of albuminuria in children with sickle cell anemia. Am J Hematol. 2019;94(1):E27–9.

Alvarez O, Miller ST, Wang WC, Luo Z, McCarville MB, Schwartz GJ, et al. Effect of hydroxyurea treatment on renal function parameters: results from the multi-center placebo-controlled BABY HUG clinical trial for infants with sickle cell anemia. Pediatr Blood Cancer. 2012;59(4):668–74.

Laurin LP, Nachman PH, Desai PC, Ataga KI, Derebail VK. Hydroxyurea is associated with lower prevalence of albuminuria in adults with sickle cell disease. Nephrol Dial Transplant. 2014;29(6):1211–8.

Quinn CT, Saraf SL, Gordeuk VR, Fitzhugh CD, Creary SE, Bodas P, et al. Losartan for the nephropathy of sickle cell anemia: a phase-2, multicenter trial. Am J Hematol. 2017;92(9):E520–8.

McClellan AC, Luthi JC, Lynch JR, Soucie JM, Kulkarni R, Guasch A, et al. High one year mortality in adults with sickle cell disease and end-stage renal disease. Br J Haematol. 2012;159(3):360–7.

Gérardin C, Moktefi A, Couchoud C, Duquesne A, Ouali N, Gataut P, et al. Survival and specific outcome of sickle cell disease patients after renal transplantation. Br J Haematol. 2019;187(5):676–80.

Ataga KI, Kutlar A, Kanter J, Liles D, Cancado R, Friedrisch J, et al. Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med. 2017;376(5):429–39.

Niihara Y, Miller ST, Kanter J, Lanzkron S, Smith WR, Hsu LL, et al. A phase 3 trial of l -glutamine in sickle cell disease. N Engl J Med. 2018;379(3):226–35.

Vichinsky E, Hoppe CC, Ataga KI, Ware RE, Nduba V, El-Beshlawy A, et al. A phase 3 randomized trial of voxelotor in sickle cell disease. N Engl J Med. 2019;381(6):509–19.

Gluckman E, Cappelli B, Bernaudin F, Labopin M, Volt F, Carreras J, et al. Sickle cell disease: an international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood. 2017;129(11):1548–56.

Eapen M, Brazauskas R, Walters MC, Bernaudin F, Bo-Subait K, Fitzhugh CD, et al. Effect of donor type and conditioning regimen intensity on allogeneic transplantation outcomes in patients with sickle cell disease: a retrospective multicentre, cohort study. Lancet Haematol. 2019;6(11):e585–96.

Bolanos-Meade J, Cooke KR, Gamper CJ, Ali SA, Ambinder RF, Borrello IM, et al. Effect of increased dose of total body irradiation on graft failure associated with HLA-haploidentical transplantation in patients with severe haemoglobinopathies: a prospective clinical trial. Lancet Haematol. 2019;6(4):e183–93.

Krishnamurti L, Ross D, Sinha C, Leong T, Bakshi N, Mittal N, et al. Comparative effectiveness of a web-based patient decision aid for therapeutic options for sickle cell disease: randomized controlled trial. J Med Internet Res. 2019;21(12):e14462.

Download references

Acknowledgements

We would like to acknowledge Lana Mucalo, MD, for supporting data collection for this manuscript.

Author information

Authors and affiliations.

Department of Pediatrics, Section of Pediatric Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI, USA

A. M. Brandow

Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA

You can also search for this author in PubMed   Google Scholar

Contributions

AB and RL contributed equally to the writing and editing of this manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to R. I. Liem .

Ethics declarations

Ethics approval and consent to participate, consent for publication, competing interests.

The authors have no competing interests to declare.

Additional information

Publisher's note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ . The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Cite this article.

Brandow, A.M., Liem, R.I. Advances in the diagnosis and treatment of sickle cell disease. J Hematol Oncol 15 , 20 (2022). https://doi.org/10.1186/s13045-022-01237-z

Download citation

Received : 30 September 2021

Accepted : 15 February 2022

Published : 03 March 2022

DOI : https://doi.org/10.1186/s13045-022-01237-z

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Sickle cell anemia

Journal of Hematology & Oncology

ISSN: 1756-8722

current research on sickle cell anemia 2022

FDA approves cure for sickle cell disease, the first treatment to use gene-editing tool CRISPR

The Food and Drug Administration on Friday approved a powerful treatment for sickle cell disease , a devastating illness that affects more than 100,000 Americans, the majority of whom are Black.

The therapy, called Casgevy, from Vertex Pharmaceuticals and CRISPR Therapeutics, is the first medicine to be approved in the United States that uses the gene-editing tool CRISPR , which won its inventors the Nobel Prize in chemistry in 2020.

“I think this is a pivotal moment in the field,” said Dr. Alexis Thompson, chief of the division of hematology at Children’s Hospital of Philadelphia, who has previously consulted for Vertex. “It’s been really remarkable how quickly we went from the actual discovery of CRISPR, the awarding of a Nobel Prize, and now actually seeing it being an approved product.”

The approval marks the first of two potential breakthroughs for the inherited blood disorder. The FDA on Friday also approved a second treatment for sickle cell disease, called Lyfgenia, a gene therapy from drugmaker Bluebird Bio. Both treatments work by genetically modifying a patient’s own stem cells.

Until now, the only known cure for sickle cell disease was a bone marrow transplant from a donor, which carries the risk of rejection by the immune system, in addition to the difficult process of finding a matching donor.

Casgevy, which was approved for people ages 12 and older, removes the need for a donor. Using CRISPR, it edits the DNA found in a patient’s stem cells to remove the gene that causes the disease.

“The patient is their own donor,” Thompson said.

“It’s a game-changer,” said Dr. Asmaa Ferdjallah, a pediatric hematologist and bone marrow transplant physician at the Mayo Clinic in Rochester, Minnesota. “To really reimagine and re-discuss sickle cell disease as a curable disease and not as this painful and debilitating chronic disease is hope enough with this news.”

Still, the new therapy is extremely expensive — $2.2 million per patient, Vertex said. The pricing strategy, experts argue, may place it out of reach for many families. What’s more, that price doesn’t include the cost of care associated with the treatment, like a stay in the hospital or chemotherapy.

“We really have to make sure that it is accessible,” said Dr. Rabi Hanna, a pediatric hematologist-oncologist at the Cleveland Clinic who has previously served on the advisory board for Vertex. “This could be an equalizer for people with sickle cell because many patients cannot pursue career options” because of the illness.

“It’s something families have been aware of in the early research stage, and they’ve been very patiently waiting for years,” Ferdjallah said. “It’s been eagerly awaited by patients and families, but also by providers and physicians.”

How Casgevy works

In patients with sickle cell disease, red blood cells, which are usually disk-shaped, take on a crescent or sickle shape. This change can cause cells to clump together, leading to clots and blockages in the blood vessels, starving tissues of oxygen. Patients can experience excruciating pain, breathing problems and stroke .

Casgevy works by editing the DNA in a patient’s stem cells — which are responsible for making the body’s blood cells — so that they no longer produce sickle-shaped cells.

While technically a one-time treatment, a number of steps that span months are required before the patient actually gets the modified stem cells. It begins with a series of blood transfusions over three to four months, after which the stem cells are extracted from the patient’s bone marrow and sent off to a lab where they are edited, Hanna said.

Before they can be reinfused into the patient, however, doctors need to make sure no flawed stem cells remain in the body. To do so, chemotherapy is used to destroy the patient’s bone marrow. 

Only then can the edited stem cells be reinfused into the patient, followed by another month or two in the hospital to allow the cells to grow and the patient to recover.

Hanna said he’s always “cautious” when telling families and patients about the one-time treatment because they may have unrealistic expectations.

“There are multiple phases of this journey,” he said.

The clinical trial included 46 people in the U.S. and abroad, 30 of whom had at least 18 months of follow-up care after the treatment. Of those, the treatment has been successful in 29.

LaRae Morning, 29, of Phoenix, was among the trial patients whose treatment was successful.

Her doctors did not expect her to live past the age of 11. Her mother lost several jobs when Morning was a child and a teenager because of her frequent hospital visits.

In April 2021, Morning joined the clinical trial at Sarah Cannon Research Institute and HCA Healthcare’s The Children’s Hospital at TriStar Centennial in Nashville, Tennessee, a decision she initially regretted. Living in Phoenix, she had to fly to Nashville once a month for treatment. It included several blood transfusions, which lasted eight hours each, and taking a medication, called plerixafor, which she recalled causing her intense stomachaches. When she started chemotherapy, her hair began to fall out and her skin changed color, resembling the appearance of a chemical burn. She also experienced nausea.

It took about six to seven months for her to feel back to normal following the CRISPR treatment. Now, she said, she’s feeling the benefits, going out to coffee shops, spending time with her friends and finishing her first semester of law school in Washington, D.C.

“Now that I’m here, I’m so happy that I did it,” she said of the trial. “I’m just like a regular person. I wake up and do a 5K. I lift weights. If I wanted to swim, I can swim. I’m still trying to know how far I can stretch it, like what are all the things I can do.”

That’s been the experience for several other patients in the trial as well, according to Dr. Monica Bhatia, chief of pediatric stem cell transplantation at NewYork-Presbyterian/Columbia University Irving Medical Center. Bhatia is a principal investigator at one of the clinical trial sites in New York City.

Following the treatment, most patients were going back to school, going to the gym or resuming other activities — “things that a lot of people take for granted,” she said — after about three to four months.

“They’re really able to live life without restrictions,” Bhatia said.

Dr. Haydar Frangoul, medical director of pediatric hematology-oncology for the Sarah Cannon Research Institute, said he is hopeful the therapy will provide relief to more patients.

“I think this is a huge moment for patients with sickle cell disease,” said Frangoul, who was the lead investigator on the clinical trial and treated Morning.

Long-term questions 

Although Casgevy has been shown to be effective, experts still don’t know about potential long-term effects, since the trial is only set to run for two years.

During a meeting in October, an FDA advisory committee discussed the risk of “off-target” effects , which refers to when the gene-editing tool makes cuts to other stretches of DNA other than the intended target and how the FDA should consider those risks moving forward.

It’s unclear what effects an off-target edit would have on a patient, but the fear is that it could result in unintended health consequences down the road, Thompson said. “To date, there do not appear to be measurable consequences.”

The FDA did, however, add a boxed warning — the strongest safety warning label— to Bluebird Bio’s Lyfgenia, noting that in rare cases the treatment can cause certain blood cancers.

Dr. Nicole Verdun, director of the Office of Therapeutic Products in the FDA's Center for Biologics Evaluation and Research, said Lyfgenia was given the warning after two patients who got the therapy in a clinical trial died from a form of leukemia.

It's unclear whether the gene therapy itself or another part of the treatment process, such as the chemotherapy, caused the cancer, but Verdun said the agency thought the deaths "rose to the level of a black-box warning." No cases were seen in the Vertex clinical trial, she said.

Bhatia is following the patients for 15 years as part of a post-approval study for Casgevy and will be monitoring for signs of long-term effects.

“Long-term follow-up is still going to be so crucial,” she said.

Christopher Vega in the hospital.

Christopher Vega, 31, from Allentown, Pennsylvania, said the possibility of long-term effects aren’t a concern for him; he is happy to be living a life free of chronic pain.

He joined the clinical trial at the Children’s Hospital of Philadelphia in late 2020. He had suffered from chronic fatigue since he was a young child and would end up in the hospital every year with a pain crisis.

“When I was younger, my mom used to always tell me things happen for a reason. And I had so much trouble believing that, because I always thought, ‘Why me?’” he said.

While the treatment process was not always easy — Vega temporarily lost his hair, felt weak and nauseous and developed skin rashes — he said it was worth it.

“I am a whole different person,” said Vega, who is now attending the Los Angeles Film School online for visual effects while taking care of his 5-year-old daughter.

“Sometimes I would get small moments of anxiety that I would have a crisis,” he said. “And after going years now, I’m slowly coming to terms with, I’m OK, and I know I’m going to be here, present.”

current research on sickle cell anemia 2022

Berkeley Lovelace Jr. is a health and medical reporter for NBC News. He covers the Food and Drug Administration, with a special focus on Covid vaccines, prescription drug pricing and health care. He previously covered the biotech and pharmaceutical industry with CNBC.

current research on sickle cell anemia 2022

Marina Kopf is an associate producer with the NBC News Health and Medical Unit.

U.S. flag

An official website of the United States government

Here’s how you know

Official websites use .gov A .gov website belongs to an official government organization in the United States.

Secure .gov websites use HTTPS A lock ( A locked padlock ) or https:// means you’ve safely connected to the .gov website. Share sensitive information only on official, secure websites.

  • Heart-Healthy Living
  • High Blood Pressure
  • Sickle Cell Disease
  • Sleep Apnea
  • Information & Resources on COVID-19
  • The Heart Truth®
  • Learn More Breathe Better®
  • Blood Diseases & Disorders Education Program
  • Publications and Resources
  • Clinical Trials
  • Blood Disorders and Blood Safety
  • Sleep Science and Sleep Disorders
  • Lung Diseases
  • Health Disparities and Inequities
  • Heart and Vascular Diseases
  • Precision Medicine Activities
  • Obesity, Nutrition, and Physical Activity
  • Population and Epidemiology Studies
  • Women’s Health
  • Research Topics
  • All Science A-Z
  • Grants and Training Home
  • Policies and Guidelines
  • Funding Opportunities and Contacts
  • Training and Career Development
  • Email Alerts
  • NHLBI in the Press
  • Research Features
  • Ask a Scientist
  • Past Events
  • Upcoming Events
  • Mission and Strategic Vision
  • Divisions, Offices and Centers
  • Advisory Committees
  • Budget and Legislative Information
  • Jobs and Working at the NHLBI
  • Contact and FAQs
  • NIH Sleep Research Plan
  • News and Events
  • < Back To All News

NIH statement on new FDA-approved gene therapies for sickle cell disease

scientific illustration - sickle cell blood cell

Today, the U.S. Food and Drug Administration (FDA) approved two gene therapies for the treatment of sickle cell disease in patients 12 years and older. About  100,000 Americans and millions of people  around the world have  sickle cell disease , a hereditary disease common among those whose ancestors come from sub-Saharan Africa, Mediterranean countries, India and the Middle East.

The U.S. National Institutes of Health (NIH) has long invested in basic genetics and genomics research, clinical trials, as well as translational medicine and social science studies, to advance our understanding of this widespread illness to help develop effective therapies.

For example:

  • NIH investments in genome sequencing technology have led to the  costs of whole genome sequencing being over a million-fold less expensive  than a couple of decades ago. Affordable sequencing technology is critical to the diagnosis of sickle cell disease.  
  • NIH researchers successfully edited the  disease-causing mutation in blood-forming cells  taken directly from people with sickle-cell disease. One of the new sickle cell treatments uses the CRISPR gene-editing system, a first for humans in the U.S.  
  • NIH funds the  Cure Sickle Cell Initiative  to help speed the development of cures for sickle cell disease, which takes advantage of the latest genetic discoveries and technological advances to move the most promising genetic-based curative therapies safely into clinical trials.  
  • NIH is working closely with the Centers for Medicare and Medicaid Services (CMS) to identify and reduce barriers to uptake and to support readiness of patients and clinicians for these therapies once available.  
  • NIH leads  The Democratizing Education for Sickle Cell Disease Gene Therapy Project , a collaborative effort that aims to help patients and their support networks navigate emerging developments in gene therapies for sickle cell disease.

"NIH celebrates this enormous milestone in treatment for sickle cell disease, the first human genetic disease that was understood at the protein and DNA levels. Researchers have worked hard to find a long-term, durable therapy for sickle cell disease. Research has enabled the use of gene therapy to make genetic changes in the bone marrow of sickle cell patients, leading to normal red blood cell levels. None of this would be possible without federal investments in basic science research."

—Eric Green, M.D., Ph.D., Director of the National Human Genome Research Institute

"We have made some exciting research advances over the years and are ready to collect on our scientific investments in sickle cell research. However, we must remember that these advances need to go hand-in-hand with scalable innovations that will ensure equitable access to life-altering care and that we must continue to engage in additional research endeavors that will minimize or eliminate potential risks that might be associated with these therapies."

—Gary H. Gibbons, M.D., Director of the National Heart, Lung, and Blood Institute

“The sickle cell disease community has historically been underserved and underacknowledged when it comes to rare genetic conditions, so it is heartening to see sickle cell disease at the forefront of gene therapy. It is critical that people with sickle cell disease who are considering gene therapy fully understand the treatment so they can make an informed decision on whether it is appropriate for them. Patients need accessible, understandable and actionable educational materials to help them make such decisions, as well as support from practitioners and the healthcare system to consider these therapies.”

—Vence L. Bonham, Jr., J.D., Acting Deputy Director of the National Human Genome Research Institute

Additional Resources

For more information, visit:

  • Sickle cell disease gene therapy education project
  • Talking Glossary of Genomic and Genetic Terms: Sickle Cell Disease
  • Sickle Cell Disease Treatment  (NHLBI)
  • Sickle Cell Disease: Research, Programs, and Progress  (NHLBI)

About the National Heart, Lung, and Blood Institute (NHLBI):  NHLBI is the global leader in conducting and supporting research in heart, lung, and blood diseases and sleep disorders that advances scientific knowledge, improves public health, and saves lives. For more information, visit  www.nhlbi.nih.gov .

About the National Institutes of Health (NIH):  NIH, the nation's medical research agency, includes 27 Institutes and Centers and is a component of the U.S. Department of Health and Human Services. NIH is the primary federal agency conducting and supporting basic, clinical, and translational medical research, and is investigating the causes, treatments, and cures for both common and rare diseases. For more information about NIH and its programs, visit  www.nih.gov .

More Information

Related health topics, health education, related news.

Dr. Gary Gibbons and Dr. Julie Panepinto

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals

Sickle cell disease articles from across Nature Portfolio

Sickle cell disease is an autosomal recessive blood disorder that can lead to anaemia. It is caused by a mutation in the haemoglobin gene, which leads to deformation of red blood cells. Deformed red blood cells can obstruct small vessels and they are prone to destruction.

Latest Research and Reviews

current research on sickle cell anemia 2022

Non-viral DNA delivery and TALEN editing correct the sickle cell mutation in hematopoietic stem cells

Sickle cell disease is a blood disorder that originates from a single point mutation in the HBB gene that codes for hemoglobin. Here, Moiani et al. developed an efficient TALEN-mediated HBB correction process that is compatible with gene therapy applications.

  • Arianna Moiani
  • Julien Valton

current research on sickle cell anemia 2022

Brain network hypersensitivity underlies pain crises in sickle cell disease

  • Minkyung Kim

current research on sickle cell anemia 2022

Risk factors for acute chest syndrome among children with sickle cell anemia hospitalized for vaso-occlusive crises

  • Faisal A. Alghamdi
  • Fawaz Al-Kasim
  • Rehab Alluqmani

current research on sickle cell anemia 2022

Development of pathophysiologically relevant models of sickle cell disease and β-thalassemia for therapeutic studies

Sickle cell disease (SCD) and β-thalassemia (BT) are globally prevalent inherited blood disorders but, despite extensive research, no ex vivo system exists for SCD and BT. Here, the authors generate pathophysiologically relevant erythroid progenitor models of SCD and BT.

  • Pragya Gupta
  • Sangam Giri Goswami
  • Sivaprakash Ramalingam

current research on sickle cell anemia 2022

The value-based price of transformative gene therapy for sickle cell disease: a modeling analysis

  • George Morgan
  • Gregory F. Guzauskas

current research on sickle cell anemia 2022

Evaluating sheep hemoglobins with MD simulations as an animal model for sickle cell disease

  • Caroline E. Kuczynski
  • Christopher D. Porada
  • Graça Almeida-Porada

Advertisement

News and Comment

current research on sickle cell anemia 2022

Autologous globin-edited HSCs ameliorate sickle cell disease

current research on sickle cell anemia 2022

Gene correction for sickle cell disease hits its prime

Prime editing can efficiently rewrite the genetic mutation causing sickle cell disease, in haematopoietic stem cells from patients.

  • Sébastien Levesque
  • Daniel E. Bauer

Health-related quality of life in sickle cell disease

  • Julie A. Panepinto
  • Gregory J. Kato
  • Wally R. Smith

Comparing health-related quality of life in chronic diseases: the importance of analyzing references

  • Christine Maynié-François
  • Stéphane Burtey

Omega-3 fatty acids are a potential therapy for patients with sickle cell disease

  • Adrian Rabinowicz
  • Kebreab Ghebremeskel

Sickle cell solutions in sight

New targets, new drug modalities and new business strategies are drawing long-awaited attention to sickle cell disease.

  • Katie Kingwell

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

current research on sickle cell anemia 2022

  • Download PDF
  • CME & MOC
  • Share X Facebook Email LinkedIn
  • Permissions

Sickle Cell Disease : A Review

  • 1 Division of General Pediatrics, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
  • 2 Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston
  • 3 School of Medicine, Division of Hematology and Oncology, University of California Davis, Sacramento
  • Comment & Response A Review of Sickle Cell Disease—Reply Patricia L. Kavanagh, MD; Titilope Fasipe, MD, PhD; Ted Wun, MD JAMA
  • Comment & Response A Review of Sickle Cell Disease Nikolaos Vlachadis, MD, DMD, MPH, MSc, DSc; Nikolaos Vrachnis, MD, PhD JAMA
  • JAMA Clinical Guidelines Synopsis Diagnosis and Management of Priapism Richard J. Fantus, MD; Robert E. Brannigan, MD; Andrew M. Davis, MD, MPH JAMA

Importance   Sickle cell disease (SCD) is an inherited disorder of hemoglobin, characterized by formation of long chains of hemoglobin when deoxygenated within capillary beds, resulting in sickle-shaped red blood cells, progressive multiorgan damage, and increased mortality. An estimated 300 000 infants are born annually worldwide with SCD. Most individuals with SCD live in sub-Saharan Africa, India, the Mediterranean, and Middle East; approximately 100 000 individuals with SCD live in the US.

Observations   SCD is diagnosed through newborn screening programs, where available, or when patients present with unexplained severe atraumatic pain or normocytic anemia. In SCD, sickling and hemolysis of red blood cells result in vaso-occlusion with associated ischemia. SCD is characterized by repeated episodes of severe acute pain and acute chest syndrome, and by other complications including stroke, chronic pain, nephropathy, retinopathy, avascular necrosis, priapism, and leg ulcers. In the US, nearly all children with SCD survive to adulthood, but average life expectancy remains 20 years less than the general population, with higher mortality as individuals transition from pediatric to adult-focused health care systems. Until 2017, hydroxyurea, which increases fetal hemoglobin and reduces red blood cell sickling, was the only disease-modifying therapy available for SCD and remains first-line therapy for most individuals with SCD. Three additional therapies, L-glutamine, crizanlizumab, and voxelotor, have been approved as adjunctive or second-line agents. In clinical trials, L-glutamine reduced hospitalization rates by 33% and mean length of stay from 11 to 7 days compared with placebo. Crizanlizumab reduced pain crises from 2.98 to 1.63 per year compared with placebo. Voxelotor increased hemoglobin by at least 1 g/dL, significantly more than placebo (51% vs 7%). Hematopoietic stem cell transplant is the only curative therapy, but it is limited by donor availability, with best results seen in children with a matched sibling donor. While SCD is characterized by acute and chronic pain, patients are not more likely to develop addiction to pain medications than the general population.

Conclusions and Relevance   In the US, approximately 100 000 people have SCD, which is characterized by hemolytic anemia, acute and chronic pain, acute chest syndrome; increased incidence of stroke, nephropathy, and retinopathy; and a life span that is 20 years shorter than the general population. While hydroxyurea is first-line therapy for SCD, L-glutamine, crizanlizumab, and voxelotor have been approved in the US since 2017 as adjunctive or second-line treatments, and hematopoietic stem cell transplant with a matched sibling donor is now standard care for severe disease.

Read More About

Kavanagh PL , Fasipe TA , Wun T. Sickle Cell Disease : A Review . JAMA. 2022;328(1):57–68. doi:10.1001/jama.2022.10233

Manage citations:

© 2024

Artificial Intelligence Resource Center

Cardiology in JAMA : Read the Latest

Browse and subscribe to JAMA Network podcasts!

Others Also Liked

Select your interests.

Customize your JAMA Network experience by selecting one or more topics from the list below.

  • Academic Medicine
  • Acid Base, Electrolytes, Fluids
  • Allergy and Clinical Immunology
  • American Indian or Alaska Natives
  • Anesthesiology
  • Anticoagulation
  • Art and Images in Psychiatry
  • Artificial Intelligence
  • Assisted Reproduction
  • Bleeding and Transfusion
  • Caring for the Critically Ill Patient
  • Challenges in Clinical Electrocardiography
  • Climate and Health
  • Climate Change
  • Clinical Challenge
  • Clinical Decision Support
  • Clinical Implications of Basic Neuroscience
  • Clinical Pharmacy and Pharmacology
  • Complementary and Alternative Medicine
  • Consensus Statements
  • Coronavirus (COVID-19)
  • Critical Care Medicine
  • Cultural Competency
  • Dental Medicine
  • Dermatology
  • Diabetes and Endocrinology
  • Diagnostic Test Interpretation
  • Drug Development
  • Electronic Health Records
  • Emergency Medicine
  • End of Life, Hospice, Palliative Care
  • Environmental Health
  • Equity, Diversity, and Inclusion
  • Facial Plastic Surgery
  • Gastroenterology and Hepatology
  • Genetics and Genomics
  • Genomics and Precision Health
  • Global Health
  • Guide to Statistics and Methods
  • Hair Disorders
  • Health Care Delivery Models
  • Health Care Economics, Insurance, Payment
  • Health Care Quality
  • Health Care Reform
  • Health Care Safety
  • Health Care Workforce
  • Health Disparities
  • Health Inequities
  • Health Policy
  • Health Systems Science
  • History of Medicine
  • Hypertension
  • Images in Neurology
  • Implementation Science
  • Infectious Diseases
  • Innovations in Health Care Delivery
  • JAMA Infographic
  • Law and Medicine
  • Leading Change
  • Less is More
  • LGBTQIA Medicine
  • Lifestyle Behaviors
  • Medical Coding
  • Medical Devices and Equipment
  • Medical Education
  • Medical Education and Training
  • Medical Journals and Publishing
  • Mobile Health and Telemedicine
  • Narrative Medicine
  • Neuroscience and Psychiatry
  • Notable Notes
  • Nutrition, Obesity, Exercise
  • Obstetrics and Gynecology
  • Occupational Health
  • Ophthalmology
  • Orthopedics
  • Otolaryngology
  • Pain Medicine
  • Palliative Care
  • Pathology and Laboratory Medicine
  • Patient Care
  • Patient Information
  • Performance Improvement
  • Performance Measures
  • Perioperative Care and Consultation
  • Pharmacoeconomics
  • Pharmacoepidemiology
  • Pharmacogenetics
  • Pharmacy and Clinical Pharmacology
  • Physical Medicine and Rehabilitation
  • Physical Therapy
  • Physician Leadership
  • Population Health
  • Primary Care
  • Professional Well-being
  • Professionalism
  • Psychiatry and Behavioral Health
  • Public Health
  • Pulmonary Medicine
  • Regulatory Agencies
  • Reproductive Health
  • Research, Methods, Statistics
  • Resuscitation
  • Rheumatology
  • Risk Management
  • Scientific Discovery and the Future of Medicine
  • Shared Decision Making and Communication
  • Sleep Medicine
  • Sports Medicine
  • Stem Cell Transplantation
  • Substance Use and Addiction Medicine
  • Surgical Innovation
  • Surgical Pearls
  • Teachable Moment
  • Technology and Finance
  • The Art of JAMA
  • The Arts and Medicine
  • The Rational Clinical Examination
  • Tobacco and e-Cigarettes
  • Translational Medicine
  • Trauma and Injury
  • Treatment Adherence
  • Ultrasonography
  • Users' Guide to the Medical Literature
  • Vaccination
  • Venous Thromboembolism
  • Veterans Health
  • Women's Health
  • Workflow and Process
  • Wound Care, Infection, Healing
  • Register for email alerts with links to free full-text articles
  • Access PDFs of free articles
  • Manage your interests
  • Save searches and receive search alerts

Sickle Cell Disease News logo

  • Small molecule shows promise in lab models for treating sickle cell

Novel molecule SR-18292 found to reduce disease severity in SCD mice

Marisa Wexler, MS avatar

by Marisa Wexler, MS | August 6, 2024

Share this article:

An oversized human hand holds a mouse alongside a rack of three vials filled with blood.

A novel small molecule called SR-18292, which acts to boost the production of fetal hemoglobin, was shown to reduce disease severity in a mouse model of sickle cell disease (SCD) and demonstrated benefits in other lab testing in a new study.

“We’ve uncovered a promising approach that can offer hope to patients with sickle cell disease who have not responded to traditional treatments,” Shuaiying Cui, PhD, a researcher at the Boston Medical Center (BMC) Center of Excellence in Sickle Cell Disease and the study’s senior author, said in a BMC press release .

Experiments in human blood cells showed the anti-sickling effects of SR-18292 combined with hydroxyurea , an established SCD treatment thought to also act on fetal hemoglobin, were more potent when compared with those of either therapy alone.

“Our research shows that combining a small molecule with hydroxyurea enhances the production of fetal hemoglobin through different mechanisms. This could provide a vital new treatment option for sickle cell disease patients who don’t respond well to hydroxyurea alone,” Cui said.

The study, “ PGC-1α agonism induces fetal hemoglobin and exerts antisickling effects in sickle cell disease ,” was published in the journal Science Advances .

An oversized red pen checks boxes labeled

1st SCD patient soon to be given therapy to restore fetal hemoglobin

Small molecule sr-18292 increased fetal hemoglobin in human blood cells.

Sickle cell disease is caused by mutations that lead to the production of an abnormal form of hemoglobin — the protein that red blood cells use to carry oxygen throughout the body. The abnormal hemoglobin tends to clump up inside red blood cells, deforming them into the sickle-like shape that gives the disease its name and ultimately drives its symptoms .

SCD specifically affects the adult version of hemoglobin. Fetal hemoglobin, or HbF for short, is an alternative version of this protein that’s made during early fetal development. Normally, the body stops making HbF and switches on the production of the adult version of hemoglobin shortly after birth.

In people with sickle cell, boosting HbF levels has been shown as a viable strategy for reducing red blood cell sickling and ultimately combatting the disease. However, treatment options targeting HbF are limited.

Previous research has shown that activating PGC-1 alpha, a protein that helps regulate the activity of various genes within cells, can increase HbF production in blood cells. Spurred on by that discovery, researchers at BMC tested the effects of SR-18292, a small molecule that activates PGC-1 alpha, in cell and animal models of SCD.

Initial experiments in human blood stem cells confirmed that SR-18292 treatment was able to increase HbF levels as expected.

Importantly, the researchers noted that the effect of SR-18292 combined with hydroxyurea was more potent at boosting HbF production than either therapy on its own.

Genetic analyses conducted in human blood stem cells also indicated that SR-18292 increased the activity of several genes that are known to boost HbF production. At the same time, the researchers also found that SR-18292 decreased the activity of genes that normally act to turn off HbF production, including BCL11A , which is the gene targeted by Casgevy (exagamglogene autotemcel) — a CRISPR/Cas9-based gene-editing therapy approved late last year for SCD.

An array of different kinds of bacteria is shown in this graphic.

Fewer infections in youth with sickle cell anemia on hydroxyurea

Scientists call sr-18292 a ‘promising’ potential scd treatment.

Following the cell experiments, the researchers tested the effects of this small molecule in a mouse model of SCD. In line with the cell experiments, results in mice showed that SR-18292 was able to boost HbF production.

SR-18292-treated mice also showed fewer signs of premature red blood cell destruction and organ damage compared with their untreated counterparts. The researchers also found evidence suggesting that SR-18292 increased the lifespan of red blood cells in SCD mice and reduced the number of sickled cells in treated animals.

“Induction of HbF by SR-18292 with improved [blood cell health] and reduced organ damage in SCD mice suggests that this small-molecule activator of PGC-1 [alpha] might be a new class of drugs that alone or in combination with other agents can enhance clinically useful increments of HbF,” the researchers wrote.

This breakthrough represents a significant step forward in BMC’s quest for more effective therapies to treat sickle cell disease for all patients.

The team noted that, in addition to suggesting that SR-18292 may be an effective sickle cell treatment, the results more broadly show “proof of principle for targeting PGC-1 [alpha] and its modulators to develop more effective HbF-boosting agents for SCD.”

Overall, the scientists concluded that “SR-18292, or agents in its class, could be a promising additional therapeutic for sickle cell disease.”

Cui said that the team hopes this treatment strategy may one day be used to help sickle cell patients worldwide manage their disease, especially those who don’t have access to cutting-edge, expensive treatments like gene therapies.

“This breakthrough represents a significant step forward in BMC’s quest for more effective therapies to treat sickle cell disease for all patients,” Cui said.

About the Author

Marisa Wexler, MS avatar

Recent Posts

  • Returning to work after an illness requires a considered approach
  • Scientist earns Trailblazer Prize for work aiding Casgevy development
  • FDA clears testing of potentially less toxic conditioning regimen for SCD
  • Advancing representation of Black and sickle cell communities in media
  • 1 step forward, 2 steps back — and trying to stay sane through it all
  • How sickle cell awareness helps both patients and caregivers
  • Hydroxyurea found not to reduce ovarian reserve in SCD patients
  • Expressing gratitude for the irreplaceable caregivers in my life
  • Eculizumab use may safely manage transfusion complication in SCD

Recommended reading

An adult stands at the bedside of a child receiving an intravenous infusion in a hospital bed.

UK investment to expand access to treatment for underserved patients

A doctor expresses concern while looking at a patient's test results.

MRI technique that measures bone marrow fat may predict SCD severity

A group of mice converge around a sprinkling of food pellets.

Reducing Dietary Iron Found to Ease Signs of SCD in Mouse Model

Subscribe to our newsletter.

Get regular updates to your inbox.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Evolution of Extracranial Internal Carotid Artery Disease in Children With Sickle Cell Anemia

Affiliations.

  • 1 Referral Center for Sickle Cell Disease, Department of Pediatric Imaging (S.V., C.O., I.C., S.B., A.K., A.N., H.O., A.T., A.S., N.B., M.E., M.A.), Robert-Debre Hospital, AP-HP, Nord, Paris University, France.
  • 2 Department of Pediatrics (G.I.), Robert-Debre Hospital, AP-HP, Nord, Paris University, France.
  • 3 Referral Center for Sickle Cell Disease, Department of Clinical Research, Intercommunal Creteil Hospital, France (F.B.).
  • PMID: 35387492
  • DOI: 10.1161/STROKEAHA.121.037980

Background: Cerebral arteriopathy in patients with sickle cell anemia mainly affects the intracranial anterior circulation. However, the extracranial internal carotid artery (eICA) can also be stenosed and responsible for ischemic lesions. In children with sickle cell anemia, we perform routine annual Doppler ultrasound assessment of the eICA and magnetic resonance imaging with 3-dimensional time-of-flight magnetic resonance angiography of the Willis circle and neck arteries in those with abnormal velocity. Our aim was to report the evolution of eICA stenoses from 2011 to the present as a function of therapy in a retrospective case-series study. We hypothesized that chronic transfusion (CTT) would be more effective than hydroxyurea and simple observation on the evolution of eICA stenosis.

Methods: Eligibility criteria were a history of eICA velocity ≥160 cm/s with a minimum Doppler and magnetic resonance imaging follow-up of 1 year. eICAs were graded for stenosis according to NASCET (The North American Symptomatic Carotid Endarterectomy Trial). Magnetic resonance imaging was investigated for ischemic lesions. Treatment with hydroxyurea and CTT were obtained from the chart review.

Results: Fifty-four patients were included. Eight patients had a stroke history. The median (range) follow-up was 4.7 years (1.1-9.2 years). On the first neck magnetic resonance angiography, stenosis was present in 48/54 (89%) patients. Kinking was found in 39/54 (72%) patients. On the last neck magnetic resonance angiography, the proportion of patients with eICA stenosis decreased to 39/54 (72%). ICA occlusion occurred in 5 patients despite CTT. Three patients had carotid webs without intracranial stenosis. The proportion of patients with improvement in stenosis score was 8% with no treatment intensification, 20% with hydroxyurea, and 48% with CTT ( P =0.016). The mean (SD) change per year in stenosis score was 0.40 (0.60) without intensification, 0.20 (0.53) with hydroxyurea, and -0.18 (0.55) with CTT ( P =0.006). Ischemic lesions were present initially in 46% of patients, and the incidence of progressive ischemic lesions was 2.5 events/100 patient-years. Cox regression analysis showed that the initial score for eICA stenosis was a significant predictive factor for the risk of new silent cerebral infarct events.

Conclusions: Our study reinforces the need to assess cervical arteries for better prevention of cerebral ischemia and encourage initiation of CTT in sickle cell anemia children with eICA stenosis.

Keywords: anemia, sickle cell; carotid arteries; magnetic resonance angiography; ultrasonography, Doppler.

PubMed Disclaimer

Similar articles

  • Incidence, kinetics, and risk factors for intra- and extracranial cerebral arteriopathies in a newborn sickle cell disease cohort early assessed by transcranial and cervical color Doppler ultrasound. Bernaudin F, Arnaud C, Kamdem A, Hau I, Madhi F, Jung C, Epaud R, Verlhac S. Bernaudin F, et al. Front Neurol. 2022 Sep 14;13:846596. doi: 10.3389/fneur.2022.846596. eCollection 2022. Front Neurol. 2022. PMID: 36188389 Free PMC article.
  • Chronic and acute anemia and extracranial internal carotid stenosis are risk factors for silent cerebral infarcts in sickle cell anemia. Bernaudin F, Verlhac S, Arnaud C, Kamdem A, Vasile M, Kasbi F, Hau I, Madhi F, Fourmaux C, Biscardi S, Epaud R, Pondarré C. Bernaudin F, et al. Blood. 2015 Mar 5;125(10):1653-61. doi: 10.1182/blood-2014-09-599852. Epub 2014 Dec 22. Blood. 2015. PMID: 25533032
  • Extracranial carotid arteriopathy in stroke-free children with sickle cell anemia: detection by submandibular Doppler sonography. Verlhac S, Balandra S, Cussenot I, Kasbi F, Vasile M, Kheniche A, Elmaleh-Bergès M, Ithier G, Benkerrou M, Bernaudin F, Sebag G. Verlhac S, et al. Pediatr Radiol. 2014 May;44(5):587-96. doi: 10.1007/s00247-014-2880-9. Epub 2014 Mar 6. Pediatr Radiol. 2014. PMID: 24595876
  • Sickle cell disease: primary stroke prevention. Gebreyohanns M, Adams RJ. Gebreyohanns M, et al. CNS Spectr. 2004 Jun;9(6):445-9. CNS Spectr. 2004. PMID: 15162093 Review.
  • Sickle Cell Disease and Stroke. Hirtz D, Kirkham FJ. Hirtz D, et al. Pediatr Neurol. 2019 Jun;95:34-41. doi: 10.1016/j.pediatrneurol.2019.02.018. Epub 2019 Feb 27. Pediatr Neurol. 2019. PMID: 30948147 Review.
  • Analysis of structural effects of sickle cell disease on brain vasculature of mice using three-dimensional quantitative phase imaging. Filan C, Song H, Platt MO, Robles FE. Filan C, et al. J Biomed Opt. 2023 Sep;28(9):096501. doi: 10.1117/1.JBO.28.9.096501. Epub 2023 Sep 9. J Biomed Opt. 2023. PMID: 37692563 Free PMC article.
  • Search in MeSH

Related information

  • PubChem Compound (MeSH Keyword)

LinkOut - more resources

Full text sources.

  • Ingenta plc
  • Ovid Technologies, Inc.
  • Wolters Kluwer
  • Genetic Alliance
  • MedlinePlus Health Information

Miscellaneous

  • NCI CPTAC Assay Portal
  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • J Hematol Oncol

Logo of jhemonc

Advances in the diagnosis and treatment of sickle cell disease

A. m. brandow.

1 Department of Pediatrics, Section of Pediatric Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI USA

2 Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL USA

Associated Data

Not applicable.

Sickle cell disease (SCD), which affects approximately 100,000 individuals in the USA and more than 3 million worldwide, is caused by mutations in the βb globin gene that result in sickle hemoglobin production. Sickle hemoglobin polymerization leads to red blood cell sickling, chronic hemolysis and vaso-occlusion. Acute and chronic pain as well as end-organ damage occur throughout the lifespan of individuals living with SCD resulting in significant disease morbidity and a median life expectancy of 43 years in the USA. In this review, we discuss advances in the diagnosis and management of four major complications: acute and chronic pain, cardiopulmonary disease, central nervous system disease and kidney disease. We also discuss advances in disease-modifying and curative therapeutic options for SCD. The recent availability of l -glutamine, crizanlizumab and voxelotor provides an alternative or supplement to hydroxyurea, which remains the mainstay for disease-modifying therapy. Five-year event-free and overall survival rates remain high for individuals with SCD undergoing allogeneic hematopoietic stem cell transplant using matched sibling donors. However, newer approaches to graft-versus-host (GVHD) prophylaxis and the incorporation of post-transplant cyclophosphamide have improved engraftment rates, reduced GVHD and have allowed for alternative donors for individuals without an HLA-matched sibling. Despite progress in the field, additional longitudinal studies, clinical trials as well as dissemination and implementation studies are needed to optimize outcomes in SCD.

Introduction

Sickle cell disease (SCD), a group of inherited hemoglobinopathies characterized by mutations that affect the β-globin chain of hemoglobin, affects approximately 100,000 people in the USA and more than 3 million people worldwide [ 1 , 2 ]. SCD is characterized by chronic hemolytic anemia, severe acute and chronic pain as well as end-organ damage that occurs across the lifespan. SCD is associated with premature mortality with a median age of death of 43 years (IQR 31.5–55 years) [ 3 ]. Treatment requires early diagnosis, prevention of complications and management of end-organ damage. In this review, we discuss recent advances in the diagnosis and management of four major complications in SCD: acute and chronic pain, cardiopulmonary disease, central nervous system disease and kidney disease. Updates in disease-modifying and curative therapies for SCD are also discussed.

Molecular basis and pathophysiology

Hemoglobin S (HbS) results from the replacement of glutamic acid by valine in the sixth position of the β-globin chain of hemoglobin (Fig.  1 ). Severe forms of SCD include hemoglobin SS due to homozygous inheritance of HbS and S/β 0 thalassemia due to co-inheritance of HbS with the β 0 thalassemia mutation. Other forms include co-inheritance of HbS with other β-globin gene mutations such as hemoglobin C, hemoglobin D-Los Angeles/Punjab or β + thalassemia. Hb S has reduced solubility and increased polymerization, which cause red blood cell sickling, hemolysis and vaso-occlusion (Table ​ (Table1) 1 ) that subsequently lead to pain episodes and end-organ damage such as cardiopulmonary, cerebrovascular and kidney disease (Table ​ (Table2 2 ).

An external file that holds a picture, illustration, etc.
Object name is 13045_2022_1237_Fig1_HTML.jpg

Genetic and molecular basis of sickle cell disease. SCD is caused by mutations in the β globin gene, located on the β globin locus found on the short arm of chromosome 11. The homozygous inheritance of Hb S or co-inheritance of Hb S with the β 0 thalassemia mutation results in the most common forms of severe SCD. Co-inheritance of Hb S with other variants such as Hb C, Hb D-Los Angeles/Punjab, Hb O-Arab or β + thalassemia also leads to clinically significant sickling syndromes (LCR, locus control region; HS, hypersensitivity site)

Sickle cell disease

Epidemiology

SCD affects primarily individuals of African or Afro-Caribbean descent

1 in 12 individuals are carriers for sickle cell trait

1 in 365 Black infants in the US are affected by SCD

Approximately 100,000 individuals in the US and millions more worldwide have SCD

Molecular basis and pathophysiology

Mutation is caused by single nucleotide substitution in the 6th codon of β-globin gene ( )

Mutation results in production of sickle hemoglobin

Common genotypes are homozygous SS disease (HbSS) and the compound heterozygous states HbSC, HbS/β and HbS/β thalassemia

Mutation leads to reduced solubility of sickle hemoglobin and increased polymerization

Pathophysiological contributors include red blood cell sickling, hemolysis, vaso-occlusion, cell adhesion, pro-inflammatory state, oxidative injury, endothelial dysfunction and hypercoagulability

Major complications and disease burden

Acute pain and chronic pain syndrome

Functional asplenia and infection

Splenic sequestration

Acute chest syndrome

Cerebrovascular disease and stroke

Neurocognitive deficits

Retinopathy

Priapism

Chronic lung disease

Pulmonary hypertension

Skin ulcers

Osteonecrosis

Chronic kidney disease

Prevalence and pathophysiologic basis of major complications in SCD

PrevalencePathophysiology and/or risk factors
Acute and chronic pain

Acute pain—represents 70% of acute care visits

Chronic pain—30% in adults, 40% in children

Tissue ischemia and infarction

Ischemia–reperfusion injury

Hemolysis-induced endothelial dysfunction

Inflammation and oxidative stress

Peripheral and central nervous system sensitization

Identifiable causes such as avascular necrosis and leg ulcers

Pulmonary hypertension10% in adults by right-heart catheterization

Intravascular hemolysis

Nitric oxide depletion

Chronic hypoxia

Diastolic dysfunction

Diffuse myocardial fibrosis

Chronic lung disease

Obstructive lung disease—16% children, 8% adults

Restrictive lung disease—7% children, 28% adults

Obstructive lung disease—atopy, airway inflammation (↑ leukotrienes)

Restrictive lung disease—recurrent acute chest syndrome

Stroke

Overt stroke—11% by 20 years old

Silent cerebral infarct—39% by 18 years old

Cerebral vasculopathy

↓ Blood oxygen content, ↑ cerebral blood flow and ↑ oxygen extraction

Nocturnal hypoxemia

Moyamoya

Sickle nephropathyChronic kidney disease—20 to 40% of adults

Medullary hypoperfusion and ischemia

Glomerular hemodynamic alterations

Hemolysis-induced oxidative injury

Endothelial damage

Vascular congestion

Hypoxia-inducible factor-1α dependent injury

Acute and chronic pain

Severe intermittent acute pain is the most common SCD complication and accounts for over 70% of acute care visits for individuals with SCD [ 4 ]. Chronic daily pain increases with older age, occurring in 30–40% of adolescents and adults with SCD [ 5 , 6 ]. Acute pain is largely related to vaso-occlusion of sickled red blood cells with ischemia–reperfusion injury and tissue infarction and presents in one isolated anatomic location (e.g., arm, leg, back) or multiple locations. Chronic pain can be caused by sensitization of the central and/or peripheral nervous system and is often diffuse with neuropathic pain features [ 7 , 8 ]. A consensus definition for chronic pain includes “Reports of ongoing pain on most days over the past 6 months either in a single location or multiple locations” [ 9 ]. Disease complications such as avascular necrosis (hip, shoulder) and leg ulcers also cause chronic pain [ 9 ].

Diagnosis of acute and chronic pain

The gold standard for pain assessment and diagnosis is patient self-report. There are no reliable diagnostic tests to confirm the presence of acute or chronic pain in individuals with SCD except when there are identifiable causes like avascular necrosis on imaging or leg ulcers on exam. The effects of pain on individuals’ function are assessed using patient-reported outcome measures (PROs) that determine to what extent pain interferes with individuals’ daily function. Tools shown to be valid, reliable and responsive can be used in clinical practice to track patients’ pain-related function over time to determine additional treatment needs and to compare to population norms [ 10 ]. There are currently no plasma pain biomarkers that improve assessment and management of SCD acute or chronic pain.

Depression and anxiety as co-morbid conditions in SCD can contribute to increased pain, more pain-related distress/interference and poor coping [ 11 ]. The prevalence of depression and anxiety range from 26–33% and 6.5–36%, respectively, in adults with SCD [ 11 – 13 ]. Adults with SCD have an 11% higher prevalence of depression compared to Black American adults without SCD [ 14 ]. Depression and anxiety can be assessed using self-reported validated screening tools (e.g., Depression: Patient Health Questionnaire (PHQ-9) [ 15 ] for adults, Center for Epidemiologic Studies Depression Scale for Children (CES-DC) [ 16 ], PROMIS assessments for adults and children; Anxiety: Generalized Anxiety Disorder 7-item (GAD-7) scale for adults, State-Trait Anxiety Inventory for Children (STAIC) [ 17 ], PROMIS assessments for adults and children). Individuals who screen positive using these tools should be referred for evaluation by a psychologist/psychiatrist.

Management of acute and chronic pain

The goal of acute pain management is to provide sufficient analgesia to return patients to their usual function, which may mean complete resolution of pain for some or return to baseline chronic pain for others. The goal of chronic pain management is to optimize individuals’ function, which may not mean being pain free. When there is an identifiable cause of chronic pain, treatment of the underlying issue (e.g., joint replacement for avascular necrosis, leg ulcer treatment) is important. Opioids, oral for outpatient management and intravenous for inpatient management, are first line therapy for acute SCD pain. In the acute care setting, analgesics should be initiated within 30–60 min of triage [ 18 ]. Ketamine, a non-opioid analgesic, can be prescribed at sub-anesthetic (analgesic) intravenous doses (0.1–0.3 mg/kg per h, maximum 1 mg/kg per h) as adjuvant treatment for acute SCD pain refractory to opioids [ 18 , 19 ]. In an uncontrolled observational study of 85 patients with SCD receiving ketamine infusions for acute pain, ketamine was associated with a decrease in mean opioid consumption by oral morphine equivalents (3.1 vs. 2.2 mg/kg/day, p  < 0.001) and reductions in mean pain scores (0–10 scale) from baseline until discontinuation of the infusion (7.81 vs. 5.44, p  < 0.001) [ 20 ]. Nonsteroidal anti-inflammatory drugs (NSAIDs) are routinely used as adjuvant therapy for acute pain treatment [ 18 ]. In a RCT ( n  = 20) of hospitalized patients with acute pain, ketorolac was associated with lower total dose of meperidine required (1866.7 ± 12.4 vs. 2804.5 ± 795.1 mg, p  < 0.05) and shorter hospitalization (median 3.3 vs. 7.2 days, p  = 0.027) [ 21 ]. In a case series of children treated for 70 acute pain events in the ED, 53% of events resolved with ketorolac and hydration alone with reduction in 100 mm visual analog scale (VAS) pain score from 60 to 13 ( p  < 0.001) [ 22 ]. Patients at risk for NSAID toxicity (e.g., renal impairment, on anticoagulation) should be identified.

Despite paucity of data, chronic opioid therapy (COT) can be considered after assessing benefits versus harms [ 23 ] and the functional status of patients with SCD who have chronic pain. Harms of COT seen in patient populations other than SCD are dose dependent and include myocardial infarction, bone fracture, increased risk of motor vehicle collisions, sexual dysfunction and mortality [ 23 ]. There are few published studies investigating non-opioid analgesics for chronic SCD pain [ 24 – 26 ]. In a randomized trial of 39 participants, those who received Vitamin D experienced a range of 6–10 pain days over 24 weeks while those who received placebo experienced 10–16 pain days, which was not significantly different [ 26 ]. In a phase 1, uncontrolled trial of 18 participants taking trifluoperazine, an antipsychotic drug, 8 participants showed a 50% reduction in the VAS (10 cm horizontal line) pain score from baseline on at least 3 assessments over 24 h without severe sedation or supplemental opioid analgesics, 7 participants showed pain reduction on 1 assessment, and the remaining 3 participants showed no reduction [ 24 ]. Although published data are not available for serotonin and norepinephrine reuptake inhibitors (SNRIs), gabapentinoids and tricyclic antidepressants (TCAs) in individuals with SCD, evidence supports their use in fibromyalgia, a chronic pain condition similar to SCD chronic pain in mechanism. A Cochrane Review that included 10 RCTs ( n  = 6038) showed that the SNRIs milnacipran and duloxetine, compared to placebo, were associated with a reduction in pain [ 27 ]. A systematic review and meta-analysis of 9 studies ( n  = 520) showed the TCA amitriptyline improved pain intensity and function [ 28 ]. Finally, a meta-analysis of 5 RCTs ( n  = 1874) of the gabapentinoid pregabalin showed a reduction in pain intensity [ 29 ]. Collectively, the indirect evidence from fibromyalgia supports the conditional recommendation in current SCD practice guidelines to consider these 3 drug classes for chronic SCD pain treatment [ 18 ]. Standard formulary dosing recommendations should be followed and reported adverse effects considered.

Non-pharmacologic therapies (e.g., integrative, psychological-based therapies) are important components of SCD pain treatment. In a case–control study of 101 children with SCD and chronic pain referred for cognitive behavioral therapy (CBT) (57 CBT, 44 no CBT) [ 30 ], CBT was associated with more rapid decrease in pain hospitalizations (estimate − 0.63, p  < 0.05) and faster reduction in hospital days over time (estimate − 5.50, p  < 0.05). Among 18 children who received CBT and completed PROs pre- and 12 months posttreatment, improvements were seen in mean pain intensity (5.47 vs. 3.76, p  = 0.009; 0–10 numeric rating pain scale), functional disability (26.24 vs. 15.18, p  < 0.001; 0–60 score range) and pain coping (8.00 vs. 9.65, p  = 0.03; 3–15 score range) post treatment [ 30 ]. In 2 uncontrolled clinical trials, acupuncture was associated with a significant reduction in pain scores by 2.1 points (0–10 numeric pain scale) in 24 participants immediately after treatment [ 31 ] or a significant mean difference in pre-post pain scores of 0.9333 (0–10 numeric pain scale) ( p  < 0.000) after 33 acupuncture sessions [ 32 ].

Cardiopulmonary disease

Cardiopulmonary disease is associated with increased morbidity and mortality in individuals with SCD. Pulmonary hypertension (PH), most commonly pulmonary arterial hypertension (PAH), is present based on right-heart catheterization in up to 10% of adults with SCD [ 33 ]. Chronic intravascular hemolysis represents the biggest risk factor for development of PAH in SCD and leads to pulmonary arteriole vasoconstriction and smooth muscle proliferation. Based on pulmonary function testing (PFT), obstructive lung disease may be observed in 16% of children and 8% of adults with SCD, while restrictive lung disease may be seen in up to 28% of adults and only 7% of children with SCD [ 34 , 35 ]. Sleep-disordered breathing, which can manifest as obstructive sleep apnea or nocturnal hypoxemia, occurs in up to 42% of children and 46% of adults with SCD [ 36 , 37 ]. Cardiopulmonary disease, including PH or restrictive lung disease, presents with dyspnea with or without exertion, chest pain, hypoxemia or exercise intolerance that is unexplained or increased from baseline. Obstructive lung disease can also present with wheezing.

Diagnosis of cardiopulmonary disease

The confirmation of PH in patients with SCD requires right-heart catheterization. Recently, the mean pulmonary artery pressure threshold used to define PH in the general population was lowered from ≥ 25 to ≥ 20 mm Hg [ 38 ]. Elevated peak tricuspid regurgitant jet velocity (TRJV) ≥ 2.5 m/s on Doppler echocardiogram (ECHO) is associated with early mortality in adults with SCD and may suggest elevated pulmonary artery pressures, especially when other signs of PH (e.g., right-heart strain, septal flattening) or left ventricular diastolic dysfunction, which may contribute to PH, are present [ 39 ]. However, the positive predictive value (PPV) of peak TRJV alone for identifying PH in adults with SCD is only 25% [ 40 ]. Increasing the peak TRJV threshold to at least 2.9 m/s has been shown to increase the PPV to 64%. For a peak TRJV of 2.5–2.8 m/s, an increased N-terminal pro-brain natriuretic peptide (NT-proBNP) ≥ 164.5 pg/mL or a reduced 6-min walk distance (6MWD) < 333 m can also improve the PPV to 62% with a false negative rate of 7% [ 33 , 40 , 41 ].

PFT, which includes spirometry and measurement of lung volumes and diffusion capacity, is standard for diagnosing obstructive and restrictive lung disease in patients with SCD. Emerging modalities include impulse oscillometry, a non-invasive method using forced sound waves to detect changes in lower airway mechanics in individuals unable to perform spirometry [ 42 ], and airway provocation studies using cold air or methacholine to reveal latent airway hyperreactivity [ 43 ]. Formal in-lab, sleep study/polysomnography remains the gold standard to evaluate for sleep-disordered breathing, which may include nocturnal hypoxemia, apnea/hypopnea events and other causes of sleep disruption. Nocturnal hypoxemia may increase red blood cell sickling, cellular adhesion and endothelial dysfunction. In 47 children with SCD, mean overnight oxygen saturation was higher in those with grade 0 compared to grade 2 or 3 cerebral arteriopathy (97 ± 1.6 vs. 93.9 ± 3.7 vs. 93.5 ± 3.0%, p  < 0.01) on magnetic resonance angiography and lower overnight oxygen saturation was independently associated with mild, moderate or severe cerebral arteriopathy after adjusting for reticulocytosis (OR 0.50, 95% CI 0.26–0.96, p  < 0.05) [ 44 ].

Management of cardiopulmonary disease

Patients with SCD who have symptoms suggestive of cardiopulmonary disease, such as worsening dyspnea, hypoxemia or reduced exercise tolerance, should be evaluated with a diagnostic ECHO and PFT. The presence of snoring, witnessed apnea, respiratory pauses or hypoxemia during sleep, daytime somnolence or nocturnal enuresis in older children and adults is sufficient for a diagnostic sleep study.

Without treatment, the mortality rate in SCD patients with PH is high compared to those without (5-year, all-cause mortality rate of 32 vs. 16%, p  < 0.001) [ 33 ]. PAH-targeted therapies should be considered for SCD patients with PAH confirmed by right-heart catheterization. However, the only RCT ( n  = 6) in individuals with SCD and PAH confirmed by right-heart catheterization (bosentan versus placebo) was stopped early for poor accrual with no efficacy endpoints analyzed [ 45 ]. In SCD patients with elevated peak TRJV, a randomized controlled trial ( n  = 74) of sildenafil, a phosphodiesterase-5 inhibitor, was discontinued early due to increased pain events in the sildenafil versus placebo arm (35 vs. 14%, p  = 0.029) with no treatment benefit [ 46 ]. Despite absence of clinical trial data, patients with SCD and confirmed PH should be considered for hydroxyurea or monthly red blood cell transfusions given their disease-modifying benefits. In a retrospective analysis of 13 adults with SCD and PAH, 77% of patients starting at a New York Heart Association (NYHA) functional capacity class III or IV achieved class I/II after a median of 4 exchange transfusions with improvement in median pulmonary vascular resistance (3.7 vs. 2.8 Wood units, p  = 0.01) [ 47 ].

Approximately 28% of children with SCD have asthma, which is associated with increased pain episodes that may result from impaired oxygenation leading to sickling and vaso-occlusion as well as with acute chest syndrome and higher mortality [ 48 – 50 ]. First line therapies include standard beta-adrenergic bronchodilators and supplemental oxygen as needed. When corticosteroids are indicated, courses should be tapered over several days given the risk of rebound SCD pain from abrupt discontinuation. Inhaled corticosteroids such as fluticasone proprionate or beclomethasone diproprionate are reserved for patients with recurrent asthma exacerbations, but their anti-inflammatory effects and impact on preventing pain episodes in patients with SCD who do not have asthma is under investigation [ 51 ]. Finally, management of sleep-disordered breathing is tailored to findings on formal sleep study in consultation with a sleep/pulmonary specialist.

Central nervous system (CNS) complications

CNS complications, such as overt and silent cerebral infarcts, cause significant morbidity in individuals with SCD. Eleven percent of patients with HbSS disease by age 20 years and 24% by age 45 years will have had an overt stroke [ 52 ]. Silent cerebral infarcts occur in 39% by 18 years and in > 50% by 30 years [ 53 , 54 ]. Patients with either type of stroke are at increased risk of recurrent stroke [ 55 ]. Overt stroke involves large-arteries, including middle cerebral arteries and intracranial internal carotid arteries, while silent cerebral infarcts involve penetrating arteries. The pathophysiology of overt stroke includes vasculopathy, increased sickled red blood cell adherence, and hemolysis-induced endothelial activation and altered vasomotor tone [ 56 ]. Overt strokes present as weakness or paresis, dysarthria or aphasia, seizures, sensory deficits, headache or altered level of consciousness, while silent cerebral infarcts are associated with cognitive deficits, including lower IQ and impaired academic performance.

Diagnosis of CNS complications in SCD

Overt stroke is diagnosed by evidence of acute infarct on brain MRI diffusion-weighted imaging and focal deficit on neurologic exam. A silent cerebral infarct is defined by a brain “MRI signal abnormality at least 3 mm in one dimension and visible in 2 planes on fluid-attenuated inversion recovery (FLAIR) T2-weighted images” and no deficit on neurologic exam [ 57 ]. Since silent cerebral infarcts cannot be detected clinically, a screening baseline brain MRI is recommended in school-aged children with SCD [ 58 ]. Recent SCD clinical practice guidelines also suggest a screening brain MRI in adults with SCD to facilitate rehabilitation services, patient and family understanding of cognitive deficits and further needs assessment [ 58 ]. An MRA should be added to screening/diagnostic MRIs to evaluate for cerebral vasculopathy (e.g., moyamoya), which may increase risk for recurrent stroke or hemorrhage [ 59 ].

Annual screening for increased stroke risk by transcranial doppler (TCD) ultrasound is recommended by the American Society of Hematology for children 2–16 years old with HbSS or HbS/β° thalassemia [ 58 ]. Increased stroke risk on non-imaging TCD is indicated by abnormally elevated cerebral blood flow velocity, defined as ≥ 200 cm/s (time-averaged mean of the maximum velocity) on 2 occasions or a single velocity of > 220 cm/s in the distal internal carotid or proximal middle cerebral artery [ 60 ]. Many centers rely on imaging TCD, which results in velocities 10–15% lower than values obtained by non-imaging protocols and therefore, require adjustments to cut-offs for abnormal velocities. Data supporting stroke risk assessment using TCD are lacking for adults with SCD and standard recommendations do not exist.

Neurocognitive deficits occur in over 30% of children and adults with severe SCD [ 61 , 62 ]. These occur as a result of overt and/or silent cerebral infarcts but in some patients, the etiology is unknown. The Bright Futures Guidelines for Health Supervision of Infants, Children and Adolescents or the Cognitive Assessment Toolkit for adults are commonly used tools to screen for developmental delays or neurocognitive impairment [ 58 ]. Abnormal results should prompt referral for formal neuropsychological evaluation, which directs the need for brain imaging to evaluate for silent cerebral infarcts and facilitate educational/vocational accommodations.

Management of CNS complications

Monthly chronic red blood cell transfusions to suppress HbS < 30% are standard of care for primary stroke prevention in children with an abnormal TCD. In an RCT of 130 children, chronic transfusions, compared to no transfusions, were associated with a difference in stroke risk of 92% (1 vs. 10 strokes, p  < 0.001) [ 60 ]. However, children with abnormal TCD and no MRI/MRA evidence of cerebral vasculopathy can safely transition to hydroxyurea after 1 year of transfusions [ 63 ]. Lifelong transfusions to maintain HbS < 30% remain standard of care for secondary stroke prevention in individuals with overt stroke [ 64 ]. Chronic monthly red blood cell transfusions should also be considered for children with silent cerebral infarct [ 58 ]. In a randomized controlled trial ( n  = 196), monthly transfusions, compared to observation without hydroxyurea, reduced risk of overt stroke, new silent cerebral infarct or enlarging silent cerebral infarct in children with HbSS or HbS/β 0 thalassemia and an existing silent cerebral infarct (2 vs. 4.8 events, incidence rate ratio of 0.41, 95% CI 0.12–0.99, p  = 0.04) [ 57 ].

Acute stroke treatment requires transfusion therapy to increase cerebral oxygen delivery. Red blood cell exchange transfusion, defined as replacement of patients’ red blood cells with donor red blood cells, to rapidly reduce HbS to < 30% is the recommended treatment as simple transfusion alone is shown to have a fivefold greater relative risk (57 vs. 21% with recurrent stroke, RR = 5.0; 95% CI 1.3–18.6) of subsequent stroke compared to exchange transfusion [ 65 ]. However, a simple transfusion is often given urgently while preparing for exchange transfusion [ 58 ]. Tissue plasminogen activator (tPA) is not recommended for children with SCD who have an acute stroke since the pathophysiology of SCD stroke is less likely to be thromboembolic in origin and there is risk for harm. Since the benefits and risks of tPA in adults with SCD and overt stroke are not clear, its use depends on co-morbidities, risk factors and stroke protocols but should not delay or replace prompt transfusion therapy.

Data guiding treatment of SCD cerebral vasculopathy (e.g., moyamoya) are limited, and only nonrandomized, low-quality evidence exists for neurosurgical interventions (e.g., encephaloduroarteriosynangiosis) [ 66 ]. Consultation with a neurosurgeon to discuss surgical options in patients with moyamoya and history of stroke or transient ischemic attack should be considered [ 58 ].

Kidney disease

Glomerulopathy, characterized by hyperfiltration leading to albuminuria, is an early asymptomatic manifestation of SCD nephropathy and worsens with age. Hyperfiltration, defined by an absolute increase in glomerular filtration rate, may be seen in 43% of children with SCD [ 67 ]. Albuminuria, defined by the presence of urine albumin ≥ 30 mg/g over 24 h, has been observed in 32% of adults with SCD [ 68 ]. Glomerulopathy results from intravascular hemolysis and endothelial dysfunction in the renal cortex. Medullary hypoperfusion and ischemia also contribute to kidney disease in SCD, causing hematuria, urine concentrating defects and distal tubular dysfunction [ 69 ]. Approximately 20–40% of adults with SCD develop chronic kidney disease (CKD) and are at risk of developing end-stage renal disease (ESRD), with rapid declines in estimated glomerular filtration rate (eGFR) > 3 mL/min/1.73 m 2 associated with increased mortality (HR 2.4, 95% CI 1.31–4.42, p  = 0.005) [ 68 ].

Diagnosis of kidney disease in SCD

The diagnosis of sickle cell nephropathy is made by detecting abnormalities such as albuminuria, hematuria or CKD rather than by distinct diagnostic criteria in SCD, which have not been developed. Traditional markers of kidney function such as serum creatinine and eGFR should be interpreted with caution in individuals with SCD because renal hyperfiltration affects their accuracy by increasing both. Practical considerations preclude directly measuring GFR by urine or plasma clearance techniques, which achieves the most accurate results. The accuracy of eGFR, however, may be improved by equations that incorporate serum cystatin C [ 70 ].

Since microalbuminuria/proteinuria precedes CKD in SCD, annual screening for urine microalbumin/protein is recommended beginning at age 10 years [ 71 ]. When evaluating urine for microalbumin concentration, samples from first morning rather than random voids are preferable to exclude orthostatic proteinuria. Recent studies suggest HMOX1 and APOL1 gene variants may be associated with CKD in individuals with SCD [ 72 ]. Potential novel predictors of acute kidney injury in individuals with SCD include urine biomarkers kidney injury molecule 1 (KIM-1) [ 73 ], monocyte chemotactic protein 1 (MCP-1) [ 74 ] and neutrophil gelatinase-associated lipocalin (NGAL) [ 75 ]. Their contribution to chronic kidney disease and interaction with other causes of kidney injury in SCD (e.g., inflammation, hemolysis) are not clear.

Management of kidney disease

Managing kidney complications in SCD should focus on mitigating risk factors for acute and chronic kidney injury such as medication toxicity, reduced kidney perfusion from hypotension and dehydration, and general disease progression, as well as early screening and treatment of microalbuminuria/proteinuria. Acute kidney injury, either an increase in serum creatinine ≥ 0.3 mg/dL or a 50% increase in serum creatinine from baseline, is associated with ketorolac use in children with SCD hospitalized for pain [ 76 ]. Increasing intravenous fluids to maintain urine output > 0.5 to 1 mL/kg/h and limiting NSAIDs and antibiotics associated with nephrotoxicity in this setting are important. Despite absence of controlled clinical trials, hydroxyurea may be associated with improvements in glomerular hyperfiltration and urine concentrating ability in children with SCD [ 77 , 78 ]. Hydroxyurea is also associated with a lower prevalence (34.7 vs. 55.4%, p  = 0.01) and likelihood of albuminuria (OR 0.28, 95% CI 0.11–0.75, p  = 0.01) in adults with SCD after adjusting for age, angiotensin-converting enzyme inhibitor (ACE-I)/angiotensin receptor blockade (ARB) use and major disease risk factors [ 79 ].

ACE-I or ARB therapy reduces microalbuminuria in patients with SCD. In a phase 2 trial of 36 children and adults, a ≥ 25% reduction in urine albumin-to-creatinine ratio was observed in 83% ( p  < 0.0001) and 58% ( p  < 0.0001) of patients with macroalbuminuria (> 300 mg/g creatinine) and microalbuminuria (30–300 mg/g creatinine), respectively, after 6 months of treatment with losartan at a dose of 0.7 mg/kg/day (max of 50 mg) in children and 50 mg daily in adults [ 80 ]. However, ACE-I or ARB therapy has not been shown to improve kidney function or prevent CKD. Hemodialysis is associated with a 1-year mortality rate of 26.3% after starting hemodialysis and an increase risk of death in SCD patients with ESRD compared to non-SCD patients with ESRD (44.6 vs. 34.5% deaths, mortality hazard ratio of 2.8, 95% CI 2.31–3.38) [ 81 ]. Renal transplant should be considered for individuals with SCD and ESRD because of recent improvements in renal graft survival and post-transplant mortality [ 82 ].

Disease-modifying therapies in SCD

Since publication of its landmark trial in 1995, hydroxyurea continues to represent a mainstay of disease-modifying therapy for SCD. Hydroxyurea induces fetal hemoglobin production through stress erythropoiesis, reduces inflammation, increases nitric oxide and decreases cell adhesion. The FDA approved hydroxyurea in 1998 for adults with SCD. Subsequently, hydroxyurea was FDA approved for children in 2017 to reduce the frequency pain events and need for blood transfusions in children ≥ 2 years of age [ 63 ]. The landscape of disease-modifying therapies, however, has improved with the recent FDA approval of 3 other treatments— l -glutamine and crizanlizumab for reducing acute complications (e.g., pain), and voxelotor for improving anemia (Table ​ (Table3) 3 ) [ 83 – 85 ]. Other therapies in current development focus on inducing fetal hemoglobin, reducing anti-sickling or cellular adhesion, or activating pyruvate kinase-R.

Major FDA-approved therapies for the treatment of SCD

Drug and FDA approvalFDA approval date and indicationsMechanism of actionDosingCommon adverse effects
Hydroxyurea

1998:

Adults to reduce frequency of painful crises

2017:

Children ≥ 2 years to reduce frequency of painful crises and need for blood transfusions

↑ Fetal Hb via temporary arrest of hematopoiesis and stress erythropoiesis

↓ Inflammation through ↓ in WBC and platelets

↓ Adhesion molecule expression

↑ Nitric oxide production

Usual starting dose 20 mg/kg/day

Dose escalate to maximum tolerated dose (~ 30–35 mg/kg/day)

Alternatively, dose escalate to absolute neutrophil count of 1500–2000/µL

Neutropenia (13%)

Thrombocytopenia (7%)

Nausea (3%)

l-glutamine

2017:

Children and adults ≥ 5 years old to reduce severe complications (sickle cell crises and acute chest syndrome)

↑ NAD redox potential in sickle red blood cells

Protects red blood cells from oxidative stress

Dose by weight

 < 30 kg—1 packet (5 g) BID

30–65 kg—2 packets (10 g) BID

 > 65 kg—3 packets (15 g) BID

May take with or without hydroxyurea

Constipation (21%)

Nausea (19%)

Abdominal pain (17%)

Headache (18%)

Cough (16%)

Crizanlizumab

2019:

Adolescents and adults ≥ 16 years old to reduce frequency of vaso-occlusive crises

Binds to P-selectin

Blocks interactions with ligands, including P-selectin glycoprotein ligand 1

5 mg/kg/dose IV on weeks 0, 2 and every 4 weeks thereafter

May take with or without hydroxyurea

Infusion-related adverse events (< 10%)

Nausea (18%)

Arthralgia (18%)

Back pain (15%)

Fever (11%)

Voxelotor

2019:

Children and adults ≥ 12 years old to increase hemoglobin concentration

Allosteric modifier of hemoglobin to stabilize oxygenated state

↓ Sickle Hb polymerization

↓ Hemolysis

1500 mg po daily

May take with or without hydroxyurea

Headache (26%)

Diarrhea (20%)

Nausea (17%)

Abdominal pain (19%)

Skin rash (14%)

Fever (12%)

Fatigue (14%)

l -glutamine

Glutamine is required for the synthesis of glutathione, nicotinamide adenine dinucleotide and arginine. The essential amino acid protects red blood cells against oxidative damage, which forms the basis for its proposed utility in SCD. The exact mechanism of benefit in SCD, however, remains unclear. In a phase 3 RCT of 230 participants (hemoglobin SS or S/β 0 thalassemia), l -glutamine compared to placebo was associated with fewer pain events (median 3 vs. 4, p  = 0.005) and hospitalizations for pain (median 2 vs. 3, p  = 0.005) over the 48-week treatment period [ 84 ]. The percentage of patients who had at least 1 episode of acute chest syndrome, defined as presence of chest wall pain with fever and a new pulmonary infiltrate, was lower in the l -glutamine group (8.6 vs. 23.1%, p  = 0.003). There were no significant between-group differences in hemoglobin, hematocrit or reticulocyte count. Common side effects of l -glutamine include GI upset (constipation, nausea, vomiting and abdominal pain) and headaches.

Crizanlizumab

P-selectin expression, triggered by inflammation, promotes adhesion of neutrophils, activated platelets and sickle red blood cells to the endothelial surface and to each other, which promotes vaso-occlusion in SCD. Crizanlizumab, given as a monthly intravenous infusion, is a humanized monoclonal antibody that binds P-selectin and blocks the adhesion molecule’s interaction with its ligand, P-selectin glycoprotein ligand 1. FDA approval for crizanlizumab was based on a phase 2 RCT ( n  = 198, all genotypes), in which the median rate of pain events (primary endpoint) was lower (1.63 vs. 2.68, p  = 0.01) and time to first pain event (secondary endpoint) was longer (4.07 vs. 1.38 months, p  = 0.001) for patients on high-dose crizanlizumab (5 mg/kg/dose) compared to placebo treated for 52 weeks (14 doses total) [ 83 ]. In this trial, patients with SCD on chronic transfusion therapy were excluded, but those on stable hydroxyurea dosing were not. Adverse events were uncommon but included headache, back pain, nausea, arthralgia and pain in the extremity.

Polymerization of Hb S in the deoxygenated state represents the initial step in red blood cell sickling, which leads to reduced red blood cell deformability and increased hemolysis. Voxelotor is a first-in-class allosteric modifier of Hb S that increases oxygen affinity. The primary endpoint for the phase 3 RCT of voxelotor ( n  = 274, all genotypes) that led to FDA approval was an increase in hemoglobin of at least 1 g/dL after 24 weeks of treatment [ 85 ]. More participants receiving 1500 mg daily of oral voxelotor versus placebo had a hemoglobin response of at least 1 g/dL (51%, 95% CI 41–61 vs. 7%, 95% CI 1–12, p < 0.001). Approximately 2/3 of the participants in these trials were on hydroxyurea, with treatment benefits observed regardless of hydroxyurea status. Despite improvements associated with voxelotor in biomarkers of hemolysis (reticulocyte count, indirect bilirubin and lactate dehydrogenase), annualized incidence rate of vaso-occlusive crisis was not significantly different among treatment groups. Adverse events included headaches, GI symptoms, arthralgia, fatigue and rash.

Curative therapies in SCD

For individuals with SCD undergoing hematopoietic stem cell transplantation (HSCT) using HLA-matched sibling donors and either myeloablative or reduced-intensity conditioning regimens, the five-year event-free and overall survival is high at 91% and 93%, respectively [ 86 ]. Limited availability of HLA-matched sibling donors in this population requires alternative donors or the promise of autologous strategies such as gene-based therapies (i.e. gene addition, transfer or editing) (Table ​ (Table4). 4 ). Matched unrelated donors have not been used routinely due to increased risk of graft-versus-host disease (GVHD) as high as 19% (95% CI 12–28) in the first 100 days for acute GVHD and 29% (95% CI 21–38) over 3 years for chronic GVHD [ 87 ]. Haplo-identical HSCT, using biological parents or siblings as donors, that incorporate post-transplant cyclophosphamide demonstrates acceptable engraftment rates, transplant-related morbidity and overall mortality [ 88 ]. Regardless of allogeneic HSCT type, older age is associated with lower event-free (102/418 vs. 72/491 events, HR 1.74, 95% CI 1.24–2.45) and overall survival (54/418 vs. 22/491 events, HR 3.15, 95% CI 1.86–5.34) in patients ≥ 13 years old compared to < 12 years old undergoing HSCT [ 87 ].

Therapies with curative intent

HLA-matched sibling donor

Standard approaches rely on myeloablative conditioning with overall and disease-free survival rates in the 90% range for children and young adults with SCD

Reduced intensity and reduced toxicity, nonmyeloablative regimens offer alternative strategies to achieve stable, mixed chimerism in adults with SCD

Umbilical cord donor

Cord units from related or unrelated donors represent alternative sources of hematopoietic stem cells for children with SCD; routine use is limited by lower total cell dose

Units from unrelated donors are associated with higher risk of graft rejection and graft-versus-host disease

HLA-matched unrelated donor

Demographics of current donor pool limit this option for Black patients with SCD

High rates of acute and chronic graft-versus-host disease remain a significant challenge

Haploidentical donor

Haploidentical donors offer most accessible donor type for children and adults with SCD

Strategies focused on T-cell depletion using post-transplant cyclophosphamide have improved engraftment rates and reduced graft-versus-host disease

Gene addition or transfer

Lentiviral-based vector encodes modified β- or γ-globin transgenes to increase anti-sickling hemoglobin production

Lentiviral-based vector transfers short-hairpin RNA (shRNA) targeting to increase γ-globin expression

Transduction efficiency is high with current vectors

Concerns remain about risk of insertional oncogenesis and long-term high-level expression

Gene editing

Genome editing focuses on correction of SCD mutation, downregulation of or mimicking of hereditary persistence of fetal hemoglobin (HPFH) mutations

Strategies rely on CRISPR-Cas9 technology or engineered DNA-cleaving enzymes such as zinc-finger nucleases (ZFNs) or transcription activator-like effector nucleases (TALENs)

Advancing research in SCD

Despite progress to date, additional high-quality, longitudinal data are needed to better understand the natural history of the disease and to inform optimal screening for SCD-related complications. In the era of multiple FDA-approved therapies with disease-modifying potential, clinical trials to evaluate additional indications and test them in combination with or compared to each other are needed. Dissemination and implementation studies are also needed to identify barriers and facilitators related to treatment in everyday life, which can be incorporated into decision aids and treatment algorithms for patients and their providers [ 89 ]. Lastly, continued efforts should acknowledge social determinants of health and other factors that affect access and disease-related outcomes such as the role of third-party payers, provider and patient education, health literacy and patient trust. Establishing evidence-derived quality of care metrics can also drive public policy changes required to ensure care optimization for this population.

Conclusions

SCD is associated with complications that include acute and chronic pain as well as end-organ damage such as cardiopulmonary, cerebrovascular and kidney disease that result in increased morbidity and mortality. Several well-designed clinical trials have resulted in key advances in management of SCD in the past decade. Data from these trials have led to FDA approval of 3 new drugs, l -glutamine, crizanlizumab and voxelotor, which prevent acute pain and improve chronic anemia. Moderate to high-quality data support recommendations for managing SCD cerebrovascular disease and early kidney disease. However, further research is needed to determine the best treatment for chronic pain and cardiopulmonary disease in SCD. Comparative effectiveness research, dissemination and implementation studies and a continued focus on social determinants of health are also essential.

Acknowledgements

We would like to acknowledge Lana Mucalo, MD, for supporting data collection for this manuscript.

Abbreviations

6-MWDSix-minute walk distance
ACE-IAngiotensin-converting enzyme inhibitor
ARBAngiotensin receptor blockade
CBTCognitive behavioral therapy
CKDChronic kidney disease
COTChronic opioid therapy
ECHOEchocardiogram
ESRDEnd stage renal disease
FLAIRFluid-attenuated inversion recovery
GFRGlomerular filtration rate
GVHDGraft-versus-host disease
HbSHemoglobin S
HSCTHematopoietic stem cell transplant
NSAIDsNonsteroidal anti-inflammatory drugs
NT-proBNPN-terminal pro-brain natriuretic peptide
NYHANew York Heart Association
PAHPulmonary arterial hypertension
PFTPulmonary function test
PHPulmonary hypertension
PPVPositive predictive value
PROsPatient-reported outcomes
RCTRandomized controlled trial
SCDSickle cell disease
SNRIsSerotonin and norepinephrine reuptake inhibitors
TCAsTricyclic antidepressants
TCDTranscranial Doppler
tPATissue plasminogen activator
TRJVTricuspid regurgitant jet velocity
VASVisual Analog Scale

Authors' contributions

AB and RL contributed equally to the writing and editing of this manuscript. All authors read and approved the final manuscript.

Availability of data and materials

Declarations.

The authors have no competing interests to declare.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

NIH Research Festival

September 23 – 25, 2024

Leveraging the Townes mice to study interactions between sickle cell disease and schistosome (human blood fluke worm) infection

  • JPK Mukendi
  • O Kamenyeva
  • ZMN Quezado

Background: Sickle cell disease (SCD) is an inherited hematologic disease due to a single nucleotide mutation in the beta chain of human hemoglobin (Hb) that produces defective Hb called HbS. Hypoxic conditions make HbS polymerize, leading to red blood cells sickling and hemolyzing. Most disease cases are in sub-Saharan Africa, where schistosomiasis, a worm infection causing severe inflammation and anemia, is also endemic. However, possible interactions between SCD and schistosomiasis remain largely unknown. This study used the Townes mouse model of SCD to determine the interactions between SCD and schistosome infection. Methods: Townes mice were infected with Schistosoma mansoni for eight weeks. Hematologic parameters were analyzed before and after infection; histology of the liver and spleen and S. mansoni egg counts were performed. Liver microcirculation and tissue were also intravitally imaged. Results: S. mansoni infection increased granulocytes (neutrophils, p=0.0005; eosinophils, p<0.0001; basophils, p<0.0001) and monocyte (p<0.0001) counts in peripheral blood but did not worsen anemia in HbSS mice (RBC, p=0.3382; Hb, p=0.7000, Hct, p=0.4590). However, it significantly decreased hepatic hemosiderosis (p<0.0001) in HbSS mice. On the contrary, SCD did not affect peri-oval granuloma formation in the liver of infected mice but caused a slight reduction of S. mansoni egg burden. Conclusion: Townes mice are susceptible to S. mansoni infection and, therefore, constitute a good model for mechanistically characterizing the interactions between SCD and schistosomiasis. This study stands as a proof of concept for the use of Townes mice in the study of worm infections in SCD and warrants further investigations.

Scientific Focus Area : Microbiology and Infectious Diseases

This page was last updated on Tuesday, August 6, 2024

Targeted therapeutic management based on phytoconstituents for sickle cell anemia focusing on molecular mechanisms: Current trends and future perspectives

  • August 2024
  • Phytomedicine
  • CC BY-NC-ND 4.0

Md. Rezaul Islam at Daffodil International University

  • Daffodil International University

Abdur Rauf at University of Swabi

  • University of Swabi

Shopnil Akash

  • This person is not on ResearchGate, or hasn't claimed this research yet.

Discover the world's research

  • 25+ million members
  • 160+ million publication pages
  • 2.3+ billion citations

No full-text available

Request Full-text Paper PDF

To read the full-text of this research, you can request a copy directly from the authors.

Hauwa Ali Buhari

  • Aisha Sa'ad Ahmad

Emmanuel Ifeanyi Obeagu

  • EVID-BASED COMPL ALT

Apollinaire Teguem Tchoulegheu

  • Prudence Josela Nya Nkwikeu
  • Natacha Lena Yembeau

Anatole Constant Pieme

  • Felix E Okieimen
  • Clifford O Ehisuoria

Ngozi O. A. Imaga

  • Oluwole Taiwo

Emmanuel Adase

  • David Okata Asamoah Agyare

Angela Snyder

  • Sangeetha Lakshmanan
  • Mary Hulihan

Laura Schieve

  • Nafisah Bisallah Lawal
  • Aisha M Alhassan

Sakariyau A. Waheed

  • Francisca Chioma Christopher

Ridwan Olamilekan Adesola

  • Ibikun Mary
  • Shengai Zhang
  • Zunmiao Wei
  • Zhongsheng Mu

Felipe Machado Mota

  • J HEMATOL ONCOL

Amanda M Brandow

  • Robert I. Liem

Carolina Mariano Pompeo

  • Assaf Malik

Sangeetha Thangaswamy

  • Craig A Branch
  • Kamalakar Ambadipudi
  • Seetharama A Acharya

Shringika Mishra

  • Maimuna Haruna
  • Eman Ali Abuagla Dafaalla

Amira Humeida

  • J TRANSL MED

Ibrahim Said Khamees

  • GENET RESOUR CROP EV
  • Gregorio Hernández-Salinas

Mario Luna-Cavazos

  • Leobigildo Córdova-Téllez
  • Wjdan A. Arishi

Hani A. Alhadrami

  • Gilberto Terra

Marco Antonio Monte

  • Joseph M. Agbedahunsi

Ahmad Hazzazi

  • Abdulaziz Mohammed Alfaqih
  • Hafiz Malhan

Taofeeq Oduola

  • F A A Adeniyi
  • E. O. Ogunyemi
  • BMC INFECT DIS
  • Ngo Linwa Esther Eleonore

Samuel Nambile Cumber

  • Thomas N Williams

Karim eldin Mohamed Ali Salih

  • Renata Mírian Nunes Eleutério
  • Francisco O. F. Nascimento
  • Tamara Gonçalves de Araújo
  • Romélia Pinheiro Gonçalves Lemes
  • Carinna Hockham
  • Samir Bhatt

Roshan Colah

  • Frédéric B Piel

Christianah Abimbola Elusiyan

  • Opeyemi Ayoade

Adewale O Adeloye

  • Clarice D. Reid

Elliott Vichinsky

  • Sudama Rathore

Viplav Prashant

  • Dr. Dolly Prashant
  • Shehu Umar Abdullahi

Adeseyemichael Akinsete

  • Eleanor A Lisbon
  • Santosh L. Saraf

David C Rees

  • Akakpo-Akue Joel

Tatiana KANGAH MIREILLE Kple

  • KRA Adou Koffi Mattieu

Joseph Djaman

  • Babatunde M. Williams
  • Mojisola C. Cyrl-Olutayo
  • J Am Med Assoc
  • Patricia L. Kavanagh
  • Titilope Fasipe
  • R. Clark Clark Brown

Andrew D Redfern

  • Eleanor Lisbon
  • Kim Smith-Whitley

Kevin Esoh

  • Kobina Dufu
  • Carsten Alt
  • Steven Strutt
  • Donna Oksenberg
  • TRANSFUS CLIN BIOL
  • Saliou Diop

France Pirenne

  • Christina Chapman
  • W Craig Hooper

Jin Han

  • Subcell Biochem

Amit Mandal

  • Recruit researchers
  • Join for free
  • Login Email Tip: Most researchers use their institutional email address as their ResearchGate login Password Forgot password? Keep me logged in Log in or Continue with Google Welcome back! Please log in. Email · Hint Tip: Most researchers use their institutional email address as their ResearchGate login Password Forgot password? Keep me logged in Log in or Continue with Google No account? Sign up

SicklED – Advocacy for Sickle Cell Education and Diagnosis

  • Sierra Leone 2024

Day 2: Visiting PHUs + Interviews

To begin our day, we traveled out to the Panlap PHU with our driver William. As we drove through the market of Makeni, people were walking everywhere. The stores of Makeni were bustling with activity as William weaved through traffic. Once we had passed the market, the activity slowly waned as we approached the PHU. With one final turn, we arrived at Panlap. Our team walked into the meeting room and explained to the head nurse that we were from Lehigh University and wished to interview a few staff members about sickle cell disease and their past experience. We rearranged the room to allow for better communication and, to our surprise, we were met with six nurses, all eager to participate. In talking with the PHU staff, the awareness and impact of sickle cell was made clear. 

The nurses emphasized some of the public’s practices, such as binding their limbs or applying heat using cooking stones. As we talked about the prevalence of sickle cell disease, its different nature than malaria, and how often PHU caregivers receive feedback from hospital referrals, we began to hand out copies of our new flipbooks. We hoped to receive constructive feedback from workers in the field to refine and improve upon our pilot version. Some recommendations they provided were the types of food available here in Makeni, to encourage not to smoke cigarettes, and advising not to drink iced water. Overall, they seemed to find our flipbook to have potential to be a strong presenting tool during outreach clinics. Although we requested the books not be used as they are only drafts and currently unapproved on the government level, we did choose to leave one copy behind for the nurses. In addition, we explained our lateral flow test project happening at home, showing the most recent pictures with lines and its ability to screen for sickle cell right in the clinic. With many “thank you”s, our team went outside to take one last picture with all of the nurses we had interviewed. 

current research on sickle cell anemia 2022

After our visit to the Panlap PHU, we traveled to the Yoni PHU. At the clinic we were welcomed by a member of the clinic and were soon greeted by the head nurse. The head nurse was a useful contact for the team last year and brought the previous team to an outreach clinic in a rural community. At the outreach clinic the nurse had brought a flipbook on Malaria and presented it to the community. This inspired the team to create the flipbook on Sickle Cell Disease. In the interview, the nurse shared her knowledge of Sickle Cell Disease. She explained that her daughter has Sickle Cell Disease so she shared her own experience of the diagnosing and management processes. She shared that money was necessary for the diagnosis and management, making the option inaccessible for many people. To help manage it, she was given medicine for free, although much of it had expired so she stopped giving it to her daughter. She also provides her daughter with pain medication and ensures her daughter is eating plenty of leafy greens. 

The nurse said that the doctors and healthcare workers have an understanding of Sickle Cell Disease and that efforts should be focused on diagnosis and treatment. She also mentioned that although sickle cell disease affects much of the community, there is a lot of misinterpretation and lack of information about the disease within communities. In areas where there are members known to have sickle cell, the nurse encourages them to plant a garden composed of leafy greens for those with sickle cell to ensure they are eating nutrient rich food. 

We then turned our attention to our flipbook. The nurse gave us very helpful feedback on our current draft. Her suggestions included editing some of the pictures to show the leafy green leaves that are readily available in Sierra Leone as well as changing the expression and depiction of some of the characters to make the pictures more representative of the disease with easier interpretation. 

After the interview, we made our way back to the World Hope Office to continue planning, debrief the morning, and catch up on paperwork. 

Leave a Reply Cancel reply

You must be logged in to post a comment.

IMAGES

  1. Rheumatological Manifestations of Sickle Cell Anemia

    current research on sickle cell anemia 2022

  2. Sickle Cell Anemia Mutation

    current research on sickle cell anemia 2022

  3. (PDF) Sickle Cell Anemia

    current research on sickle cell anemia 2022

  4. (PDF) Seminar on Sickle Cell Anemia

    current research on sickle cell anemia 2022

  5. Sickle Cell Anaemia

    current research on sickle cell anemia 2022

  6. Sickle Cell Disease

    current research on sickle cell anemia 2022

VIDEO

  1. New research in sickle cell anemia

  2. Breakthrough treatment for sickle cell anemia patients

  3. Mitanin CHW Video on Sickle Cell Anemia 2022

  4. Sickle Cell Anemia groundbreaking research in Mineral Balancing & Genetic upgrades & development

  5. Sickle Cell Anemia role of Minerals in the body part 1

  6. Future Frontiers: Research on Sickle Cell Anemia Disease

COMMENTS

  1. Sickle Cell Disease Research

    The National Institutes of Health (NIH) has supported research on sickle cell disease since before the NHLBI was founded in 1948. With each decade that followed, the NHLBI has kept a sustained focus on advancing the understanding of sickle cell disease and improving clinical care. We lead and support research and programs on sickle cell disease in the United States and around the world ...

  2. Researchers study a new way to treat sickle cell disease

    Researchers study a new way to treat sickle cell disease. June 16, 2022. Activating a protein in red blood cells may improve anemia and alleviate acute episodes of severe pain for people living with sickle cell disease. Swee Lay Thein, M.B., D.Sc., a senior investigator and chief of NHLBI's Sickle Cell Branch, shares insight into a decade ...

  3. Advances in the diagnosis and treatment of sickle cell disease

    Sickle cell disease (SCD), which affects approximately 100,000 individuals in the USA and more than 3 million worldwide, is caused by mutations in the βb globin gene that result in sickle hemoglobin production. Sickle hemoglobin polymerization leads to red blood cell sickling, chronic hemolysis and vaso-occlusion. Acute and chronic pain as well as end-organ damage occur throughout the ...

  4. Clinical genome editing to treat sickle cell disease—A brief update

    Sickle cell disease (SCD) is one of the most common hemoglobinopathies, which comprises a group of disorders that are characterized by faulty hemoglobin production ( 1, 2 ). Hemoglobin, a two-way respiratory carrier in red blood cells (RBCs), is responsible for transporting oxygen to tissues and returning carbon dioxide to the lung.

  5. Gene therapies close in on a cure for sickle-cell disease

    Sickle-cell disease presents a near-ideal opportunity to tap the power of gene therapy because the disorder typically arises from a mutation in a single nucleotide in one gene. That gene encodes ...

  6. FDA approves cure for sickle cell disease, the first treatment to use

    The FDA approved a new treatment for sickle cell disease. The therapy is first to use the ground-editing tool CRISPR.

  7. Biologic and Clinical Efficacy of LentiGlobin for Sickle Cell Disease

    Sickle cell disease is characterized by the painful recurrence of vaso-occlusive events. Gene therapy with the use of LentiGlobin for sickle cell disease (bb1111; lovotibeglogene autotemcel) consis...

  8. NIH statement on new FDA-approved gene therapies for sickle cell

    Today, the U.S. Food and Drug Administration (FDA) approved two gene therapies for the treatment of sickle cell disease in patients 12 years and older. About 100,000 Americans and millions of people around the world have sickle cell disease , a hereditary disease common among those whose ancestors come from sub-Saharan Africa, Mediterranean countries, India and the Middle East.

  9. Recent Advances in the Treatment of Sickle Cell Disease

    In 1949, Linus Pauling showed that an abnormal protein (hemoglobin S, HbS) was the cause of sickle cell anemia (SCA), making SCD the first molecular disease and motivating an enormous amount of scientific and medical research.

  10. Sickle cell disease

    Sickle cell disease is an autosomal recessive blood disorder that can lead to anaemia. It is caused by a mutation in the haemoglobin gene, which leads to deformation of red blood cells. Deformed ...

  11. Revisiting anemia in sickle cell disease and finding the balance with

    Abstract Chronic hemolytic anemia and intermittent acute pain episodes are the 2 hallmark characteristics of sickle cell disease (SCD). Anemia in SCD not only signals a reduction of red cell mass and oxygen delivery, but also ongoing red cell breakdown and release of cell-free hemoglobin, which together contribute to a number of pathophysiological responses and play a key role in the ...

  12. Effective therapies for sickle cell disease: are we there yet?

    Sickle cell disease (SCD) is a common genetic blood disorder associated with acute and chronic pain, progressive multiorgan damage, and early mortality. Recent advances in technologies to manipulate the human genome, a century of research and the development of techniques enabling the isolation, efficient genetic modification, and reimplantation of autologous patient hematopoietic stem cells ...

  13. Current Research on Sickle Cell Disease

    Learn about current research efforts to treat sickle cell disease, including gene therapy and drugs that increase fetal hemoglobin.

  14. Sickle Cell Anemia News -- ScienceDaily

    Being in Space Destroys More Red Blood Cells. Jan. 14, 2022 — A world-first study has revealed how space travel can cause lower red blood cell counts, known as space anemia. Analysis of 14 ...

  15. Evidence-based management of pregnant women with sickle cell disease in

    The pathophysiology of SCD is a result of HbS in low oxygen conditions giving rise to rigid and fragile sickle-shaped red cells. 3 This leads to an increase in the breakdown of these cells, resulting in anemia and the sickle-shaped red cells polymerizing and causing the clinical features of acute pain, significant anemia, shortness of breath ...

  16. American Society of Hematology 2020 guidelines for sickle cell disease

    The most relevant guidelines are the 2014 National Heart, Lung, and Blood Institute Expert Panel Report of the Evidence-Based Management of Sickle Cell Disease, 12 the 2018 Standard for Clinical Care of Adults with Sickle Cell Disease in the United Kingdom, 168 the 2018 International Collaboration for Transfusion Medicine Guidelines on Red ...

  17. Sickle Cell Disease : A Review

    This Review summarizes the pathophysiology and diagnosis for sickle cell disease (SCD), management for SCD and its related complications, and prognosis for patients with SCD.

  18. PDF Sickle cell anemia therapy in 2023

    Pathophysiology of sickle cell disease (SCD) Redrawn from Kavanagh Pl, et al. JAMA 2022;328:57-68 PATHOPHYSIOLOGY AND COMPLICATIONS Timeline of SCD complications

  19. Small molecule shows promise in lab models for treating sickle cell

    A novel small molecule called SR-18292 reduced disease severity in a mouse model of SCD and showed anti-sickling effects in human blood cells.

  20. UCSF Sickle Cell Anemia Clinical Trials

    Mitapivat (AG-348) in Participants With Sickle Cell Disease (RISE UP) open to eligible people ages 16 years and up. This clinical trial is a Phase 2/3 study that will determine the recommended dose of mitapivat and evaluate the efficacy and safety of mitapivat in sickle cell disease by testing how well mitapivat works compared to placebo to ...

  21. PGC-1α agonism induces fetal hemoglobin and exerts ...

    Sickle cell disease (SCD) is caused by a single missense mutation in the adult β-globin gene that codes for sickle hemoglobin (HbS; α 2 β S 2).Deoxygenated HbS tends to form polymers that damage the red blood cell (RBC) causing hemolytic anemia, vaso-occlusion, and acute and chronic pain with progressive multiorgan damage, all of which lead to early mortality (1-3).

  22. Health Supervision for Children and Adolescents With Sickle Cell

    Sickle cell disease (SCD) is a group of complex genetic disorders of hemoglobin with multisystem manifestations. The scope of this clinical report is such that in-depth recommendations for management of all complications is not possible. Rather, the authors present an overview focused on the practical management of children and adolescents with SCD and the complications that are of particular ...

  23. Evolution of Extracranial Internal Carotid Artery Disease in Children

    Background: Cerebral arteriopathy in patients with sickle cell anemia mainly affects the intracranial anterior circulation. However, the extracranial internal carotid artery (eICA) can also be stenosed and responsible for ischemic lesions. In children with sickle cell anemia, we perform routine annual Doppler ultrasound assessment of the eICA and magnetic resonance imaging with 3-dimensional ...

  24. World Sickle Cell Day 2022: Progress & prospects

    World Sickle Cell Day 2022: Progress & prospects. See commentary "Biomarkers for early diagnosis of diabetic kidney disease: still a long way to go" in volume 156 on page 14. World Sickle Cell Day, an annual event held on June 19, was established by a resolution of the United Nations in 2008, almost a century after sickle cell disease was ...

  25. Sickle cell disease in India: The quest for a cure

    India has one of the highest prevalences of sickle cell ... The anemia and medical history prompted an obstetrician to order a blood test for sickle cell disease during Sanodiya's first ...

  26. Advances in the diagnosis and treatment of sickle cell disease

    Abstract Sickle cell disease (SCD), which affects approximately 100,000 individuals in the USA and more than 3 million worldwide, is caused by mutations in the βb globin gene that result in sickle hemoglobin production. Sickle hemoglobin polymerization leads to red blood cell sickling, chronic hemolysis and vaso-occlusion.

  27. Leveraging the Townes mice to study interactions between sickle cell

    Background: Sickle cell disease (SCD) is an inherited hematologic disease due to a single nucleotide mutation in the beta chain of human hemoglobin (Hb) that produces defective Hb called HbS. Hypoxic conditions make HbS polymerize, leading to red blood cells sickling and hemolyzing.

  28. Targeted therapeutic management based on phytoconstituents for sickle

    The aim of the current study was to evaluate of the anti-sickling activity of the NS extracts, forty patients with sickle cell anemia were recruited for the study.

  29. American Society of Hematology 2020 guidelines for sickle cell disease

    Background: The management of acute and chronic pain for individuals living with sickle cell disease (SCD) is a clinical challenge. This reflects the paucity of clinical SCD pain research and limited understanding of the complex biological differences between acute and chronic pain.

  30. Day 2: Visiting PHUs + Interviews

    Langer-Simon Award for Bioscience Research, 2022; Presenters US Naval Academy Science and Engineering Conference, 2023 ... Easy-to-Use, Sickle Cell Anemia Screening Device for use in Low and Middle Income Countries. Sierra Leone 2024; Day 2: Visiting PHUs + Interviews. ... The nurse gave us very helpful feedback on our current draft. Her ...