scholarly journals Genetic modifiers of fetal hemoglobin affect the course of sickle cell disease in patients treated with hydroxyurea

Haematologica ◽  
2021 ◽  
Author(s):  
Pierre Allard ◽  
Nareen Alhaj ◽  
Stephan Lobitz ◽  
Holger Cario ◽  
Andreas Jarisch ◽  
...  

The course of sickle cell disease (SCD) is modified by polymorphisms boosting fetal hemoglobin (HbF) synthesis. However, it has remained an open question how these polymorphisms affect patients who are treated with the HbF-inducing drug hydroxyurea/hydroxycarbamide. The German SCD registry offers the opportunity to answer this question, because >90% of patients are treated according to national guidelines recommending the use of hydroxyurea in all patients above 2 years of age. We analyzed the modifying effect of HbF-related genetic polymorphisms in 417 patients with homozygous SCD >2 years who received hydroxyurea. HbF levels were correlated with higher total hemoglobin levels, lower rates of hemolysis, a lower frequency of painful crises and of red blood cell transfusions. The minor alleles of the polymorphisms in the γ-globin promoter (rs7482144), BCL11A (rs1427407) and HMIP (rs66650371) were strongly associated with increased HbF levels. However, these associations did not translate into lower frequencies of vasoocclusive events that did not differ between patients either carrying or not carrying the HMIP and BCL11A polymorphisms. Patients on hydroxyurea carrying the γ- globin promoter polymorphism demonstrated substantially higher hemoglobin levels (p

Blood ◽  
2012 ◽  
Vol 120 (18) ◽  
pp. 3822-3828 ◽  
Author(s):  
Christopher J. Bean ◽  
Sheree L. Boulet ◽  
Dorothy Ellingsen ◽  
Meredith E. Pyle ◽  
Emily A. Barron-Casella ◽  
...  

Abstract Sickle cell disease is a common hemolytic disorder with a broad range of complications, including vaso-occlusive episodes, acute chest syndrome (ACS), pain, and stroke. Heme oxygenase-1 (gene HMOX1; protein HO-1) is the inducible, rate-limiting enzyme in the catabolism of heme and might attenuate the severity of outcomes from vaso-occlusive and hemolytic crises. A (GT)n dinucleotide repeat located in the promoter region of the HMOX1 gene is highly polymorphic, with long repeat lengths linked to decreased activity and inducibility. We examined this polymorphism to test the hypothesis that short alleles are associated with a decreased risk of adverse outcomes (hospitalization for pain or ACS) among a cohort of 942 children with sickle cell disease. Allele lengths varied from 13 to 45 repeats and showed a trimodal distribution. Compared with children with longer allele lengths, children with 2 shorter alleles (4%; ≤ 25 repeats) had lower rates of hospitalization for ACS (incidence rate ratio 0.28, 95% confidence interval, 0.10-0.81), after adjusting for sex, age, asthma, percentage of fetal hemoglobin, and α-globin gene deletion. No relationship was identified between allele lengths and pain rate. We provide evidence that genetic variation in HMOX1 is associated with decreased rates of hospitalization for ACS, but not pain. This study is registered at www.clinicaltrials.gov as #NCT00072761.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3641-3641 ◽  
Author(s):  
Evadnie Rampersaud ◽  
Lance E. Palmer ◽  
Jane S. Hankins ◽  
Vivien A Sheehan ◽  
Wenjian Bi ◽  
...  

Abstract Although sickle cell disease (SCD) is a monogenic disorder, the severity and specific organ dysfunction and failure are strongly influenced by genetic modifiers. Rapid identification of all modifiers in patients and well-phenotyped cohorts will better define the impact of relevant variants on clinical status, inform disease biology, and identify new therapeutic strategies. We created the Sickle Genome Project (SGP), a whole genome sequencing (WGS) strategy, to define genomic variation and modifiers of SCD. We performed WGS on 871 African American SCD patients from St. Jude Children's Research Hospital who participated in the Sickle Cell Clinical Research and Intervention Program (SCCRIP, Hankins et al. Pediatr Blood Cancer, 2018) and Texas Children's Hospital Hematology Center (TCHC). We developed robust pipelines for accurate detection of single nucleotide polymorphisms (SNPs), identification of structural variants and data retrieval/sharing via the St. Jude Cloud platform (to be described elsewhere). Notable findings include: 1) Confirmed associations of common genetic modifiers with SCD phenotypes, including levels of fetal hemoglobin (BCL11A, HBS1L-MYB, HBB), bilirubin (UGT1A1), and microalbuminuria (APOL1). Additional associations approaching genome-wide significance require further investigation, including replication in independent samples. 2) Improved determination of the SCD modifier α-thalassemia. The most common α-thalassemia mutations in SCD are 3.7 kb or 4.2 kb deletions (-α3.7 and -α4.2 alleles), which arose from recombination between homologous HBA1 and HBA2 genes and are difficult to map using standard WGS reads. Three independent crossover events are described for -α3.7 and one for -α4.2 in SCD cohorts. We developed a novel approach to identify α-globin gene deletions by local de novo assembly of WGS data and coverage depth analysis. We identified 5 -α3.7 alleles (frequencies 0.77-32.12%) and 7 -α4.2 alleles (frequencies 0.19-5.77%). Collectively, the frequency of all -α alleles was 57%, reflecting at least 12 distinct recombination events, greatly exceeding previously published counts. These findings better define the evolution of α-globin genes to allow improved understanding of their regulation and influence on SCD. 3) Characterization of β0-thalassemia alleles. Mutations in the extended β-globin locus influence SCD phenotypes. Five SGP patients had large β-globin (HBB) deletions associated with elevated fetal hemoglobin, which ameliorates symptoms of SCD. Twenty-three patients had HbSβ0-thalassemia, which reduces the severity of some SCD phenotypes. Overall, 48.6% (18/37) of patients clinically designated as HbSβ0 -thalassemia had no identified β-thalassemia mutation. Moreover, 4/680 patients (0.6%) designated HbSS were identified to be β0-thalassemia heterozygotes. The MCV, RBC and %HbA2 distributions overlapped substantially in correct vs. incorrect genotype assignments. Improved discrimination of HbSβ0 vs HbSS genotypes by WGS will better define associated phenotype differences to impact clinical care. 4) Determination of a genetic variant linked to vaso-occlusive crisis (VOC). Previously, a single GWAS study linked rs3115229, located 63.7 kb 5′ upstream of the KIAA1109 gene, with VOC at borderline significance (P = 5.63 × 10−8) (Chaturvedi et al, Blood 130, 2017). Using WGS data for 327 SGP participants (HbSS or HbSβ0-thalassemia) enrolled in the SCCRIPP study, we found strong association (p = 7 x 10-5) between the onset of VOC and a 4-SNP diplotype within an adjacent LD block of the KIAA1109-TENR-IL2-IL21 region (chr4: 122.8Mb - 123.8Mb) which has been previously associated with numerous inflammatory disorders. We validated this association using imputed genome-wide array data in an independent group of SCD patients (Sleep and Asthma Cohort, n= 181 patients, p = 0.05) (Cohen et al, Ann Am Thorac Soc, 2016). This works provides confirmation that the region surrounding KIAA1109 is associated with pain crisis in SCD. Our studies provide new information on the genomic architecture of SCD patients and delineate a consolidated approach for future applications of precision medicine. Disclosures Hankins: Novartis: Research Funding; Global Blood Therapeutics: Research Funding; NCQA: Consultancy; bluebird bio: Consultancy. Estepp:Global Blood Therapeutics: Consultancy, Research Funding; ASH Scholar: Research Funding; NHLBI: Research Funding; Daiichi Sankyo: Consultancy.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2216-2216
Author(s):  
Vivien A Sheehan ◽  
Jacy R Crosby ◽  
Jonathan Michael Flanagan ◽  
Thad A Howard ◽  
Nicole A. Mortier ◽  
...  

Abstract Hydroxyurea is a safe and effective therapy for sickle cell disease (SCD), with the majority of its benefit correlating with the amount of fetal hemoglobin (HbF) produced at maximum tolerated dose, or MTD. There is substantial individual variability in HbF response to hydroxyurea, with baseline HbF levels accounting for approximately 40% of the observed variability in final HbF at MTD. Several genetic modifiers of baseline, or endogenous, HbF levels have previously been identified by genome wide association studies. These include certain beta-globin haplotypes, and polymorphisms at the BCL11A and HBS1L-MYB gene loci. The effect of these known genetic modifiers of baseline HbF on drug response has been investigated in a small (n=38) cohort of pediatric patients treated with hydroxyurea in several centers with different treatment guidelines. In this limited group, no association was found between the BCL11A or HBS1L-MYB variants and the change in HbF at MTD (ΔHbF; final HbF minus baseline HbF). Co-inheritance of alpha thalassemia has been reported to have a negative effect on HbF response to hydroxyurea. To independently verify these findings in a larger sample size, we tested the effect of four BCL11A single nucleotide polymorphisms (SNPs), three HBS1L-MYB SNPs, the XmnI polymorphism, and co-inheritance of α-thalassemia on ΔHbF at MTD in a cohort of 171 pediatric SCA patients, treated prospectively and uniformly on HUSTLE (NCT NCT00305175) and SWiTCH (NCT 00122980) protocols. These patients represent the most accurate hydroxyurea phenotypes available, as all were meticulously titrated to MTD, and had complete laboratory data demonstrating compliance, such as absolute neutrophil count and absolute reticulocyte count within the therapeutic range. In our cohort, the BCL11A SNPs rs1427407, rs4671393 and rs11886868 were significantly associated with baseline HbF (Table 1). We saw no association between baseline HbF and any of the HBS1L-MYB SNPs, XmnI, α-thalassemia or BCL11A SNP rs7599488. In contrast to other reports, we found that coinheritance of α-thalassemia did not affect hydroxyurea treatment response (p=0.088). We found that BCL11A SNPs rs1427407, rs4671393 and rs11886868 were significantly associated with reduced ΔHbF following hydroxyurea treatment (Table 1), where individuals with the BCL11A SNPs had a smaller ΔHbF compared to individuals without the polymorphisms, as shown by the negative baseline β-value. For example, one BCL11A rs1427407 SNP is associated with a ΔHbF 3.46 percentage points lower than individuals without the SNP at MTD, with an additive, dose effect of the SNP at the second allele; homozygous individuals have higher baseline HbF, lower ΔHbF compared to heterozygotes or wild-type individuals (Figure 1). The other variants HBS1L-MYB SNPs, XmnI, α-thalassemia or BCL11A SNP rs7599488 did not significantly impact ΔHbF. None of the tested polymorphisms, including the BCL11A SNPs, were associated with a significant difference in final HbF levels. Individuals with higher baseline HbF due to BCL11A polymorphisms demonstrate a statistically significant lower rise in HbF in response to hydroxyurea than individuals without these polymorphisms. Identification of more variants associated with baseline and ΔHbF through next generation sequencing will help elucidate whether the negative effect of high baseline on ΔHbF is a BCL11A specific effect, or a manifestation of a general threshold effect, that there is a maximum amount of HbF an individual is able to achieve through hydroxyurea induction.Table 1Association between BCL11A SNPs and response to hydroxyurea.GeneSNP IDBaseline β-value (lnHbF)Baseline p-valueΔHbF β-value (%HbF)ΔHbF p-valueBCL11Ars14274070.3569.05x10-5-3.461.02x10-3BCL11Ars46713930.272.17x10-3-2.627.21x10-3BCL11Ars118868680.131.61x10-4-3.007.86x10-4BCL11Ars75994880.080.30-0.780.33Figure 1Effect of BCL11A rs1427407 on Hydroxyurea Response. Average baseline and ΔHbF values are shown for a sample BCL11A variant.Figure 1. Effect of BCL11A rs1427407 on Hydroxyurea Response. Average baseline and ΔHbF values are shown for a sample BCL11A variant. Disclosures: Off Label Use: Hydroxyurea is not FDA approved for use in pediatric sickle cell patients.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3747-3747
Author(s):  
Margaret A. Keller ◽  
Meggin K. Borowski ◽  
Marcella Devoto ◽  
Hui-Hua Li ◽  
Morgan Brown ◽  
...  

Abstract Renal insufficiency is the source of significant morbidity and mortality in adults with sickle cell disease (SCD). A population of adult patients with SCD and end-stage renal disease (ESRD) requiring hemodialysis was examined for factors that predict ESRD. ESRD patients (mean age 36.8 +/− 7.74) were compared with patients without ESRD over 50 years old (mean age 59.3 +/− 8.31). We found that the ESRD population was characterized by more males (14 males, 8 females ESRD vs. 4 males, 15 females non-ESRD), a higher occurrence of the CAR beta-like globin cluster haplotype (44 % ESRD vs. 11 % non-ESRD) and relatively lower fetal hemoglobin levels (3.6 +/− 2.16 ESRD vs. 8.6 +/− 6.35 non-ESRD). In order to further examine potential predictors of renal disease, including genetic modifiers unlinked to the beta-globin cluster, we examined a broader population of patients with elevated creatinine, defined as levels greater than 1.0 mg/dl, who did or did not require hemodialysis. We hypothesize that gender, beta-like globin cluster haplotype, fetal hemoglobin levels and polymorphisms in critical regulators of vasomotor tone are predictors of renal insufficiency in SCD. Genotyping of a single nucleotide polymorphism (SNP) in endothelial nitric oxide synthase (eNOS or NOS3) and an insertion/deletion polymorphism (I/D) in the angiotensin-converting enzyme (ACE) gene will allow us to determine if these polymorphisms are over-represented in this cohort. The eNOS T-786C SNP alters the level of transcription of the eNOS gene and the amount of eNOS activity. We recently showed that this polymorphism was associated with a history of acute chest syndrome in females with SCD (Br. J. Haematol., 2004, 124:240). The ACE I/D polymorphism has been associated with altered plasma ACE levels. Twenty-one adult patients with sickle cell disease with renal insufficiency and 25 age- and gender-matched control patients seen at Thomas Jefferson University are being examined. Genotype results and statistical analysis will be presented. It is hoped that identification of predictors of renal insufficiency in patients with SCD might lead to early identification of at-risk patients, allowing for therapeutic intervention.


2008 ◽  
Vol 105 (33) ◽  
pp. 11869-11874 ◽  
Author(s):  
G. Lettre ◽  
V. G. Sankaran ◽  
M. A. C. Bezerra ◽  
A. S. Araujo ◽  
M. Uda ◽  
...  

Hematology ◽  
2013 ◽  
Vol 2013 (1) ◽  
pp. 362-369 ◽  
Author(s):  
Deepa Manwani ◽  
Paul S. Frenette

Abstract Recurrent and unpredictable episodes of vaso-occlusion are the hallmark of sickle cell disease. Symptomatic management and prevention of these events using the fetal hemoglobin–reactivating agent hydroxyurea are currently the mainstay of treatment. Discoveries over the past 2 decades have highlighted the important contributions of various cellular and soluble participants in the vaso-occlusive cascade. The role of these elements and the opportunities for therapeutic intervention are summarized in this review.


2002 ◽  
Vol 2 ◽  
pp. 1706-1728 ◽  
Author(s):  
Martin H. Steinberg

High fetal hemoglobin (HbF) levels inhibit the polymerization of sickle hemoglobin (HbS) and reduce the complications of sickle cell disease. Pharmacologic agents that can reverse the switch from γ- to β-chain synthesis — γ-globin chains characterize HbF, and sickle β-globin chains are present in HbS — or selectively increase the proportion of adult erythroid precursors that maintain the ability to produce HbF are therapeutically useful. Hydroxyurea promotes HbF production by perturbing the maturation of erythroid precursors. This treatment increases the total hemoglobin concentration, reduces the vaso-occlusive complications of pain and acute chest syndrome, and attenuates mortality in adults. It is a promising beginning for pharmacologic therapy of sickle cell disease. Still, its effects are inconsistent, trials in infants and children are ongoing, and its ultimate value — and peril — when started early in life are still unknown.


Blood ◽  
1997 ◽  
Vol 90 (2) ◽  
pp. 891-892
Author(s):  
Rita Selby ◽  
Eric Nisbet-Brown ◽  
Raveen K. Basran ◽  
Lebe Chang ◽  
Nancy F. Olivieri

Blood ◽  
2012 ◽  
Vol 120 (3) ◽  
pp. 528-537 ◽  
Author(s):  
Karina Yazdanbakhsh ◽  
Russell E. Ware ◽  
France Noizat-Pirenne

Abstract Red blood cell transfusions have reduced morbidity and mortality for patients with sickle cell disease. Transfusions can lead to erythrocyte alloimmunization, however, with serious complications for the patient including life-threatening delayed hemolytic transfusion reactions and difficulty in finding compatible units, which can cause transfusion delays. In this review, we discuss the risk factors associated with alloimmunization with emphasis on possible mechanisms that can trigger delayed hemolytic transfusion reactions in sickle cell disease, and we describe the challenges in transfusion management of these patients, including opportunities and emerging approaches for minimizing this life-threatening complication.


Sign in / Sign up

Export Citation Format

Share Document