Learning Not to Judge a Book By Its Cover: A Case Series of Malignancy Masquerading As Common Presentations of Sickle Cell Disease

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4872-4872 ◽  
Author(s):  
Ahmar Urooj Zaidi ◽  
Lo'Rell Martin ◽  
Michael U Callaghan ◽  
Sharada A. Sarnaik ◽  
Jeffrey W Taub

Abstract Background: Sickle cell disease affects 100,000 people in the United States. Cancer does not appear to be more common in individuals with sickle cell disease however many presenting signs and symptoms of cancer overlap with signs and symptoms of sickle cell disease, complicating diagnosis. Furthermore, sickle cell disease can complicate surgical, chemo- and radio- therapies for malignancy. Methods: We reviewed the past 46 years experience of cancers in children with sickle cell at a large urban pediatric referral center. We identified 5 cases and reviewed the presentations, treatments, response to therapy, sickle cell complication and laboratory values, including transfusion needs and effect of chemotherapy on fetal hemoglobin levels. Results: We identified five cases of malignancies in children with sickle cell disease including cases of acute lymphoblastic leukemia (ALL), Hodgkin's lymphoma, Non-Hodgkin's lymphoma (NHL), Wilms tumor and renal cell carcinoma. An additional patient with sickle cell trait was diagnosed with metastatic renal medullary carcinoma. The patients were ages 6-18 years old at diagnosis and 4 were female. All had delays in diagnosis as their initial signs and symptoms overlapped with sickle cell disease (abdominal mass mimicking splenomegaly, low blood counts, fever and posterior reversible encephalopathy syndrome, hematuria). All five achieved complete remission with multi-modality therapy (excluding stem cell transplants) and are currently doing well 1- 20 years after treatment. Admissions for pain were reduced during and for some period after cancer therapy and 2 patients had marked, and in one case, prolonged elevation of fetal hemoglobin after therapy. Discussion: Children with sickle cell develop cancers that can be confused for sickle cell related complications. Cancer therapy can be well tolerated and successful in children with sickle cell. Sickle cell complications are decreased during cancer therapy likely secondary to frequent therapy related transfusion and therapy induced increases in fetal hemoglobin. Disclosures Callaghan: Biogen: Honoraria; Bayer: Honoraria; CSL Behring: Honoraria; Baxalta: Honoraria, Research Funding; Grifols: Honoraria; Roche: Honoraria, Research Funding.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1013-1013
Author(s):  
Frédéric Galactéros ◽  
Ersi Voskaridou ◽  
Anoosha Habibi ◽  
Giovanna Cannas ◽  
Laure Joseph ◽  
...  

Hydroxyurea (HU) is approved in the EU and USA for prevention of vaso-occlusive crises (VOC) including acute chest syndromes (ACS) in patients over 2 years with sickle-cell disease (SCD). The major benefits of HU in SCD are directly related to its abilities to increase HbF, decrease sickling of red blood cells and hemolysis, leading to reduction of vaso-occlusive episodes, need for blood transfusions and consequently reduction of morbidity and mortality. Adherence to the treatment is paramount for effectiveness, but in spite of proven benefits, barriers to adherence persist.[1] ESCORT-HU study (European Sickle Cell Disease COhoRT - HydroxyUrea), is a multicentric, prospective, non-interventional European study designed to evaluate the safety profile of HU in real life. Patients were enrolled from January 2009 to June 2017 with a follow-up of up to 10 years. All interruptions and resumptions of HU treatment exceeding 15 days were recorded in this study. We hereby present the analysis of the group of patients who self-discontinued HU at least once during the study before informing their caregiver, with a view to identify potential barriers to long-term adherence. In total, 1906 patients were enrolled in ESCORT-HU from 63 centers in France, Germany, Greece and Italy. Of these, 619 patients (32%) stopped HU for over 15 days at least once, and around a third (11% of all patients) were due to patient's will. The mean duration of HU treatment before the first discontinuation was 4.8 ± 5.1 years. Data are summarized in table 1. Compared to the rest of the cohort, the 'treatment discontinuation' group had similar distribution by gender and indication for HU prescription, but a higher proportion of adults stopped HU more than 15 days. It is notable that the proportion of patients with SC genotype was higher in the 'treatment discontinuation' group (4.5% vs 1.7%). The patients in the 'treatment discontinuation' group had more frequent SCD symptoms before enrolment in the study (table 2). Hematological and clinical improvement compared to the baseline was observed in both groups. However, average mean Corpuscular Volume (MCV) and Fetal Hemoglobin percentage (HbF%) were lower and mean percentages of patients with SCD symptoms were higher over the three years of follow-up in the 'treatment discontinuation' group, suggesting that HU daily dose was insufficient (table 2). Sixty patients have no treatment resumption date reported which suggest a permanent interruption of their treatment. Among them 32% preferred to switch to another HU medicinal product and 13% have safety issue (table 3). Understanding and managing self-discontinuation of HU before taking medical advice is challenging for the physician. It is tempting to speculate that it may be due, at least in part, to lack of effectiveness potentially due to an underdosage of the treatment. Resistance to the treatment may also be suggested based on past literature data revealing a great variability in the response (determined by HbF%) to HU therapy. There is evidence that genetic modifiers affect individual response to HU.[2],[3] Finally, weariness from long-term use may also explain the patient's wish to discontinue HU. But treatment at optimal effective should be the primary goal of caregivers. [1]Smaldone A., Manwani D., Green NS, Greater number of perceived barriers to hydroxyurea associated with poorer health-related quality of life in youth with sickle cell disease, Pediatr Blood Cancer. 2019 [2] Steinberg MH, Voskaridou E, Kutlar A, Loukopoulos D, Koshy M, et al. (2003). Concordant fetal hemoglobin response to hydroxyurea in siblings with sickle cell disease. Am J Hematol 72: 121-126 [3] Ware RE, Despotovic JM, Mortier NA, Flanagan JM, He J, et al. (2011) Pharmacokinetics, pharmacodynamics, and pharmacogenetics of hydroxyurea treatment for children with sickle cell anemia. Blood 118: 4985-4991 Disclosures Galactéros: Addmedica: Membership on an entity's Board of Directors or advisory committees. Voskaridou:Celgene Corporation: Consultancy, Research Funding; Protagonist: Research Funding; Genesis: Consultancy, Research Funding; Acceleron: Consultancy, Research Funding; Addmedica: Membership on an entity's Board of Directors or advisory committees. Cannas:Addmedica: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 967-967
Author(s):  
Nicolas Hebert ◽  
Erica B. Esrick ◽  
Myriam Armant ◽  
Christian Brendel ◽  
Marioara Felicia Ciuculescu ◽  
...  

Abstract NH and EE equally contributed. ADW and PB co-signed. The expression of fetal hemoglobin (HbF) is one of the main targets of sickle cell disease treatment, as it inhibits the polymerization of hemoglobin S. The hypothesis of an inhibitory threshold of HbF per red blood cell (RBC) has been suggested, 1 although not well defined, as the overall percentage of HbF does not reflect the heterogeneous distribution of HbF per cell. Likewise, the qualitative analysis of RBCs containing HbF, called F cells, is neither reproducible nor clinically interpretable, due to low expression. 2 We have developed a technique for measuring the amount of HbF per cell, to determine thresholds of HbF expression per RBC correlated with clinical and biological effects. 2 Among genes controlling its expression, BCL11A has a major repressive effect on the expression of gamma globin/HbF during the fetal to adult hemoglobin switch. Post-transcriptional silencing of BCL11A, using lentivirus expression of a shRNA embedded in a microRNA architecture (shmiR) to re-activate γ-globin expression, is safe and demonstrates high levels of %HbF in a pilot clinical study (NCT 03282656). 3 Here, we show the quantitative measurement of HbF per RBC and reticulocyte. Methods: During patient follow-up, HbF quantification per single cell RBC was performed using a fluorescent HbF antibody. 2 Addition of an anti-CD71 fluorescent antibody allowed selection of reticulocyte sub-populations for determining their HbF content. Fold-increase in percentage of RBC versus percentage of reticulocyte were calculated. Kinetics of HbF/RBC and HbF/Reticulocyte were modeled using mixed effects polynomial linear regression to account for the correlation between repeated data over time. Results: With a median follow-up of 15 months [12-20] after gene transfer, figure 1 shows the mathematical modeling of single-RBC HbF measurement representing RBC percentage containing at least 2, 4, 6, 8 and 10 pg of HbF. Percentage of RBC above each threshold was higher compared to 14 hydroxyurea treated patients for 6 months. Figure 2 shows fold increase between reticulocytes and RBCs with same thresholds of HbF/cell. For low thresholds, RBCs were found in same percentage as reticulocytes whereas RBCs containing increasing levels of HbF were found in higher percentage than reticulocytes, until 6pg/cell showing a clear selective advantage for red cells with a threshold ≥ 6pg/cell of HbF. Figure 3 shows different kinetics of HbF increase according to two different transduction strategies with 2 enhancers in patients 2-4 compared to one enhancer in patients 6-8. Conclusion: BCL11A down-regulation in six clinical trial subjects was associated with an in vivo selection process RBCs with ≥ 6pg HbF per cell attained with different engraftment kinetics, depending on transduction processes, and ultimately stable high level and broadly distributed HbF. 1 Steinberg MH, Chui DH, Dover GJ, Sebastiani P, Alsultan A. Fetal hemoglobin in sickle cell anemia: a glass half full? Blood. 2014 Jan 23;123(4):481-5. 2 Hebert N, Rakotoson MG, Bodivit G, et al. Individual red blood cell fetal hemoglobin quantification allows to determine protective thresholds in sickle cell disease. Am. J. Hematol. 3 Esrick EB, Lehmann LE, Biffi A, et al. Post-Transcriptional Genetic Silencing of BCL11A to Treat Sickle Cell Disease. N. Engl. J. Med. 2021;384(3):205-215. Figure 1 Figure 1. Disclosures Esrick: bluebird bio: Consultancy. Audureau: GBT: Honoraria. Higgins: Sebia, Inc.: Honoraria; Danaher Diagnostics: Consultancy. Williams: BioMarin: Membership on an entity's Board of Directors or advisory committees, Other: Insertion Site Advisory Board; Geneception: Membership on an entity's Board of Directors or advisory committees, Other: Scientific Advisory Board; Emerging Therapy Solutions: Membership on an entity's Board of Directors or advisory committees, Other: Chief Scientific Chair; Beam Therapeutics: Membership on an entity's Board of Directors or advisory committees, Other: Scientific Advisory Board; Alerion Biosciences: Other: Co-founder (now licensed to Avro Bio, potential for future milestones/royalties); Novartis: Membership on an entity's Board of Directors or advisory committees, Other: Steering Committee, Novartis ETB115E2201 (eltrombopag in aplastic anemia). Advisory fees donated to NAPAAC.; Orchard Therapeutics: Membership on an entity's Board of Directors or advisory committees, Other: Membership on a safety advisory board (SAB): SAB position ended 05/20/2021. Co-founder , Patents & Royalties: Potential for future royalty/milestone income, X-SCID. Provided GMP vector for clinical trial, Research Funding; bluebird bio: Membership on an entity's Board of Directors or advisory committees, Other: Insertion Site Analysis Advisory Board, Patents & Royalties: BCH licensed certain IP relevant to hemoglobinopathies to bluebird bio. The current license includes the potential for future royalty/milestone income. Bluebird has indicated they will not pursue this as a clinical program and BCH is negotiating return of, Research Funding. Bartolucci: AGIOS: Consultancy; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Lecture fees, Steering committee, Research Funding; Jazz Pharma: Other: Lecture fees; Emmaus: Consultancy; Addmedica: Consultancy, Other: Lecture fees, Research Funding; INNOVHEM: Other: Co-founder; Hemanext: Consultancy; GBT: Consultancy; Bluebird: Consultancy, Research Funding; F. Hoffmann-La Roche Ltd: Consultancy; Fabre Foundation: Research Funding.


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 ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4808-4808
Author(s):  
Salam Alkindi ◽  
Muna Almaskari ◽  
Shoaib Alzadjali ◽  
Rhea Misquith ◽  
Rajagopal Krishnamoorthy ◽  
...  

Abstract Abstract 4808 Although sickle cell disease (SCD) is a recessively inherited monogenic disorder, its clinical expression is variable from patient to patient, even within the same family; and, both heritable and environmental factors significantly contribute to this inter-patient difference in disease severity. The most prominent genetic modifiers include co-inherited alpha thalassaemia, over expression of fetal hemoglobin (HbF) in adulthood, and the excessive production of insoluble bilirubin. The purpose of this study was to ascertain the emerging role of HbF by studying several known SNP's in Chromosome 2, 6 and 11 that are involved in modulating the fetal hemoglobin levels in patients from Oman. This case-control study was conducted at Sultan Qaboos University Hospital from February to May 2010. Eighty eight SCD patients were consecutively studied along with forty four Omani blood donors who were selected randomly to complete a 2:1 case-control enrollment after an informed consent. Whole blood samples were collected in EDTA tubes from SCD patients and donors. DNA was extracted from the whole blood after Complete Blood Count and High Performance Liquid Chromatography were performed on the patient and donor samples. The molecular analysis included genotyping of BCL11A, HBSIL-MYB, HBG2 SNP's by direct DNA sequencing with 3100 genetic analyzer (ABI systems, Illinois, USA) using Tag SNP's in each category namely rs11886868, rs4671393 (BCL11A-Ch.2), rs7776054, rs9399137, rs4895441 (HBSIL-MYB-Ch.6), & rs7482144 (HBG2-Ch.11) to correlate these SNP's with the HbF expression in the SCD patients. A stepwise regression analysis result of the 6 SNP's genotyped at the BCL11A, HBSIL-MYB, and HBG2 gene locus were correlated to the HbF levels in our SCD study cohort and is shown below. At each step, the least significant SNP was removed, until all SNP's left in the multivariate model were significant. The model at step 4 has the best goodness of fit value, as assessed by the likelihood ratio test, than models 1, 2 & 3. [Table] Step 1: HbF levels~rs7482144 + rs7776054 + rs9399137 + rs4895441 + rs11886868 + rs4671393 Step 2: HbF levels~rs7482144 + rs7776054 + rs9399137 + rs11886868 + rs4671393 Step 3: HbF levels~rs7482144 + rs9399137 + rs11886868+ rs4671393 Step 4: HbF levels~rs7482144 + rs9399137 + rs4671393Table:Fetal hemoglobin association results for SNPs at the BCL11A, HBS1L-MYB, and HBG2 gene loci in the sickle cell cohort from Oman.SNP'sChromosomeMAF# (allele)Effect sizeStandard Errorp-valuers7482144110.262 (A)4.1610.6890.000*rs939913760.0875 (C)5.5631.2660.000*rs467139320.59 (A)-1.6610.8360.05*#MAF, minor allele frequency. Minor alleles (positive strand) are given in parenthesis.*A p value of <0.05 was considered as highly significant. Our study has demonstrated a significant association between the BCL11A, HBSIL-MYB, HBG2 SNP's and HbF levels. Together these 6 SNP's accounted for ~46% variation in the HbF levels in our SCD study subjects and in part explains the clinical heterogeneity seen in these patients. Three SNP's one each in Chromosome 2, 6 and 11 namely rs4671393, rs9399137 and rs7482144 respectively demonstrated the strongest effect on HbF levels. Disclosures: Alkindi: Sultan Qaboos University: Employment, Research Funding. Krishnamoorthy: INSERM U763: Employment, Research Funding. Pathare: Sultan Qaboos University: Employment, Research Funding.


1987 ◽  
Vol 73 (5) ◽  
pp. 523-524 ◽  
Author(s):  
Fortunato Morabito ◽  
Vincenzo Callea ◽  
Maura Brugiatelli ◽  
Domenico D'Ascola ◽  
Alfio Palazzolo ◽  
...  

A case of non-Hodgkin's lymphoma with leukemic spread in a patient affected with homozygous sickle cell disease is reported. This association has not been previously described. A correlation between the malignancy and the hemoglobinopathy could not be etiologically ascertained; therefore, an alternative explanation to a chance event cannot be offered.


2018 ◽  
Vol 40 (2) ◽  
pp. 118-120 ◽  
Author(s):  
Zainab Abdulmajeed Toorani ◽  
Srishma Sridhar ◽  
Wilfredo Roque

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 723-723
Author(s):  
Lance E. Palmer ◽  
Xin Zhou ◽  
Clay McLeod ◽  
Evadnie Rampersaud ◽  
Jeremie H. Estepp ◽  
...  

Abstract With the increase in availability of high depth whole genome sequencing (WGS) data of individuals with sickle cell disease (SCD), easy access to the raw sequencing data remains an issue due to technical and regulatory challenges. A compliant system that can provide facile data access would accelerate scientific discovery of genetic variants associated with clinical phenotypes. Cloud storage and computing provide an ultimate solution to this data access, which we have shown through the St. Jude Cloud (https://stjude.cloud) where over 5000 whole genome sequences for pediatric cancer patients are being shared in collaboration with DNANexus and Microsoft. Here we expand the St. Jude Cloud to sickle cell disease data through the Sickle Genome Project (SGP) Data Portal (https://pecan.stjude.org/permalink/sgp) to allow instantaneous raw data access (following data access committee approval), as well as visualization of genotype calls at individual level in a novel genome. The SGP WGS data was generated from 871 patients from St. Jude Children's Research Hospital (St. Jude) through the Sickle Cell Clinical Research and Intervention Program (SCCRIP, Pediatr Blood Cancer. 2018 May 24: e27228) and from Baylor College of Medicine (BCM). All study participants provided informed consent for genomic study and data sharing on IRB-approved research protocols. The SGP data portal will have multi-tiered access. All users will have access to a general heat map view which shows anonymized patient clinical values (e.g., fetal hemoglobin (HbF), mean corpuscular volume (MCV), hemoglobin concentration (Hb)) and relevant SCD modifying variants (e.g., Beta-globin locus, MYB, BCL11A, HBA). The GenomePaint bowser allows for viewing coding and noncoding variants. Displayed with each variant will be a visual indication of the median fetal hemoglobin values for patients homozygous for the reference allele, heterozygous, or homozygous for the alternative allele. The browser also displays erythroid specific DNA-accessibility and epigenetic marks and indicates variant that may disrupt erythroid specific transcription factor binding sites (GATA1 and BCL11A). For anonymization purposes, within the genome browser and heat map views, clinical values and the patients age will be binned into ranges when displayed as single or low count values. Lastly, the ProteinPaint tool (Zhang and Zhou, Nature Gen, Dec 29, 2015) will enable visualization and filtering of variants with reference to protein domain and amino acid sequence. To access processed data such as BAM and VCF files for downstream analyses, a user will be required to apply for access which will be adjudicated by a data access committee. Verified researchers will be granted access to clinical data in a manner consistent with the protocol specific informed consent documentation and protocol under which the sequencing was performed. This may include coded clinical and demographic data when specified by the research protocol and informed consent The SGP data set will be one of the first WGS datasets from primarily African American Sickle cell patients to be made available to clinicians and researchers worldwide. In addition, no SCD-centric data portal exists that contains controlled access to data and provides graphical tools for visual analysis. The combination of the visual tools and ability to download tools provides the scientific community an invaluable resource for studying sickle cell disease. Figure. Figure. Disclosures Estepp: Daiichi Sankyo: Consultancy; NHLBI: Research Funding; Global Blood Therapeutics: Consultancy, Research Funding; ASH Scholar: Research Funding. Hankins:Global Blood Therapeutics: Research Funding; bluebird bio: Consultancy; NCQA: Consultancy; Novartis: Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5889-5889
Author(s):  
Stuthi Pavani Perimbeti ◽  
Kevin Ye Hou ◽  
Sabarina Ramanathan ◽  
Adonas Woodard ◽  
Daniel Kyung ◽  
...  

Abstract Introduction: Case reports have suggested that there is an increased risk of hematological malignancies with sickle cell disease (SCD). We aimed to investigate the prevalence and mortality of select hematological malignancies, including: acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), polycythemia vera (PV), essential thrombocytopenia (ET), Hodgkin's lymphoma (HL) and non-Hodgkin's lymphoma (NHL), primary myelofibrosis (PMF) in patients hospitalized with SCD. Methods: We queried the Healthcare Cost and Utilization Project's Nationwide Inpatient Sample to identify the hospitalizations with SCD using ICD-9 codes 282.6, 282.60, 282.61, 282.62, 282.63, 282.64, 282.68, 282.69 from 1999 to 2014. Cases were then stratified based on the concurrent presence of the above hematological malignancies. The percentages of patients with each type of hematological malignancy were obtained using Chi-square analysis. In addition, we compared outcomes between patients with and without SCD who had hematological malignancies. Bivariate analysis for in-hospital mortality percentage was performed using Chi-square test. Multivariate analysis to evaluate the risk of death during hospitalization was performed using Cox proportional hazard regression with alpha set at 0.05. Results: There were 307,424 admissions (weighted=1,513,168) with SCD. Within these admissions, 0.04% (n=516) were associated with AML, 0.05% (n=720) with HL, 0.02% (n=47) with ALL, 0.07% (n=1,028) with NHL, 0.69% (n=10,654) with ET, 0.01% (n=20) with PMF and 0.01% (n=18) with PV. The hazard ratio for mortality (95% C.I.) with SCD compared to without SCD was 3.60 (1.24-4.67) for AML (p <0.001), 4.56 (2.78-6.78) for ET (p <0.001), 2.37 (1.41-5.67) for NHL (p=0.003), 1.5 (0.8-4.5) for HL (p=0.08), 1.1 (0.5-3.2) for ALL (p=0.04), 1.3 (1.1-3.2) for PMF (p= 0.03) and 1.7 (0.6-2.9) for PV (p=0.05). Discussion: The most frequently encountered hematological malignancy in patients with SCD was ET, followed by NHL and then HL. After controlling for multiple confounders including age, race, sex, comorbidities and socioeconomic status, the hazard of death during hospitalization with SCD, ET, AML, NHL and ALL are significantly higher compared to those without SCD. While uncommonly encountered, concomitant hematological malignancy and SCD portends a significantly worse outcome, particularly in ET and AML. Potential explanations include iron overload from prior transfusions, increased infection risk due to asplenia and vascular damage from previous vaso-occlusive events. Newer advances in the management of SCD might improve subsequent outcomes in hematological malignancies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3969-3969
Author(s):  
So Hyun Julie Park Park ◽  
Mingming Cao ◽  
Yankai Zhang ◽  
Vivien A. Sheehan ◽  
Gang Bao

Abstract Introduction: Several gene editing strategies have been developed to cure sickle cell disease (SCD), including the use of CRISPR/Cas9 to edit beta-globin (HBB), gamma-globin (HBG), or B-cell lymphoma/leukemia 11A (BCL11A) in hematopoietic stem and progenitor cells (HSPCs) from patients with SCD. Although high gene-editing rates can be achieved and off-target effects reduced, new challenges in applying the gene-editing strategies, including unintended gene modifications, need to be addressed in order to cure SCD with high efficacy and safety. To date, due to limitations in sequencing methods, studies on CRISPR/Cas9 genome editing for treating SCD only identified small insertions/deletions (INDELs); the extent and consequences of unintended large gene modifications are generally unknown. Here we provide accurate quantification and profiling of unintended gene modifications due to Cas9 induced double-stranded breaks (DSBs) in SCD HSPCs, including large deletions, insertions, and complex chromosomal arrangements, and the comparison of different approaches. Methods: R-66S gRNA targets the sickle mutation on the HBB. R-02 gRNA generates a DSB 16 bp away from the sickle mutation site. SD-02 gRNA introduces a 13-bp Hereditary Persistence of Fetal Hemoglobin (HPFH) deletion as a major INDEL in the HBG1/HBG2 promoter to reactivate fetal hemoglobin (HbF). BCL11A gRNA targets the GATA1 site at the BCL11A erythroid enhancer to induce HbF. R-66S, R-02, SD-02, and BCL11A gRNAs were respectively complexed with SpyCas9 and delivered as ribonucleoprotein (RNP) to SCD HSPCs. To accurately quantify CRISPR/Cas9 induced large modifications in gene-edited SCD HSPCs, we used PacBio Single Molecule, Real-Time (SMRT) Sequencing with Unique Molecular Identifiers (UMI). The 5-6 kb region around the Cas9 cut-site was dual-UMI tagged using two PCR cycles. The second and third PCR was performed with minimal cycle numbers to enrich the UMI-tagged template molecules. The SMRTbell library composed of edited and unedited SCD HSPCs samples was sequenced on a PacBio Sequel II 8M flowcell using the circular consensus sequencing (CCS) mode. The PacBio subreads were converted to HiFi reads and subjected to UMI consensus read generation and variant calling. Results: SMRT-seq with UMI revealed high rates and broad spectra of unintended large deletions (&gt; 200 bp) induced by Cas9 cutting at HBB, HBG1, and BCL11A genes in RNP treated samples, with respectively R-66S RNP, 31.7%; R-02 RNP, 17.4%; SD-02 RNP, 13.3%; BCL11A RNP, 40%. The large deletions have a very broad distribution of sizes and locations. In addition, we found large insertions (&gt; 50 bp) and local complex chromosomal rearrangements at the Cas9 cut-sites. Therefore, the current assessment of gene-editing rates using short-read Next Generation Sequencing (NGS) misses a substantial proportion of Cas9-cutting induced large gene modifications, resulting in an inaccurate measure of both allele and genotype frequencies. Discussions: We found that unintended on-target large deletions occur at high rates at HBB, HBG1, and BCL11A in gene-edited SCD HSPCs. These results raise significant safety concerns regarding gene-editing of HSPCs to treat SCD. Our results demonstrate the importance of detecting and quantifying all possible CRISPR/Cas9 gene-editing outcomes to ensure the efficient and safe translation of gene-editing-based strategies to cure SCD and other human diseases. Additional work is required to determine the functional consequences of the unintended gene modifications and the persistence of the unintended large gene modifications at the on-target cut-sites. Disclosures Sheehan: Forma Therapeutics: Research Funding; Beam Therapeutics: Research Funding; Novartis: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document