Skip to main content
Kidney360 logoLink to Kidney360
. 2023 May 31;4(7):997–1005. doi: 10.34067/KID.0000000000000162

Emerging Therapies and Advances in Sickle Cell Disease with a Focus on Renal Manifestations

Mofiyin Obadina 1,, Sam Wilson 1,2, Vimal K Derebail 3, Jane Little 1,2
PMCID: PMC10371301  PMID: 37254256

Abstract

The underlying mechanisms of disease in sickle cell disease (SCD) contribute to a multifaceted nephropathy, commonly manifested as albuminuria. In severe SCD genotypes (e.g., Hemoglobin SS [HbSS]), albuminuria and CKD are major predictors of mortality in this population. Therefore, the monitoring and management of renal function is an intrinsic part of comprehensive care in SCD. Management of nephropathy in SCD can be accomplished with SCD-directed therapies and/or CKD-directed therapies. In the past 5 years, novel disease-modifying and palliative therapies have been approved in SCD to target aspects of the disease, such as anemia, inflammation, and vasculopathy. Along with conventional hydroxyurea and chronic transfusion, l-glutamine, crizanlizumab, and voxelotor have all been shown to mitigate some adverse effect of SCD, and their effect on nephropathy is being investigated. CKD-directed therapies such as renin–angiotensin–aldosterone system blockers have long been used in SCD nephropathy; however, more complete long-term studies on benefits are needed. Given the effect of renal disease on survival, further assessment of the mechanisms and efficacy of these SCD-directed or CKD-directed therapeutic agents is essential.

Keywords: CKD, vascular

Introduction

Sickle cell disease (SCD) is a highly prevalent inherited blood disorder worldwide occurring in 300,000 births annually.1 It comprises sickle cell anemia (SCA) and variant SCD (compound heterozygous Hemoglobin SC or HbSb+ thalassemia), disorders with genotypic and phenotypic variability arising from a mutation in the hemoglobin B globin gene that produces sickle hemoglobin (HbS). Deoxygenated HbS (deoxy-HbS) polymerizes, resulting in fragile red blood cells (RBCs), chronic hemolysis with anemia and inflammation, vasculopathy, intermittent vaso-occlusive episodes (VOEs), and ultimately chronic multiorgan damage.2 In the more clinically severe SCA (homozygous HbSS or HbSb0 thalassemia), a multiorgan vasculopathy predominates and carries an increased risk of premature death.3 Advances in pediatric care in the United States48 has shifted the burden of disease to this new population of young adults with SCD who face a shorter lifespan compared with the general population.9,10 In this population, kidney disease is prevalent and can progress to CKD which affects mortality. The goal of this review is to discuss advances in SCD-focused therapies and their efficacy in reversing or slowing progression of CKD. In addition, we will describe the role of specific CKD-directed therapies in sickle nephropathy.

Renal Manifestation in SCD

The kidneys are sensitive to chronic hemolysis, anemia, and vaso-occlusion that are hallmarks of SCD, resulting in regional dysfunction1113 in the kidney (Figure 1).

Figure 1.

Figure 1

Clinical manifestation of SCA by region of the kidneys. SCA, sickle cell anemia.

The renal cortex has a relatively low rate of oxygen extraction but receives a high rate of blood flow, which facilitates high oxygen consumption,14,15 making this region sensitive to hypoxia and a likely site for deoxy-HbS polymerization. The renal medullary environment is hypoxic, acidotic, and hyperosmotic, contributing to HbS polymerization and recurrent vaso-occlusion, resulting in ischemia there and in the adjacent renal papillae.13,16

In the glomerulus, there is evidence for endothelial injury in the arterioles and glomerular hyperfiltration, believed to be stimulated by anemia, vasodilation, and medullary ischemia.1619 Free hemoglobin from intravascular hemolysis is directly toxic to the renal tubules through oxidative damage, inflammation, and depletion of nitric oxide (NO) with resultant vasculopathy.2022 In addition, free hemoglobin in the proximal tubules likely reduces albumin reabsorption contributing to proteinuria.23 Studies show that iron deposition in the tubular epithelium2426 in SCA are consistent with hemolysis, not chronic transfusion, as the proximate cause of this insult. Cellular damage to the proximal and distal convoluted tubules has been described, and there is a clinical association between hemolysis and albuminuria in this population.27,28

Sickle cell nephropathy can therefore appear as glomerular hyperfiltration, albuminuria, FSGS, impaired urinary concentration ability (hyposthenuria), metabolic acidosis, hematuria, papillary necrosis, and/or CKD (defined as depressed eGFR and/or proteinuria).16,29 Albuminuria, generally accepted as an early indicator of glomerular injury, can be seen in up to 30% of children and 60% of adults with HbSS, and associates with increasing age.30,31 In a longitudinal, prospective multicenter study with children and adults, albuminuria at >100 mg/g creatinine was associated with a significantly increased risk of persistent albuminuria and a faster rate of decline in eGFR.32 Persistence of albuminuria predicts the risk of progression of renal disease; thus, early detection of albuminuria and initiation of therapy to halt progression is the general approach to managing sickle cell nephropathy. Genetic modifiers that decrease risk of progression to CKD include higher fetal hemoglobin levels and coinherited α-thalassemia deletions which have net effect of decreasing the sickling effect of HbS. Genetic modifiers that increase the risk of progression to CKD include variants in MYH9, APOL1, and HMOX1.3335 Of note, albuminuria and CKD are common in variant SCD, with or without concomitant diabetes, hypertension, or other glomerulopathy.31 Before determining that a patient has SCD-related kidney disease, clinicians should exclude other potential causes of non–sickle cell–related kidney disease in people with SCD.

CKD is a significant predictor of risk of death in patients with SCD3639 and was present in 12% of an observational cohort (N=1056) of people living with SCA, with a mean age of onset of 37 years. In this cohort, most deaths (73.3%) occurred in people with prior organ damage, and renal failure was present in nearly one-third (29.4%) of those patients; CKD predicted death with an adjusted odds ratio (OR) of 2.04 (95% confidence interval [CI], 1.27 to 3.26).40

SCD-Directed Therapeutics in Nephropathy

Hydroxyurea

Hydroxyurea, the cornerstone of disease-modifying therapy in SCA, was approved by the US Food and Drug Administration (FDA) in 1998, initially for the indication of reducing frequency of painful VOEs in SCA.41 Hydroxyurea is a ribonucleotide reductase inhibitor that exerts its effect through multiple mechanisms, including increasing expression of fetal hemoglobin, reducing white blood cell and platelet counts, and increasing NO levels.42 Cumulatively, hydroxyurea decreases RBC sickling, thereby likely reducing downstream endothelial injury, intravascular hemolysis, and depletion of endogenous NO. Hydroxyurea use has been associated with reduced frequency of acute chest syndrome, reduced need for blood transfusions, likely improved mortality,43,44 decreased prevalence, and/or improvement of albuminuria.4548 A prospective study exploring the efficacy of hydroxyurea and angiotensin-converting enzyme inhibitors (ACEi) in 191 children with SCA, after a mean follow-up of 2.19 years, showed normalization of microalbuminuria in 44% of patients on hydroxyurea. In that study, 16 of 17 hydroxyurea-treated patients without microalbuminuria at the start of the study remained albuminuria-free throughout.49 Another cross-sectional study of albuminuria in 149 adults with SCD found a lower prevalence of albuminuria among patients on hydroxyurea; multivariate analyses showed hydroxyurea was associated with a lower likelihood of albuminuria (OR, 0.28; 95% CI, 0.11 to 0.75; P = 0.01).50 However, smaller shorter observational studies have not always noted significant benefit.51,52

Studies assessing hydroxyurea's effect on eGFR have showed conflicting results.53 The Pediatric Hydroxyurea Phase III Clinical Trial,54 a multicenter, phase III placebo-controlled study of 193 children with SCA, did not identify significant differences in eGFR between the two groups. However, they did note improve urine concentrating capacity in hydroxyurea-treated children, perhaps because of preserved tubular function. These results are limited by the young cohort (mean age 13.8 months) and short follow-up period.

The timing and dose of hydroxyurea initiation may affect albuminuria outcomes. In a retrospective analysis of data from 88 children with SCA,48 children who started hydroxyurea before age 10 years were less likely to develop albuminuria, compared with those who started hydroxyurea after 10 years (hazard ratio, 0.49; 95% CI, 0.25 to 0.97; P = 0.038); children on hydroxyurea in whom baseline albuminuria resolved received a higher dose of hydroxyurea (26.7±3.8 mg/kg per day versus 21.1±3.9 mg/kg per day; P = 0.003). Larger controlled studies including adult patients are needed.

Transfusion

Chronic RBC transfusions (simple and exchange) can increase hemoglobin levels and decrease the amount of sickle RBCs in circulation, which has been shown to reduce markers of endothelial injury.55 Older studies have demonstrated improved urinary concentration ability with simple transfusions; however, the capacity for recovery might be reduced with age.56,57 A retrospective review of 120 children and young adults with SCD found that patients who started chronic transfusion therapy at a younger age had a lower prevalence of microalbuminuria.52 They concluded that chronic transfusions were renoprotective when started younger than 9 years, while acknowledging significant transfusion-related adverse effects limiting routine use. However, another retrospective study evaluating risk of CKD progression in children did not note significant differences in the prevalence of microalbuminuria among treatment groups of hydroxyurea, chronic transfusions, or observation.51

l-Glutamine

l-glutamine was approved by the FDA in 2017, with the indication of reducing frequency of VOEs.58 l-glutamine is an essential amino acid, and its mechanism of action in SCD is unknown. l-glutamine is essential for the synthesis of antioxidants NAD and GSH. As part of the oxidative stress that leads to hemolysis of HbS-containing RBCs, there is increased production of reactive oxidant species and free radicals that consume reduced NAD (reduced nicotinamide adenine dinucleotide) and GSH.5961 The depletion of reduced nicotinamide adenine dinucleotide and GSH is mitigated by glutamine, which is believed to decrease RBC damage and the threshold for hemolysis. Although one study found significantly decreased markers of hemolysis (reticulocytes and lactate dehydrogenase) in 19 patients on l-glutamine,62 the phase III approval study did not reproduce this finding.58 Previous studies support the role of glutamine in improving NAD redox potential and decreasing endothelial adhesion.61 Observational studies have focused on l-glutamine's effect on pain63 and other complications, and its effect on progression of renal disease in SCD is unknown.

Other reviews have discussed alternative mechanisms by which glutamine could affect erythrocytes.64 These include the suppression of inflammatory pathways and modulation of intestinal microbiome and barrier function to create a less aggressive SCD phenotype60 and glutamine acting to reduce resting energy expenditure.65 As with all supplements, caution should be used in patients with underlying liver disease or renal disease. Post hoc analysis of a randomized clinical trial assessing the use of l-glutamine in critically ill patients identified the greatest potential for harm in those with evidence for renal dysfunction at enrollment into the study.66

Crizanlizumab

The monoclonal antibody crizanlizumab was approved by the FDA in 2019, with the indication of reducing VOE frequency.67 Crizanlizumab inhibits P-selectin binding to its ligands, presumably blocking P-selectin–mediated adhesion of inflammatory cells to the endothelium, thus reducing vaso-occlusion. Given its recent approval, there are ongoing evaluations68 into its clinical benefits, including its effect on nephropathy. The Study Exploring the Effect of Crizanlizumab on Kidney Function in Patients With Chronic Kidney Disease Caused by Sickle Cell Disease trial (NCT04053764) is a phase II study comparing crizanlizumab and standard of care with standard of care alone in patients with SCD-associated CKD. It is based on previous studies showing increased P-selectin expression in kidney ischemia-reperfusion injury.69 Its primary end point is the proportion of patients who experience ≥30% decrease in albuminuria at 12 months. In addition, the Study of Two Doses of Crizanlizumab Versus Placebo in Adolescent and Adult Sickle Cell Disease Patients trial (NCT03814746) is a phase III placebo-controlled trial comparing efficacy and safety of crizanlizumab doses with a secondary outcome of albuminuria prevalence.

Voxelotor

Voxelotor was approved by the FDA in 2019, with the indication of increasing hemoglobin concentration.70 Voxelotor selectively and reversibly binds to hemoglobin, keeping it longer in the oxy-Hb or R-state. This results in delayed and decreased HbS polymerization, with less hemolysis and a modest increase in Hb concentration. The results from the long-term follow-up of the initial Hemoglobin Oxygen Affinity Modulation to Inhibit HbS Polymerization study71 confirmed a durable increase in hemoglobin and noted significant improvement in markers of hemolysis (lactate dehydrogenase, indirect bilirubin, and reticulocytes) in the group randomized to 1500 mg of voxelotor. Given the noted effect on hemolysis markers,72,73 a small retrospective study evaluated albuminuria before and after voxelotor initiation in ten adult patients with SCA.74 They reported a 25% reduction in albuminuria from baseline in all patients placed on therapy, with a median decrease by 109 (−38 to −379) mg/g, compared with age-matched and sex-matched untreated controls that showed a 34% increase in albuminuria.

The initial voxelotor trial70 noted that the change in erythropoietin levels were similar across the groups, with a trend toward a lower erythropoietin level in the 1500 mg voxelotor group through week 24. This finding suggests that higher voxelotor hemoglobin occupancy, and higher hemoglobin, blunts the hypoxia-mediated induction of erythropoietin. An open-label study75 assessing voxelotor in participants with hepatic and renal impairment suggests no effect of kidney function on its excretion (comparable half-life values) but recommended dose reduction in severe hepatic impairment.

Like crizanlizumab, there are several ongoing trials further evaluating safety and efficacy including a randomized trial comparing risk of CKD progression in patients receiving voxelotor compared with standard of care (NCT04335721). In addition, a next generation Hb-modifying agent, GBT021601, is being studied in a phase II/III clinical trial (NCT05431088) in adults and children.

CKD-Directed Therapeutics in SCD

ACEi/Angiotensin Receptor Blockers

Angiotensin inhibition with ACEi and angiotensin receptor blockers, cumulatively known as renin–angiotensin–aldosterone system (RAAS) blockers, used in other albuminuric/proteinuric kidney diseases and CKD,7678 have long been used in the treatment of sickle nephropathy; however, hyperkalemia has limited widespread adoption in real-world settings. Falk et al.,13 in a prospective study investigating enalapril in sickle nephropathy, found a significant decline in urinary protein excretion in ten adults after 2 weeks on enalapril. They also noted that after discontinuation of therapy, mean protein excretion remained lower than baseline. These findings were corroborated in a placebo-controlled randomized control trial of 22 adults79 where at 6 months of therapy with captopril, there was significant difference in the absolute (−63 mg/24 h [95% CI, 40 to 86]) and percentage (−54% [95% CI, 22% to 85%]) change in albuminuria with treatment.

More recently, angiotensin receptor blockers have been evaluated in a phase I study in which patients with SCA and persistent albuminuria (N=12) were placed on losartan for 1 year with renal function assessments at short (1–2 months) and long (≥12 months) time points.80 The albumin excretion rate decreased significantly in short-term but not at ≥12 months, although only eight patients completed long-term therapy. eGFR did not change significantly; thus, the use of losartan was believed to decrease albumin excretion with stable GFR. Another phase II multicenter trial with 6 months of losartan in adults and children with SCA met their primary end point of ≥25% reduction in albuminuria in the group of patients with microalbuminuria and macroalbuminuria.81

A large single-center retrospective review followed 86 patients on RAAS blockers for a median of 2.28 years compared with 68 patients not on therapy followed for 2.24 years.82 While not statistically significant, the odds of improved proteinuria over time were higher in the group on RAAS blockers (OR, 1.36; P = 0.063). There was a statistically significant difference in the rate of eGFR decline between the groups with slower rate in the group on therapy.

More long-term assessments of the efficacy and tolerance of RAAS blockade in this setting are needed, as well as prospective testing for synergism with SCD-related therapies.

Sodium–Glucose Cotransporter-2 Inhibitors and Mineralocorticoid Receptor Antagonists

Sodium–glucose cotransporter-2 inhibitors, used as antiglycemic agents now, have indication for use in CKD and albuminuric kidney diseases. Dapagliflozin and empagliflozin have demonstrated efficacy in slowing down CKD progression, independent of diabetes diagnosis,83,84 and canagliflozin has shown similar efficacy in albuminuric nephropathies.85

Finerenone, a nonsteroidal, selective mineralocorticoid receptor antagonist, was approved for diabetic kidney disease based on the Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease study.86 The placebo-controlled randomized study of participants with CKD receiving maximum tolerated doses of RAAS blockers showed significantly decreased risk of CKD progression in the finerenone group compared with placebo group but also higher rates of hyperkalemia. Because of its proposed ability to abrogate inflammation and fibrosis, both relevant in SCD nephropathy, some have suggested finerenone may have particular benefit for this disease entity.87

The use of these therapies in SCD nephropathy has not yet been investigated; therefore, the benefits in this setting are unclear.

Emerging Therapies

Endothelin Receptor Antagonists

Endothelin (ET)-1 is a peptide that functions through binding to its receptors, ETA and ETB, of which ETA has proinflammatory, vasoconstrictive, and nociceptive actions. In SCD-associated oxidative stress, hypoxia, and vascular injury, the cellular synthesis of ET-1 is increased.88 ET-1 signaling contributes to development of chronic pain, vasculopathy, pulmonary hypertension, and CKD in SCD.88 Investigation of ET-1's role in nephropathy has shown high urinary ET-1 levels in patients with SCD and positive correlation between urinary albumin-creatinine ratio and serum ET-1 levels.89 In mouse models, ambrisentan, a selective ETA antagonist, has demonstrated efficacy in preventing nephropathy from the level of the glomerulus to the tubules.90 It is now being evaluated in a phase I trial (NCT02712346) with a secondary outcome measure of urinary microalbuminuria/proteinuria. Atrasentan, another selective ETA antagonist, which has been studied in diabetic nephropathy and CKD,91 is being evaluated (NCT04573920) for efficacy and safety in proteinuric glomerular diseases. Sparsentan, a dual angiotensin receptor and ETA antagonist, has been shown to decrease proteinuria in FSGS92 and is undergoing additional evaluation of its long-term efficacy in FSGS (NCT03493685) and in IgA nephropathy (NCT03762850). On the basis of prespecified interim analysis of the latter study which demonstrated greater reduction in proteinuria at 36 weeks when compared with irbesartan, sparsentan recently received accelerated approval for IgA nephropathy.93

Pyruvate Kinase Activators

Pyruvate kinase (PK) catalyzes the last step in glycolysis, resulting in generation of ATP. PK has a role in energy homeostasis, membrane integrity, and maintenance of 2,3-diphosphoglycerate (2,3-DPG), an intermediate of glycolysis. Elevated 2,3-DPG levels decrease oxygen affinity and pH in erythrocytes,94,95 which promotes deoxy-HbS polymerization and RBC sickling. PK activators increase ATP and reduce 2,3-DPG, thus reducing HbS polymerization, supporting RBC health and membrane integrity.

Mitapivat (AG-348) has demonstrated the ability to stabilize PK in sickle erythrocytes. A current phase II open-label study assessing efficacy and safety, reported on nine participants followed over 8 weeks.96 They saw improvements in anti-HbS polymerization, noted by a reduction in the point of sickling (a novel biomarker demonstrating when sickling is initiated in vitro), an increase in measured Hb-oxygen affinity, ATP/2,3-DPG ratios, and total hemoglobin, with decreases in 2,3-DPG levels and hemolysis markers. The investigators are continuing the study over an extension period as a phase II/III study (NCT05031780). Etavopivat (FT-4202) is another PK activator that is being evaluated in SCD through the Study of Etavopivat in Adults and Adolescents With Sickle Cell Disease (NCT04624659).

While these studies are not focused on nephropathy, data for GFR and albuminuria will be captured for both agents. The upstream targeting of HbS polymerization, as for voxelotor, might be predicted to result in improvements in hemolysis and anemia, with subsequent mitigation of kidney damage.

Special Considerations in SCD Population

Iron Chelators

Iron overload is an expected complication of chronic or recurrent transfusion therapy, indicated in many SCD-specific settings. Thus, iron chelation becomes necessary to prevent morbidity. Available chelating agents are deferoxamine, deferasirox, and deferiprone, and the choice of initiation depends on multiple factors. Deferasirox, a widely used oral chelator, has been implicated in kidney-specific adverse events ranging from dose-dependent but reversible elevations in creatinine to renal failure97,98; therefore, deferasirox should be used cautiously and with frequent monitoring of renal function.

Nonsteroidal Anti-Inflammatory Drugs

Nonsteroidal anti-inflammatory drugs (NSAIDs) play a role in SCD-mediated pain management but should be used sparingly in the setting of sickle nephropathy. In the presence of nephropathy, the vasoconstrictive mechanism of NSAIDs can worsen glomerular hyperfiltration99 and contribute to renal insufficiency. In addition, recurrent use of NSAIDs carries the risk for acute kidney injury, which itself is a factor for developing CKD.100

Nephropathy, up to and including ESKD, in SCD is an important predictor of mortality. The pathophysiology of SCD-associated nephropathy is multifactorial; however, the proximate cause is hemoglobin polymerization and its downstream consequences.

Newer SCD-directed therapies target aspects of the disease implicated in renal damage, including rate of RBC polymerization, anemia, hemolysis, endothelial adhesion, and vascular inflammation (Figure 2). The use of RAAS blockers, sodium–glucose cotransporter-2 inhibitors, and avoidance of nephrotoxins are universal approaches to manage CKD that are being extended to SCD-related kidney disease.

Figure 2.

Figure 2

Schematic demonstrating the major mechanisms of SCA on vasculopathy and point of effect of targeted therapeutics. Cr, crizanlizumab; HU, hydroxyurea; L-g, l-glutamine; SCA, sickle cell anemia; Vx, voxelotor.

The literature contains mostly small, observational studies, so there is need for larger, longer-term, prospective, controlled, and real-world studies to better evaluate the effect of interventions on albuminuria, eGFR, tubular dysfunction, and overall renal health. Future directions for research include elucidating mechanisms of action of these therapeutics, clarifying the role of biomarkers in diagnosing renal damage and its response to therapy, and testing for potential synergism between therapeutic strategies.

Disclosures

V.K. Derebail reports the following—consultancy: Bayer, Forma Therapeutics, Merck, Novartis, and Travere; honoraria: UpToDate; and research funding: Site PI for Clinical Trials (Boehringer Ingelheim, Chemocentryx, Gilead, Hansa, Infla-RX, Merck, Travere, and Vertex). J. Little reports the following—advisory or leadership role: Husband Tom Hostetter advisor for Tricida is not being paid for this currently or in recent past; patents or royalties: On patents currently being used at BioChip Labs and Hemex, but I receive no royalties or financial rewards from this; research funding: BlueBird Bio and GBT (Pfizer); and other interests or relationships: Member of National Association of Sickle Cell Centers and receive funding; receive funding from NHLBI; on study adjudication for Forma/Norva Nordisk Ph III Hibiscus Study. All remaining authors have nothing to disclose.

Funding

M. Obadina: National Heart, Lung, and Blood Institute (T32HL007149-47). V.K. Derebail: National Heart, Lung, and Blood Institute (5R01HL159376) and US Food and Drug Administration (5R01FD006030).

Author Contributions

Conceptualization: Jane Little, Mofiyin Obadina.

Visualization: Mofiyin Obadina.

Writing – original draft: Mofiyin Obadina.

Writing – review & editing: Vimal K. Derebail, Jane Little, Mofiyin Obadina, Sam Wilson.

References

  • 1.Piel FB, Hay SI, Gupta S, Weatherall DJ, Williams TN. Global burden of sickle cell anaemia in children under five, 2010–2050: modelling based on demographics, excess mortality, and interventions. PLoS Med. 2013;10(7):e1001484. doi: 10.1371/journal.pmed.1001484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kato GJ Piel FB Reid CD, et al. Sickle cell disease. Nat Rev Dis Primers. 2018;4(1):18010. doi: 10.1038/nrdp.2018.10 [DOI] [PubMed] [Google Scholar]
  • 3.Platt OS Brambilla DJ Rosse WF, et al. Mortality in sickle cell disease: life expectancy and risk factors for early death. N Engl J Med. 1994;330(23):1639–1644. doi: 10.1056/NEJM199406093302303 [DOI] [PubMed] [Google Scholar]
  • 4.Gaston MH Verter JI Woods G, et al. Prophylaxis with oral penicillin in children with sickle cell anemia: a randomized trial. N Engl J Med. 1986;314(25):1593–1599. doi: 10.1056/NEJM198606193142501 [DOI] [PubMed] [Google Scholar]
  • 5.Vichinsky E, Hurst D, Earles A, Kleman K, Lubin B. Newborn screening for sickle cell disease: effect on mortality. Pediatrics. 1988;81(6):749–755. doi: 10.1542/peds.81.6.749 [DOI] [PubMed] [Google Scholar]
  • 6.Adams RJ McKie VC Hsu L, et al. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography. N Engl J Med. 1998;339(1):5–11. doi: 10.1056/NEJM199807023390102 [DOI] [PubMed] [Google Scholar]
  • 7.Quinn CT, Rogers ZR, Buchanan GR. Survival of children with sickle cell disease. Blood. 2004;103(11):4023–4027. doi: 10.1182/blood-2003-11-3758 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bernaudin F Verlhac S Arnaud C, et al. Impact of early transcranial Doppler screening and intensive therapy on cerebral vasculopathy outcome in a newborn sickle cell anemia cohort. Blood. 2011;117(4):1130–1140. doi: 10.1182/blood-2010-06-293514 [DOI] [PubMed] [Google Scholar]
  • 9.Quinn CT, Rogers ZR, McCavit TL, Buchanan GR. Improved survival of children and adolescents with sickle cell disease. Blood. 2010;115(17):3447–3452. doi: 10.1182/blood-2009-07-233700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.DeBaun MR Ghafuri DL Rodeghier M, et al. Decreased median survival of adults with sickle cell disease after adjusting for left truncation bias: a pooled analysis. Blood. 2019;133(6):615–617. doi: 10.1182/blood-2018-10-880575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Van Eps L, Pinedo-Veels C, de Vries GH, de Koning J. Nature of concentrating defect in sickle-cell nephropathy. Microradioangiographic studies. Lancet. 1970;295(7644):450–452. doi: 10.1016/s0140-6736(70)90836-6 [DOI] [PubMed] [Google Scholar]
  • 12.Powars DR Elliott-Mills DD Chan L, et al. Chronic renal failure in sickle cell disease: risk factors, clinical course, and mortality. Ann Intern Med. 1991;115(8):614–620. doi: 10.7326/0003-4819-115-8-614 [DOI] [PubMed] [Google Scholar]
  • 13.Falk RJ, Scheinman J, Phillips G, Orringer E, Johnson A, Jennette JC. Prevalence and pathologic features of sickle cell nephropathy and response to inhibition of angiotensin-converting enzyme. N Engl J Med. 1992;326(14):910–915. doi: 10.1056/NEJM199204023261402 [DOI] [PubMed] [Google Scholar]
  • 14.O'Connor PM. Renal oxygen delivery: matching delivery to metabolic demand. Clin Exp Pharmacol Physiol. 2006;33(10):961–967. doi: 10.1111/j.1440-1681.2006.04475.x [DOI] [PubMed] [Google Scholar]
  • 15.Layton AT. Recent advances in renal hypoxia: insights from bench experiments and computer simulations. Am J Physiol Renal Physiol. 2016;311(1):F162–F165. doi: 10.1152/ajprenal.00228.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Saraf SL, Derebail VK, Gordeuk VR, Little JA. Sickle nephropathy. In: Gladwin MT, Kato GJ, Novelli EM, editors. Sickle Cell Disease. McGraw Hill; 2021:303–325. [Google Scholar]
  • 17.Tharaux PL Hagège I Placier S, et al. Urinary endothelin-1 as a marker of renal damage in sickle cell disease. Nephrol Dial Transplant. 2005;20(11):2408–2413. doi: 10.1093/ndt/gfi111 [DOI] [PubMed] [Google Scholar]
  • 18.Ataga KI Brittain JE Moore D, et al. Urinary albumin excretion is associated with pulmonary hypertension in sickle cell disease: potential role of soluble FMS-like tyrosine kinase-1. Eur J Haematol. 2010;85(3):257–263. doi: 10.1111/j.1600-0609.2010.01471.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ataga KI, Gordeuk VR, Agodoa I, Colby JA, Gittings K, Allen IE. Low hemoglobin increases risk for cerebrovascular disease, kidney disease, pulmonary vasculopathy, and mortality in sickle cell disease: a systematic literature review and meta-analysis. PLoS One. 2020;15(4):e0229959. doi: 10.1371/journal.pone.0229959 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kato GJ McGowan V Machado RF, et al. Lactate dehydrogenase as a biomarker of hemolysis-associated nitric oxide resistance, priapism, leg ulceration, pulmonary hypertension, and death in patients with sickle cell disease. Blood. 2006;107(6):2279–2285. doi: 10.1182/blood-2005-06-2373 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tracz MJ, Alam J, Nath KA. Physiology and pathophysiology of heme: implications for kidney disease. J Am Soc Nephrol. 2007;18(2):414–420. doi: 10.1681/ASN.2006080894 [DOI] [PubMed] [Google Scholar]
  • 22.Gladwin MT, Kanias T, Kim-Shapiro DB. Hemolysis and cell-free hemoglobin drive an intrinsic mechanism for human disease. J Clin Invest. 2012;122(4):1205–1208. doi: 10.1172/JCI62972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Eshbach ML, Kaur A, Rbaibi Y, Tejero J, Weisz OA. Hemoglobin inhibits albumin uptake by proximal tubule cells: implications for sickle cell disease. Am J Physiol Cell Physiol. 2017;312(6):C733–C740. doi: 10.1152/ajpcell.00021.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lusco MA, Fogo AB, Najafian B, Alpers CE. AJKD atlas of renal pathology: sickle cell nephropathy. Am J Kidney Dis. 2016;68(1):e1–e3. doi: 10.1053/j.ajkd.2016.05.003 [DOI] [PubMed] [Google Scholar]
  • 25.Vasavda N, Gutiérrez L, House MJ, Drašar E, St Pierre TG, Thein SL. Renal iron load in sickle cell disease is influenced by severity of haemolysis. Br J Haematol. 2012;157(5):599–605. doi: 10.1111/j.1365-2141.2012.09093.x [DOI] [PubMed] [Google Scholar]
  • 26.Donnola SB Piccone CM Lu L, et al. Diffusion tensor imaging MRI of sickle cell kidney disease: initial results and comparison with iron deposition. NMR Biomed. 2018;31(3):e3883. doi: 10.1002/nbm.3883 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hamideh D Raj V Harrington T, et al. Albuminuria correlates with hemolysis and NAG and KIM-1 in patients with sickle cell anemia. Pediatr Nephrol. 2014;29(10):1997–2003. doi: 10.1007/s00467-014-2821-8 [DOI] [PubMed] [Google Scholar]
  • 28.Day TG, Drasar ER, Fulford T, Sharpe CC, Thein SL. Association between hemolysis and albuminuria in adults with sickle cell anemia. Haematologica. 2012;97(2):201–205. doi: 10.3324/haematol.2011.050336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sharpe CC, Thein SL. Sickle cell nephropathy – a practical approach. Br J Haematol. 2011;155(3):287–297. doi: 10.1111/j.1365-2141.2011.08853.x [DOI] [PubMed] [Google Scholar]
  • 30.Ataga KI, Derebail VK, Archer DR. The glomerulopathy of sickle cell disease. Am J Hematol. 2014;89(9):907–914. doi: 10.1002/ajh.23762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Drawz P Ayyappan S Nouraie M, et al. Kidney disease among patients with sickle cell disease, hemoglobin SS and SC. Clin J Am Soc Nephrol. 2016;11(2):207–215. doi: 10.2215/CJN.03940415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Niss O Lane A Asnani MR, et al. Progression of albuminuria in patients with sickle cell anemia: a multicenter, longitudinal study. Blood Adv. 2020;4(7):1501–1511. doi: 10.1182/bloodadvances.2019001378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ashley-Koch AE Okocha EC Garrett ME, et al. MYH9 and APOL1 are both associated with sickle cell disease nephropathy. Br J Haematol. 2011;155(3):386–394. doi: 10.1111/j.1365-2141.2011.08832.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zahr RS Rampersaud E Kang G, et al. Children with sickle cell anemia and APOL1 genetic variants develop albuminuria early in life. Haematologica. 2019;104(9):e385–e387. doi: 10.3324/haematol.2018.212779 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ataga KI, Saraf SL, Derebail VK. The nephropathy of sickle cell trait and sickle cell disease. Nat Rev Nephrol. 2022;18(6):361–377. doi: 10.1038/s41581-022-00540-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.McClellan AC Luthi JC Lynch JR, et al. High one year mortality in adults with sickle cell disease and end-stage renal disease. Br J Haematol. 2012;159(3):360–367. doi: 10.1111/bjh.12024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Elmariah H Garrett ME De Castro LM, et al. Factors associated with survival in a contemporary adult sickle cell disease cohort. Am J Hematol. 2014;89(5):530–535. doi: 10.1002/ajh.23683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gladwin MT Barst RJ Gibbs JS, et al. Risk factors for death in 632 patients with sickle cell disease in the United States and United Kingdom. PLoS One. 2014;9(7):e99489. doi: 10.1371/journal.pone.0099489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Maitra P Caughey M Robinson L, et al. Risk factors for mortality in adult patients with sickle cell disease: a meta-analysis of studies in North America and Europe. Haematologica. 2017;102(4):626–636. doi: 10.3324/haematol.2016.153791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Powars DR, Chan LS, Hiti A, Ramicone E, Johnson C. Outcome of sickle cell anemia: a 4-decade observational study of 1056 patients. Medicine (Baltimore). 2005;84(6):363–376. doi: 10.1097/01.md.0000189089.45003.52 [DOI] [PubMed] [Google Scholar]
  • 41.Charache S Terrin ML Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. N Engl J Med. 1995;332(20):1317–1322. doi: 10.1056/NEJM199505183322001 [DOI] [PubMed] [Google Scholar]
  • 42.McGann PT, Ware RE. Hydroxyurea therapy for sickle cell anemia. Expert Opin Drug Saf. 2015;14(11):1749–1758. doi: 10.1517/14740338.2015.1088827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Voskaridou E Christoulas D Bilalis A, et al. The effect of prolonged administration of hydroxyurea on morbidity and mortality in adult patients with sickle cell syndromes: results of a 17-year, single-center trial (LaSHS). Blood. 2010;115(12):2354–2363. doi: 10.1182/blood-2009-05-221333 [DOI] [PubMed] [Google Scholar]
  • 44.Steinberg MH McCarthy WF Castro O, et al. The risks and benefits of long-term use of hydroxyurea in sickle cell anemia: a 17.5-year follow-up. Am J Hematol. 2010;85(6):403–408. doi: 10.1002/ajh.21699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Becton LJ Kalpatthi RV Rackoff E, et al. Prevalence and clinical correlates of microalbuminuria in children with sickle cell disease. Pediatr Nephrol. 2010;25(8):1505–1511. doi: 10.1007/s00467-010-1536-8 [DOI] [PubMed] [Google Scholar]
  • 46.Bartolucci P Habibi A Stehlé T, et al. Six months of hydroxyurea reduces albuminuria in patients with sickle cell disease. J Am Soc Nephrol. 2016;27(6):1847–1853. doi: 10.1681/ASN.2014111126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Tehseen S, Joiner CH, Lane PA, Yee ME. Changes in urine albumin to creatinine ratio with the initiation of hydroxyurea therapy among children and adolescents with sickle cell disease. Pediatr Blood Cancer. 2017;64(12):e26665. doi: 10.1002/pbc.26665 [DOI] [PubMed] [Google Scholar]
  • 48.Zahr RS Hankins JS Kang G, et al. Hydroxyurea prevents onset and progression of albuminuria in children with sickle cell anemia. Am J Hematol. 2019;94(1):E27–E29. doi: 10.1002/ajh.25329 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.McKie KT, Hanevold CD, Hernandez C, Waller JL, Ortiz L, McKie KM. Prevalence, prevention, and treatment of microalbuminuria and proteinuria in children with sickle cell disease. J Pediatr Hematol Oncol. 2007;29(3):140–144. doi: 10.1097/MPH.0b013e3180335081 [DOI] [PubMed] [Google Scholar]
  • 50.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):1211–1218. doi: 10.1093/ndt/gft295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lebensburger J, Johnson SM, Askenazi DJ, Rozario NL, Howard TH, Hilliard LM. Protective role of hemoglobin and fetal hemoglobin in early kidney disease for children with sickle cell anemia. Am J Hematol. 2011;86(5):430–432. doi: 10.1002/ajh.21994 [DOI] [PubMed] [Google Scholar]
  • 52.Alvarez O, Montane B, Lopez G, Wilkinson J, Miller T. Early blood transfusions protect against microalbuminuria in children with sickle cell disease. Pediatr Blood Cancer. 2006;47(1):71–76. doi: 10.1002/pbc.20645 [DOI] [PubMed] [Google Scholar]
  • 53.Aygun B, Mortier NA, Smeltzer MP, Shulkin BL, Hankins JS, Ware RE. Hydroxyurea treatment decreases glomerular hyperfiltration in children with sickle cell anemia. Am J Hematol. 2013;88(2):116–119. doi: 10.1002/ajh.23365 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Alvarez O Miller ST Wang WC, 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):668–674. doi: 10.1002/pbc.24100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hyacinth HI, Adams RJ, Voeks JH, Hibbert JM, Gee BE. Frequent red cell transfusions reduced vascular endothelial activation and thrombogenicity in children with sickle cell anemia and high stroke risk. Am J Hematol. 2014;89(1):47–51. doi: 10.1002/ajh.23586 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Keitel HG, Thompson D, Itano HA. Hyposthenuria in sickle cell anemia: a reversible renal defect. J Clin Invest. 1956;35(9):998–1007. doi: 10.1172/JCI103360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.van Eps L, Schouten H, La Porte-Wijsman LW, Struyker Boudier AM. The influence of red blood cell transfusions on the hyposthenuria and renal hemodynamics of sickle cell anemia. Clin Chim Acta. 1967;17(3):449–461. doi: 10.1016/0009-8981(67)90222-7 [DOI] [PubMed] [Google Scholar]
  • 58.Niihara Y Miller ST Kanter J, et al. A phase 3 trial of l-glutamine in sickle cell disease. N Engl J Med. 2018;379(3):226–235. doi: 10.1056/NEJMoa1715971 [DOI] [PubMed] [Google Scholar]
  • 59.Morris CR Suh JH Hagar W, et al. Erythrocyte glutamine depletion, altered redox environment, and pulmonary hypertension in sickle cell disease. Blood. 2008;111(1):402–410. doi: 10.1182/blood-2007-04-081703 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jafri F Seong G Jang T, et al. L-glutamine for sickle cell disease: more than reducing redox. Ann Hematol. 2022;101(8):1645–1654. doi: 10.1007/s00277-022-04867-y [DOI] [PubMed] [Google Scholar]
  • 61.Niihara Y Matsui NM Shen YM, et al. L-glutamine therapy reduces endothelial adhesion of sickle red blood cells to human umbilical vein endothelial cells. BMC Hematol. 2005;5(1):4. doi: 10.1186/1471-2326-5-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Elenga N, Loko G, Etienne-Julan M, Al-Okka R, Adel AM, Yassin MA. Real-World data on efficacy of L-glutamine in preventing sickle cell disease-related complications in pediatric and adult patients. Front Med (Lausanne). 2022;9:931925. doi: 10.3389/fmed.2022.931925 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wilson S, Wright F, Carden MA. L-glutamine decreases opioid use in individuals with sickle cell disease and chronic pain: a case series. Blood. 2019;134(suppl 1):4849. doi: 10.1182/blood-2019-126179 [DOI] [Google Scholar]
  • 64.Quinn CT. L-glutamine for sickle cell anemia: more questions than answers. Blood. 2018;132(7):689–693. doi: 10.1182/blood-2018-03-834440 [DOI] [PubMed] [Google Scholar]
  • 65.Williams R Olivi S Li CS, et al. Oral glutamine supplementation decreases resting energy expenditure in children and adolescents with sickle cell anemia. J Pediatr Hematol Oncol. 2004;26(10):619–625. doi: 10.1097/01.mph.0000140651.65591.b8 [DOI] [PubMed] [Google Scholar]
  • 66.Heyland DK Elke G Cook D, et al. Glutamine and antioxidants in the critically ill patient: a post hoc analysis of a large-scale randomized trial. JPEN J Parenter Enteral Nutr. 2015;39(4):401–409. doi: 10.1177/0148607114529994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ataga KI Kutlar A Kanter J, et al. Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med. 2017;376(5):429–439. doi: 10.1056/NEJMoa1611770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Liles DK Shah NR Scullin B, et al. Long-term biological effects in sickle cell disease: insights from a post-crizanlizumab study. Br J Haematol. 2021;195(4):e150–e153. doi: 10.1111/bjh.17739 [DOI] [PubMed] [Google Scholar]
  • 69.Singbartl K, Green SA, Ley K. Blocking P-selectin protects from ischemia/reperfusion-induced acute renal failure. FASEB J. 2000;14(1):48–54. doi: 10.1096/fasebj.14.1.48 [DOI] [PubMed] [Google Scholar]
  • 70.Vichinsky E Hoppe CC Ataga KI, et al. A phase 3 randomized trial of voxelotor in sickle cell disease. N Engl J Med. 2019;381(6):509–519. doi: 10.1056/NEJMoa1903212 [DOI] [PubMed] [Google Scholar]
  • 71.Howard J Ataga KI Brown RC, et al. Voxelotor in adolescents and adults with sickle cell disease (HOPE): long-term follow-up results of an international, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Haematol. 2021;8(5):e323–e333. doi: 10.1016/S2352-3026(21)00059-4 [DOI] [PubMed] [Google Scholar]
  • 72.Muschick K, Fuqua T, Stoker-Postier C, Anderson AR. Real-world data on voxelotor to treat patients with sickle cell disease. Eur J Haematol. 2022;109(2):154–161. doi: 10.1111/ejh.13782 [DOI] [PubMed] [Google Scholar]
  • 73.Estepp JH Kalpatthi R Woods G, et al. Safety and efficacy of voxelotor in pediatric patients with sickle cell disease aged 4 to 11 years. Pediatr Blood Cancer. 2022;69(8):e29716. doi: 10.1002/pbc.29716 [DOI] [PubMed] [Google Scholar]
  • 74.Han J, Molokie RE, Hussain F, Njoku F, Gordeuk VR, Saraf SL. Voxelotor and albuminuria in adults with sickle cell anaemia. Br J Haematol. 2022;197(5):e63–e64. doi: 10.1111/bjh.18076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Preston RA Marbury T Balaratnam G, et al. Pharmacokinetics of voxelotor in patients with renal and hepatic impairment. J Clin Pharmacol. 2021;61(4):493–505. doi: 10.1002/jcph.1757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lewis EJ, Hunsicker LG, Bain RP, Rohde RD. The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med. 1993;329(20):1456–1462. doi: 10.1056/NEJM199311113292004 [DOI] [PubMed] [Google Scholar]
  • 77.Casas JP Chua W Loukogeorgakis S, et al. Effect of inhibitors of the renin-angiotensin system and other antihypertensive drugs on renal outcomes: systematic review and meta-analysis. Lancet. 2005;366(9502):2026–2033. doi: 10.1016/S0140-6736(05)67814-2 [DOI] [PubMed] [Google Scholar]
  • 78.Kunz R, Friedrich C, Wolbers M, Mann JF. Meta-analysis: effect of monotherapy and combination therapy with inhibitors of the renin angiotensin system on proteinuria in renal disease. Ann Intern Med. 2008;148(1):30–48. doi: 10.7326/0003-4819-148-1-200801010-00190 [DOI] [PubMed] [Google Scholar]
  • 79.Foucan L, Bourhis V, Bangou J, Mérault L, Etienne-Julan M, Salmi RL. A randomized trial of captopril for microalbuminuria in normotensive adults with sickle cell anemia. Am J Med. 1998;104(4):339–342. doi: 10.1016/s0002-9343(98)00056-4 [DOI] [PubMed] [Google Scholar]
  • 80.Yee ME, Lane PA, Archer DR, Joiner CH, Eckman JR, Guasch A. Losartan therapy decreases albuminuria with stable glomerular filtration and permselectivity in sickle cell anemia. Blood Cells Mol Dis. 2018;69:65–70. doi: 10.1016/j.bcmd.2017.09.006 [DOI] [PubMed] [Google Scholar]
  • 81.Quinn CT Saraf SL Gordeuk VR, et al. Losartan for the nephropathy of sickle cell anemia: a phase-2, multicenter trial. Am J Hematol. 2017;92(9):E520–E528. doi: 10.1002/ajh.24810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Thrower A, Ciccone EJ, Maitra P, Derebail VK, Cai J, Ataga KI. Effect of renin-angiotensin-aldosterone system blocking agents on progression of glomerulopathy in sickle cell disease. Br J Haematol. 2019;184(2):246–252. doi: 10.1111/bjh.15651 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Heerspink HJL Stefánsson BV Correa-Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436–1446. doi: 10.1056/NEJMoa2024816 [DOI] [PubMed] [Google Scholar]
  • 84.Herrington WG Staplin N Wanner C, et al.; The EMPA-KIDNEY Collaborative Group. Empagliflozin in patients with chronic kidney disease. N Engl J Med. 2023;388(2):117–127. doi: 10.1056/NEJMoa2204233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Perkovic V Jardine MJ Neal B, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380(24):2295–2306. doi: 10.1056/NEJMoa1811744 [DOI] [PubMed] [Google Scholar]
  • 86.Bakris GL Agarwal R Anker SD, et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med. 2020;383(23):2219–2229. doi: 10.1056/NEJMoa2025845 [DOI] [PubMed] [Google Scholar]
  • 87.Epstein M. Aldosterone and mineralocorticoid receptor signaling as determinants of cardiovascular and renal injury: from hans selye to the present. Am J Nephrol. 2021;52(3):209–216. doi: 10.1159/000515622 [DOI] [PubMed] [Google Scholar]
  • 88.Fox BM, Kasztan M. Endothelin receptor antagonists in sickle cell disease: a promising new therapeutic approach. Life Sci. 2016;159:15–19. doi: 10.1016/j.lfs.2016.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ataga KI Derebail VK Caughey M, et al. Albuminuria is associated with endothelial dysfunction and elevated plasma endothelin-1 in sickle cell anemia. PLoS One. 2016;11(9):e0162652. doi: 10.1371/journal.pone.0162652 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kasztan M Fox BM Speed JS, et al. Long- term endothelin-A receptor antagonism provides robust renal protection in humanized sickle cell disease mice. J Am Soc Nephrol. 2017;28(8):2443–2458. doi: 10.1681/ASN.2016070711 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Heerspink HJL Parving HH Andress DL, et al. Atrasentan and renal events in patients with type 2 diabetes and chronic kidney disease (SONAR): a double-blind, randomised, placebo-controlled trial. Lancet. 2019;393(10184):1937–1947. doi: 10.1016/S0140-6736(19)30772-X [DOI] [PubMed] [Google Scholar]
  • 92.Trachtman H Nelson P Adler S, et al. DUET: a phase 2 study evaluating the efficacy and safety of sparsentan in patients with FSGS. J Am Soc Nephrol. 2018;29(11):2745–2754. doi: 10.1681/ASN.2018010091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Heerspink HJL Radhakrishnan J Alpers CE, et al. Sparsentan in patients with IgA nephropathy: a prespecified interim analysis from a randomised, double-blind, active-controlled clinical trial. Lancet. 2023;401(10388):1584–1594. doi: 10.1016/S0140-6736(23)00569-X [DOI] [PubMed] [Google Scholar]
  • 94.Charache S, Grisolia S, Fiedler AJ, Hellegers AE. Effect of 2,3-diphosphoglycerate on oxygen affinity of blood in sickle cell anemia. J Clin Invest. 1970;49(4):806–812. doi: 10.1172/JCI106294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Poillon WN, Kim BC, Labotka RJ, Hicks CU, Kark JA. Antisickling effects of 2,3-diphosphoglycerate depletion. Blood. 1995;85(11):3289–3296. doi: 10.1182/blood.V85.11.3289.bloodjournal85113289 [DOI] [PubMed] [Google Scholar]
  • 96.van Dijk MJ Rab MAE van Oirschot BA, et al. Safety and efficacy of mitapivat, an oral pyruvate kinase activator, in sickle cell disease: a phase 2, open-label study. Am J Hematol. 2022;97(7):E226–E229. doi: 10.1002/ajh.26554 [DOI] [PubMed] [Google Scholar]
  • 97.Vichinsky E Torres M Minniti CP, et al. Efficacy and safety of deferasirox compared with deferoxamine in sickle cell disease: two-year results including pharmacokinetics and concomitant hydroxyurea. Am J Hematol. 2013;88(12):1068–1073. doi: 10.1002/ajh.23569 [DOI] [PubMed] [Google Scholar]
  • 98.Scoglio M Cappellini MD D'Angelo E, et al. Kidney tubular damage secondary to deferasirox: systematic literature review. Children (Basel). 2021;8(12):1104. doi: 10.3390/children8121104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Allon M, Lawson L, Eckman JR, Delaney V, Bourke E. Effects of nonsteroidal anti-inflammatory drugs on renal function in sickle cell anemia. Kidney Int. 1988;34(4):500–506. doi: 10.1038/ki.1988.209 [DOI] [PubMed] [Google Scholar]
  • 100.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):1451–1456. doi: 10.1007/s00467-017-3623-6 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Kidney360 are provided here courtesy of American Society of Nephrology

RESOURCES