151
|
Pendergrast J, Ajayi LT, Kim E, Campitelli MA, Graves E. Sickle cell disease in Ontario, Canada: an epidemiologic profile based on health administrative data. CMAJ Open 2023; 11:E725-E733. [PMID: 37582620 PMCID: PMC10435244 DOI: 10.9778/cmajo.20220145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND The number of patients with sickle cell disease in Ontario, Canada, is unknown. In the absence of a formal registry, we performed a study to determine an approximate census via analysis of health administrative databases. METHODS We identified Ontario patients with a diagnosis of sickle cell disease through queries of the Discharge Abstract Database, National Ambulatory Care Reporting System and Newborn Screening Ontario database. The period of inquiry was Apr. 1, 2007, through Mar. 31, 2017. We identified repeat interactions by the same patient by cross-referencing provincial health insurance plan numbers. RESULTS We documented health care system interactions for 3418 unique patients (1912 [55.9%] female, median age at the time of identification 24 yr). Over the 10-year study period, patients visited the emergency department a median of 2 (interquartile range [IQR] 1-7) times and an average of 6.69 (standard deviation [SD] 26.71) times, and were admitted to hospital a median of 1 (IQR 1-5) time and an average of 4.38 (SD 8.53) times for treatment related to sickle cell disease. A total of 229 patients (6.7%) died during the study period, with an average age at death of 55 years. Even without accounting for the effects of immigration, the rate of natural increase slowed slightly over the study period owing to a decrease in the annual number of affected births. INTERPRETATION The estimated prevalence of patients with sickle cell disease in Ontario in 2007/08-2016/17 was 1 in 4200, and affected patients' need for hospital-based care was substantial, although highly variable. Similar queries of health administrative databases may be feasible in other Canadian provinces.
Collapse
Affiliation(s)
- Jacob Pendergrast
- Department of Medical Oncology and Hematology (Pendergrast), University Health Network; Department of Medicine (Pendergrast), University of Toronto; Sickle Cell Awareness Group of Ontario (Tunji Ajayi); ICES Central (Kim, Campitelli, Graves), Toronto, Ont.
| | - Lanre Tunji Ajayi
- Department of Medical Oncology and Hematology (Pendergrast), University Health Network; Department of Medicine (Pendergrast), University of Toronto; Sickle Cell Awareness Group of Ontario (Tunji Ajayi); ICES Central (Kim, Campitelli, Graves), Toronto, Ont
| | - Eliane Kim
- Department of Medical Oncology and Hematology (Pendergrast), University Health Network; Department of Medicine (Pendergrast), University of Toronto; Sickle Cell Awareness Group of Ontario (Tunji Ajayi); ICES Central (Kim, Campitelli, Graves), Toronto, Ont
| | - Michael A Campitelli
- Department of Medical Oncology and Hematology (Pendergrast), University Health Network; Department of Medicine (Pendergrast), University of Toronto; Sickle Cell Awareness Group of Ontario (Tunji Ajayi); ICES Central (Kim, Campitelli, Graves), Toronto, Ont
| | - Erin Graves
- Department of Medical Oncology and Hematology (Pendergrast), University Health Network; Department of Medicine (Pendergrast), University of Toronto; Sickle Cell Awareness Group of Ontario (Tunji Ajayi); ICES Central (Kim, Campitelli, Graves), Toronto, Ont
| |
Collapse
|
152
|
Castro-Sesquen YE, Saraf SL, Gordeuk VR, Nekhai S, Jerebtsova M. Use of multiple urinary biomarkers for the early detection of chronic kidney disease in sickle cell anemia. Blood Adv 2023; 7:2606-2608. [PMID: 36634264 PMCID: PMC10250912 DOI: 10.1182/bloodadvances.2022008006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/08/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Affiliation(s)
| | - Santosh L. Saraf
- Section of Hematology-Oncology, Department of Medicine, University of Illinois Chicago, Chicago, IL
| | - Victor R. Gordeuk
- Section of Hematology-Oncology, Department of Medicine, University of Illinois Chicago, Chicago, IL
| | - Sergei Nekhai
- Department of Microbiology, Howard University, Washington, DC
- Center for Sickle Cell Disease, Howard University, Washington, DC
- Department of Medicine, College of Medicine, Howard University, Washington, DC
| | | |
Collapse
|
153
|
Antonelli Rossi DA, De Araujo Junior JA, Luvizutto GJ, Bazan R, Salmazo PS, Modolo GP, Hueb JC, Nunes HRDC, Hokama NK, Minicucci MF, Roscani MG, Zanati Bazan SG. Effect of a Physical Exercise Program on the Inflammatory Response, Cardiac Functions, Functional Capacity, and Quality of Life in Patients with Sickle Cell Disease. J Clin Med 2023; 12:3952. [PMID: 37373647 PMCID: PMC10299033 DOI: 10.3390/jcm12123952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/13/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION The beneficial effects of physical exercise on functional capacity and inflammatory response are well-known in cardiovascular diseases; however, studies on sickle cell disease (SCD) are limited. It was hypothesized that physical exercise may exert a favorable effect on the inflammatory response of SCD patients, contributing to an improved quality of life. This study aimed to evaluate the effect of a regular physical exercise program on the anti-inflammatory responses in SCD patients. METHODS A non-randomized clinical trial was conducted in adult SCD patients. The patients were divided into two groups: 1-Exercise Group, which received a physical exercise program three times a week for 8 weeks, and; 2-Control Group, with routine physical activities. All patients underwent the following procedures initially and after eight weeks of protocol: clinical evaluation, physical evaluation, laboratory evaluation, quality of life evaluation, and echocardiographic evaluation. STATISTICAL ANALYSIS Comparisons between groups were made using Student's t-test, Mann-Whitney test, chi-square test, or Fisher's exact test. Spearman's correlation coefficient was calculated. The significance level was set at p < 0.05. RESULTS There was no significant difference in inflammatory response between the Control and Exercise Groups. The Exercise Group showed an improvement in peak VO2 values (p < 0.001), an increase in the distance walked (p < 0.001), an improvement in the limitation domain due to the physical aspects of the 36-Item Short Form Health Survey (SF-36) quality of life questionnaire (p = 0.022), and an increase in physical activity related to leisure (p < 0.001) and walking (p = 0.024) in the International Physical Activity Questionnaire (IPAQ). There was a negative correlation between IL-6 values and distance walked on the treadmill (correlation coefficient -0.444, p = 0.020) and the estimated peak VO2 values (correlation coefficient -0.480; p = 0.013) in SCD patients in both groups. CONCLUSIONS The aerobic exercise program did not change the inflammatory response profile of SCD patients, nor did it show unfavorable effects on the parameters evaluated, and patients with lower functional capacity were those with the highest levels of IL-6.
Collapse
Affiliation(s)
- Daniele Andreza Antonelli Rossi
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | - Jonas Alves De Araujo Junior
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | - Gustavo José Luvizutto
- Department of Applied Physical Therapy, Federal University of Triângulo Mineiro, Uberaba 38065-430, Brazil;
| | - Rodrigo Bazan
- Department of Neurology, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-970, Brazil; (R.B.); (G.P.M.)
| | - Péricles Sidnei Salmazo
- Department of Medicine, Faculty of Medical and Health Sciences, Pontifical Catholic University of São Paulo, Sorocaba 18052-490, Brazil;
| | - Gabriel Pinheiro Modolo
- Department of Neurology, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-970, Brazil; (R.B.); (G.P.M.)
| | - João Carlos Hueb
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | | | - Newton Key Hokama
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | - Marcos Ferreira Minicucci
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | - Meliza Goi Roscani
- Department of Medicine, Federal University of Sao Carlos-UFSCar, São Carlos 13565-251, Brazil;
| | - Silméia Garcia Zanati Bazan
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| |
Collapse
|
154
|
Ma L, Yang S, Peng Q, Zhang J, Zhang J. CRISPR/Cas9-based gene-editing technology for sickle cell disease. Gene 2023; 874:147480. [PMID: 37182559 DOI: 10.1016/j.gene.2023.147480] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023]
Abstract
Sickle cell disease (SCD) is the most common monogenic hematologic disorder and is essentially congenital hemolytic anemia caused by an inherited point mutation in the β-globin on chromosome 11. Although the genetic basis of SCD was revealed as early as 1957, treatment options for SCD have been very limited to date. Hematopoietic stem cell transplantation (HSCT) was thought to hold promise as a cure for SCD, but the available donors were still only 15% useful. Gene therapy has advanced rapidly into the 21st century with the promise of a cure for SCD, and gene editing strategies based on the cluster-based regularly interspaced short palindromic repeat sequence (CRISPR)/Cas9 system have revolutionized the field of gene therapy by precisely targeting genes. In this paper, we review the pathogenesis and therapeutic approaches of SCD, briefly summarize the delivery strategies of CRISPR/Cas9, and finally discuss in depth the current status, application barriers, and solution directions of CRISPR/Cas9 in SCD. Through the review in this paper, we hope to provide some references for gene therapy in SCD.
Collapse
Affiliation(s)
- Liangliang Ma
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China
| | - Shanglun Yang
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China
| | - Qianya Peng
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China
| | - Jingping Zhang
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China
| | - Jing Zhang
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China.
| |
Collapse
|
155
|
Ataga KI, Zhou Q, Saraf SL, Hankins JS, Ciccone EJ, Loehr LR, Garrett ME, Ashley-Koch AE, Cai J, Telen MJ, Derebail VK. Sex differences in progression of kidney disease in sickle cell disease. Haematologica 2023; 108:1436-1441. [PMID: 36546431 PMCID: PMC10153522 DOI: 10.3324/haematol.2022.281677] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Kenneth I Ataga
- Center for Sickle Cell Disease, University of Tennessee Health Scienter Center, Memphis, TN.
| | - Qingning Zhou
- Department of Mathematics and Statistics, University of North Carolina at Charlotte, Charlotte, NC
| | | | - Jane S Hankins
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Emily J Ciccone
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Laura R Loehr
- Division of General Medicine and Clinical Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC
| | | | - Jianwen Cai
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Marilyn J Telen
- Division of Hematology, Duke University Medical Center, Durham, NC
| | - Vimal K Derebail
- UNC Kidney Center, Division of Nephrology and Hypertension, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
156
|
Poku BA, Atkin KM, Kirk S. Self-management interventions for children and young people with sickle cell disease: A systematic review. Health Expect 2023; 26:579-612. [PMID: 36597596 PMCID: PMC10010100 DOI: 10.1111/hex.13692] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/03/2022] [Accepted: 12/11/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Increasing numbers of interventions are being developed to support self-management for children and young people (CYP) with sickle cell disease (SCD), but no systematic review has systematically synthesized this evidence regarding their characteristics, effectiveness, acceptability and feasibility for all published intervention types. METHODS The Joanna Briggs Institute guidelines for mixed-method reviews were followed. A systematic search of eight databases and key journals was conducted from their inception to November 2021. Primary research of self-management interventions targeting CYP with SCD aged 8-24 years and reporting any health/social outcome and acceptability data were included. Design-specific standardized critical appraisal instruments were used. Two independent reviewers screened and appraised the articles. A third reviewer resolved disagreements. RESULTS Of 1654 articles identified, 38 studies were included. Methodological quality was moderate. Most studies evaluated SCD education, psycho-behavioural, psychosocial and skills training and/or social support interventions. They appear to demonstrate short-term improvements in knowledge, social functioning and medical adherence outcomes. Interventions that were multifaceted in content, combined technological platforms and in-person group-based formats and involved peers, family and care providers were more acceptable and effective. The long-term impact of interventions was limited, including CYP's involvement in the intervention development and implementation. CONCLUSIONS There is inconclusive evidence for any self-management programme. Nonetheless, support from family, peers and care providers appears to be important for self-management interventions' effectiveness and acceptability. Future research needs to prioritize CYP involvement in both intervention design and delivery, their wider social context and include CYP with SCD from non-Black backgrounds. PATIENT AND PUBLIC CONTRIBUTION Three young people with SCD recruited acted as the review advisors. They were formally trained in the review process and involved in every aspect of the review: the design, conduct and interpretation of the findings. CYP involvement in the interventions' development and implementation was analysed as part of the review. This systematic review was conducted as part of a wider research project titled: Understanding fatigue experiences of CYP with SCD to guide the co-development of a fatigue self-management intervention. Two of the young advisors involved in the review were also involved in the development of the project funding application.
Collapse
Affiliation(s)
- Brenda A Poku
- Division of Nursing, Midwifery and Social Work, School of Health Sciences, University of Manchester, Manchester, UK
| | | | - Sue Kirk
- Division of Nursing, Midwifery and Social Work, School of Health Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
157
|
Lu HY, Orkin SH, Sankaran VG. Fetal Hemoglobin Regulation in Beta-Thalassemia. Hematol Oncol Clin North Am 2023; 37:301-312. [PMID: 36907604 DOI: 10.1016/j.hoc.2022.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
β-thalassemia is caused by mutations that reduce β-globin production, causing globin chain imbalance, ineffective erythropoiesis, and consequent anemia. Increased fetal hemoglobin (HbF) levels can ameliorate the severity of β-thalassemia by compensating for the globin chain imbalance. Careful clinical observations paired with population studies and advances in human genetics have enabled the discovery of major regulators of HbF switching (i.e. BCL11A, ZBTB7A) and led to pharmacological and genetic therapies for treating β-thalassemia patients. Recent functional screens using genome editing and other emerging tools have identified many new HbF regulators, which may improve therapeutic HbF induction in the future.
Collapse
Affiliation(s)
- Henry Y Lu
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Karp Family Research Laboratories, Boston Children's Hospital, 1 Blackfan Street, Boston, MA 02115, USA. https://twitter.com/realhenrylu
| | - Stuart H Orkin
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Karp Family Research Laboratories, Boston Children's Hospital, 1 Blackfan Street, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Karp Family Research Laboratories, Boston Children's Hospital, 1 Blackfan Street, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
158
|
Mohamed Jiffry MZ, Hassan R, Davis A, Scharf S, Walgamage T, Ahmed-Khan MA, Dandwani M. Sickle Cell Anemia Associated With Increased In-Hospital Mortality in Post-Cardiac Arrest Patients. Cureus 2023; 15:e37987. [PMID: 37223169 PMCID: PMC10202522 DOI: 10.7759/cureus.37987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2023] [Indexed: 05/25/2023] Open
Abstract
Introduction Sickle cell anemia (SCA) is a hemoglobinopathy that arises from a point mutation in the beta-globin gene, which causes the polymerization of deoxygenated hemoglobin that leads to a wide variety of clinical complications. Deaths in patients with SCA most commonly arise from renal, cardiovascular disease, infections, and stroke. In-hospital cardiac arrest has been found to be more common in older patients and those on ventilatory life support, among others. This study aims to provide more insight into how SCA affects the risk of in-hospital mortality in post-cardiac arrest patients. Methods The National Inpatient Survey database years 2016 to 2019 was utilized. The International Classification of Diseases, Tenth Revision, Procedure Coding System (ICD-10 PCS) codes for cardiopulmonary resuscitation were used to identify in-hospital cardiac arrest (IHCA) patients. ICD-10 Clinical Modification (CM) codes were used to identify SCA and other medical comorbidities. Categorical data was compared using Person's chi-square test, and continuous variables were compared using the independent samples t-test. Multinomial logistic regression was used to study the effects of SCA on post-arrest in-hospital mortality controlling for age, Charlson comorbidity score, and demographic variables. Binomial logistic regression models for dichotomous variables were utilized in the subgroup and secondary outcomes analysis. Results In patients with IHCA, patients who had SCA were found to have a significantly increased risk of in-hospital mortality adjusted for baseline characteristics and Charlson comorbidity score (OR: 1.16, 95% CI: 1.02-1.32, p=0.0025). Patient characteristics most strongly associated with an increased risk of in-hospital mortality in this cohort were found to be Black race (OR: 1.92, 95% CI: 1.87-1.97, p<0.001) and self-payer status (OR: 2.14, 95% CI: 2.06-2.22, p<0.001). Subgroup analysis revealed only patients with sickle cell disease had a statistically significant increased risk of in-hospital mortality in this cohort (OR: 4.41, 95% CI: 3.5-5.55, p<0.001), and patients with sickle cell trait did not. Conclusion In patients with IHCA, SCA is associated with an increased risk of in-hospital mortality. This risk was confined to patients with sickle cell disease and not patients with sickle cell trait.
Collapse
Affiliation(s)
| | - Rehana Hassan
- School of Medicine, American University of the Caribbean, Cupecoy, SXM
| | - Alexis Davis
- School of Medicine, American University of the Caribbean, Cupecoy, SXM
| | - Shelbie Scharf
- School of Medicine, American University of the Caribbean, Cupecoy, SXM
| | | | - Mohammad A Ahmed-Khan
- Department of Internal Medicine, University of Vermont, Burlington, USA
- Internal Medicine, Danbury Hospital, Danbury, USA
| | | |
Collapse
|
159
|
Sadeghi A, Taherifard E, Dehdari Ebrahimi N, Rafiei E, Hadianfard F, Taherifard E. Effects of l-arginine supplementation in patients with sickle cell disease: A systematic review and meta-analysis of clinical trials. Health Sci Rep 2023; 6:e1167. [PMID: 37064309 PMCID: PMC10090802 DOI: 10.1002/hsr2.1167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 04/18/2023] Open
Abstract
Background and Aims Previous studies have shown that supplementation of some amino acids such as l-arginine or its precursors could exert beneficial effects in patients with sickle cell disease (SCD). The objective of this study is to systematically review the literature to assess the effect of arginine administration on the clinical and paraclinical parameters of patients with SCD. Methods Four online databases of PubMed, Web of Sciences, Scopus, and Embase were selected for systematic search. Eligible studies were clinical trials that evaluated the effect of arginine usage in patients with SCD. Effects sizes were calculated using weighted mean difference (WMD) and Hedge's g and they were pooled using random-effects modeling with Hartung-Knapp adjustment. Additional analyses were also conducted. Results Twelve studies containing detail of 399 patients with SCD were found to be eligible. The data synthesis showed that l-arginine significantly increased the level of NO metabolites (Hedge's g: 1.50, 0.48-1.82, I 2: 88%) and hemoglobin F (WMD: 1.69%, 0.86-2.52, I 2: 0%) and significantly decreased systolic blood pressure (WMD: -8.46 mmHg, -15.58 to -1.33, I 2: 53%) and aspartate transaminase (Hedge's g: -0.49, -0.73 to -0.26, I 2: 0%). However, there were no significant effects on hemoglobin, reticulocyte, malondialdehyde and diastolic blood pressure, and alanine transaminase. Conclusion Our meta-analysis showed that l-arginine use for SCD could be beneficial, increase hemoglobin F and exert blood pressure-lowering and hepatoprotective properties. However, for a firm conclusion and widespread use of l-arginine for these patients, more studies are needed.
Collapse
Affiliation(s)
- Alireza Sadeghi
- Student Research CommitteeShiraz University of Medical SciencesShirazIran
| | - Ehsan Taherifard
- Internal Medicine DepartmentShiraz University of Medical SciencesShirazIran
| | | | - Elham Rafiei
- Student Research CommitteeShiraz University of Medical SciencesShirazIran
| | - Farshad Hadianfard
- Student Research CommitteeShiraz University of Medical SciencesShirazIran
| | - Erfan Taherifard
- Internal Medicine DepartmentShiraz University of Medical SciencesShirazIran
| |
Collapse
|
160
|
Oyedeji CI, Hodulik KL, Telen MJ, Strouse JJ. Management of Older Adults with Sickle Cell Disease: Considerations for Current and Emerging Therapies. Drugs Aging 2023; 40:317-334. [PMID: 36853587 PMCID: PMC10979738 DOI: 10.1007/s40266-023-01014-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/01/2023]
Abstract
People with sickle cell disease (SCD) are living longer than ever before, with the median survival increasing from age 14 years in 1973, beyond age 40 years in the 1990s, and as high as 61 years in recent cohorts from academic centers. Improvements in survival have been attributed to initiatives, such as newborn screening, penicillin prophylaxis, vaccination against encapsulated organisms, better detection and treatment of splenic sequestration, and improved transfusion support. There are an estimated 100,000 people living with SCD in the United States and millions of people with SCD globally. Given that the number of older adults with SCD will likely continue to increase as survival improves, better evidence on how to manage this population is needed. When managing older adults with SCD (defined herein as age ≥ 40 years), healthcare providers should consider the potential pitfalls of extrapolating evidence from existing studies on current and emerging therapies that have typically been conducted with participants at mean ages far below 40 years. Older adults with SCD have historically had little to no representation in clinical trials; therefore, more guidance is needed on how to use current and emerging therapies in this population. This article summarizes the available evidence for managing older adults with SCD and discusses potential challenges to using approved and emerging drugs in this population.
Collapse
Affiliation(s)
- Charity I Oyedeji
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Duke Claude D. Pepper Older Americans Independence Center, Durham, NC, USA.
- Department of Medicine, and Duke Comprehensive Sickle Cell Center, Duke University School of Medicine, 315 Trent Dr., Suite 266, DUMC Box 3939, Durham, NC, 27710, USA.
| | - Kimberly L Hodulik
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Department of Pharmacy, Duke University Hospital, Durham, NC, USA
| | - Marilyn J Telen
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - John J Strouse
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Claude D. Pepper Older Americans Independence Center, Durham, NC, USA
- Department of Medicine, and Duke Comprehensive Sickle Cell Center, Duke University School of Medicine, 315 Trent Dr., Suite 266, DUMC Box 3939, Durham, NC, 27710, USA
- Division of Pediatric Hematology-Oncology, Duke University, Durham, NC, USA
| |
Collapse
|
161
|
Sesti-Costa R, Costa FF, Conran N. Role of Macrophages in Sickle Cell Disease Erythrophagocytosis and Erythropoiesis. Int J Mol Sci 2023; 24:ijms24076333. [PMID: 37047304 PMCID: PMC10094208 DOI: 10.3390/ijms24076333] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited blood disorder caused by a β-globin gene point mutation that results in the production of sickle hemoglobin that polymerizes upon deoxygenation, causing the sickling of red blood cells (RBCs). RBC deformation initiates a sequence of events leading to multiple complications, such as hemolytic anemia, vaso-occlusion, chronic inflammation, and tissue damage. Macrophages participate in extravascular hemolysis by removing damaged RBCs, hence preventing the release of free hemoglobin and heme, and triggering inflammation. Upon erythrophagocytosis, macrophages metabolize RBC-derived hemoglobin, activating mechanisms responsible for recycling iron, which is then used for the generation of new RBCs to try to compensate for anemia. In the bone marrow, macrophages can create specialized niches, known as erythroblastic islands (EBIs), which regulate erythropoiesis. Anemia and inflammation present in SCD may trigger mechanisms of stress erythropoiesis, intensifying RBC generation by expanding the number of EBIs in the bone marrow and creating new ones in extramedullary sites. In the current review, we discuss the distinct mechanisms that could induce stress erythropoiesis in SCD, potentially shifting the macrophage phenotype to an inflammatory profile, and changing their supporting role necessary for the proliferation and differentiation of erythroid cells in the disease. The knowledge of the soluble factors, cell surface and intracellular molecules expressed by EBI macrophages that contribute to begin and end the RBC’s lifespan, as well as the understanding of their signaling pathways in SCD, may reveal potential targets to control the pathophysiology of the disease.
Collapse
|
162
|
Diamantopoulos PT, Anastasopoulou A, Dimopoulou M, Samarkos M, Gogas H. Challenges in the treatment of melanoma with BRAF and MEK inhibitors in patients with sickle cell disease: case report and review of the literature. Ther Adv Hematol 2023; 14:20406207231155991. [PMID: 36936358 PMCID: PMC10021083 DOI: 10.1177/20406207231155991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/23/2023] [Indexed: 03/17/2023] Open
Abstract
Patients with sickle cell disease (SCD) suffer from complications due to anemia, inflammation, and vaso-occlusion. Factors that trigger sickling and/or inflammation may initiate such complications, while treatment with hydroxyurea (HU) reduces their emergence and prolongs survival. On the contrary, inhibition of the BRAF-MEK-ERK pathway with BRAF and MEK inhibitors (BRAF/MEKi) has revolutionized treatment of melanoma but their use has been correlated with inflammatory adverse events. Thus, treatment of patients with SCD with BRAF/MEKi may be quite challenging and pyrexia in those patients should be managed as a medical emergency. In this article, intrigued by the case of a 36-year-old female patient with S/β-thal under HU who was treated with dabrafenib and trametinib for melanoma, we analyze the mechanisms underlying inflammation and vaso-occlusion in SCD, the mechanisms of pyrexia and inflammation induced by BRAF/MEKi, their potential interconnections, the shared role of the inflammasome in these two entities, and the protective effect of HU in SCD. Since SCD is the most common inheritable blood disorder, the administration of BRAF/MEKi for melanoma in patients with SCD may be a rather common challenge. Thus, proper treatment with HU may pave the way for an uneventful management of such patients.
Collapse
Affiliation(s)
- Panagiotis T Diamantopoulos
- First Department of Internal Medicine, Laikon
General Hospital, National and Kapodistrian University of Athens, 17 Agiou
Thoma street, Goudi, 11527, Athens, Greece
| | - Amalia Anastasopoulou
- First Department of Internal Medicine, Laikon
General Hospital, National and Kapodistrian University of Athens, Athens,
Greece
| | | | - Michalis Samarkos
- First Department of Internal Medicine, Laikon
General Hospital, National and Kapodistrian University of Athens, Athens,
Greece
| | - Helen Gogas
- First Department of Internal Medicine, Laikon
General Hospital, National and Kapodistrian University of Athens, Athens,
Greece
| |
Collapse
|
163
|
Peslak SA, Demirci S, Chandra V, Ryu B, Bhardwaj SK, Jiang J, Rupon JW, Throm RE, Uchida N, Leonard A, Essawi K, Bonifacino AC, Krouse AE, Linde NS, Donahue RE, Ferrara F, Wielgosz M, Abdulmalik O, Hamagami N, Germino-Watnick P, Le A, Chu R, Hinds M, Weiss MJ, Tong W, Tisdale JF, Blobel GA. Forced enhancer-promoter rewiring to alter gene expression in animal models. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:452-465. [PMID: 36852088 PMCID: PMC9958407 DOI: 10.1016/j.omtn.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023]
Abstract
Transcriptional enhancers can be in physical proximity of their target genes via chromatin looping. The enhancer at the β-globin locus (locus control region [LCR]) contacts the fetal-type (HBG) and adult-type (HBB) β-globin genes during corresponding developmental stages. We have demonstrated previously that forcing proximity between the LCR and HBG genes in cultured adult-stage erythroid cells can activate HBG transcription. Activation of HBG expression in erythroid cells is of benefit to patients with sickle cell disease. Here, using the β-globin locus as a model, we provide proof of concept at the organismal level that forced enhancer rewiring might present a strategy to alter gene expression for therapeutic purposes. Hematopoietic stem and progenitor cells (HSPCs) from mice bearing human β-globin genes were transduced with lentiviral vectors expressing a synthetic transcription factor (ZF-Ldb1) that fosters LCR-HBG contacts. When engrafted into host animals, HSPCs gave rise to adult-type erythroid cells with elevated HBG expression. Vectors containing ZF-Ldb1 were optimized for activity in cultured human and rhesus macaque erythroid cells. Upon transplantation into rhesus macaques, erythroid cells from HSPCs expressing ZF-Ldb1 displayed elevated HBG production. These findings in two animal models suggest that forced redirection of gene-regulatory elements may be used to alter gene expression to treat disease.
Collapse
Affiliation(s)
- Scott A. Peslak
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Vemika Chandra
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Byoung Ryu
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Saurabh K. Bhardwaj
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jing Jiang
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Jeremy W. Rupon
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Robert E. Throm
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Khaled Essawi
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
- Department of Medical Laboratory Science, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | | | - Allen E. Krouse
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Nathaniel S. Linde
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Robert E. Donahue
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Francesca Ferrara
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Matthew Wielgosz
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Osheiza Abdulmalik
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicole Hamagami
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paula Germino-Watnick
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anh Le
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Rebecca Chu
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Malikiya Hinds
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mitchell J. Weiss
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Wei Tong
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - John F. Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Gerd A. Blobel
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
164
|
Segura EER, Ayoub PG, Hart KL, Kohn DB. Gene Therapy for β-Hemoglobinopathies: From Discovery to Clinical Trials. Viruses 2023; 15:713. [PMID: 36992422 PMCID: PMC10054523 DOI: 10.3390/v15030713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Investigations to understand the function and control of the globin genes have led to some of the most exciting molecular discoveries and biomedical breakthroughs of the 20th and 21st centuries. Extensive characterization of the globin gene locus, accompanied by pioneering work on the utilization of viruses as human gene delivery tools in human hematopoietic stem and progenitor cells (HPSCs), has led to transformative and successful therapies via autologous hematopoietic stem-cell transplant with gene therapy (HSCT-GT). Due to the advanced understanding of the β-globin gene cluster, the first diseases considered for autologous HSCT-GT were two prevalent β-hemoglobinopathies: sickle cell disease and β-thalassemia, both affecting functional β-globin chains and leading to substantial morbidity. Both conditions are suitable for allogeneic HSCT; however, this therapy comes with serious risks and is most effective using an HLA-matched family donor (which is not available for most patients) to obtain optimal therapeutic and safe benefits. Transplants from unrelated or haplo-identical donors carry higher risks, although they are progressively improving. Conversely, HSCT-GT utilizes the patient's own HSPCs, broadening access to more patients. Several gene therapy clinical trials have been reported to have achieved significant disease improvement, and more are underway. Based on the safety and the therapeutic success of autologous HSCT-GT, the U.S. Food and Drug Administration (FDA) in 2022 approved an HSCT-GT for β-thalassemia (Zynteglo™). This review illuminates the β-globin gene research journey, adversities faced, and achievements reached; it highlights important molecular and genetic findings of the β-globin locus, describes the predominant globin vectors, and concludes by describing promising results from clinical trials for both sickle cell disease and β-thalassemia.
Collapse
Affiliation(s)
- Eva Eugenie Rose Segura
- Molecular Biology Interdepartmental Doctoral Program, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| | - Paul George Ayoub
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Kevyn Lopez Hart
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Donald Barry Kohn
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Pediatrics (Hematology/Oncology), David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center for Stem Cell Research and Regenerative Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
165
|
Belisário AR, Simões E Silva AC, Moura ICG, Carneiro-Proietti AB, Sabino EC, Loureiro P, Máximo C, Flor-Park MV, Rodrigues DDOW, Ozahata MC, Mota RA, Dinardo CL, Kelly S, Custer B. Estimated glomerular filtration rate in Brazilian adults with sickle cell disease: results from the REDS-III multicenter cohort study. Ann Hematol 2023; 102:1019-1027. [PMID: 36884065 DOI: 10.1007/s00277-023-05150-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 02/25/2023] [Indexed: 03/09/2023]
Abstract
Chronic kidney disease (CKD) has a significant impact on sickle cell disease (SCD) morbidity and mortality. Early identification of individuals at highest risk of developing CKD may allow therapeutic intervention to prevent worse outcomes. This study aimed to evaluate the prevalence and risk factors for reduced estimated glomerular filtration rate (eGFR) among adults with SCD in Brazil. Participants in the REDS-III multicenter SCD cohort with more severe genotypes aged ≥ 18 years with at least two serum creatinine values were analyzed. The eGFR was calculated using the Jamaica Sickle Cell Cohort Study GFR equation. The eGFR categories were defined according to the K/DOQI. Participants with eGFR ≥ 90 were compared to those with those with eGFR < 90. Among the 870 participants, 647 (74.4%) had eGFR ≥ 90, 211 (24.3%) had eGFR 60 to 89, six (0.7%) had eGFR 30 to 59, and six (0.7%) had ESRD. Male sex (OR: 37.3; 95%CI: 22.4-65.1), higher age (OR: 1.04; 95%CI: 1.02-1.06), higher diastolic blood pressure (OR: 1.03; 95%CI: 1.009-1.06), lower Hb (OR: 0.80; 95%CI: 0.68-0.93), and lower reticulocytes (OR: 0.94; 95%CI: 0.89-0.99) levels were independently associated with eGFR < 90. There was a trend towards higher odds of death in participants with eGFR < 90 (OR: 1.8; 95%CI: 0.95-3.32; p = 0.065). In turn, participants with eGFR < 60 had a 12.2 (95%CI: 2.1-96.9) times higher odds for death when compared to those with eGFR ≥ 60. In this study, eGFR < 90 was observed in one-quarter of adults. Older age, male sex, higher diastolic blood pressure, lower hemoglobin, and lower reticulocyte levels were associated with occurrence of eGFR < 90. Estimated GFR < 60 increased the risk of mortality.
Collapse
Affiliation(s)
- André Rolim Belisário
- Centro de Tecidos Biológicos de Minas Gerais, Fundação Hemominas, Rua das Goiabeiras, 779, Lagoa Santa, MG, 33400-000, Brazil. .,Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Ana Cristina Simões E Silva
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Anna Bárbara Carneiro-Proietti
- Centro de Tecidos Biológicos de Minas Gerais, Fundação Hemominas, Rua das Goiabeiras, 779, Lagoa Santa, MG, 33400-000, Brazil
| | - Ester Cerdeira Sabino
- Faculdade de Medicina (FMUSP) and Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
| | - Paula Loureiro
- Fundação Hemope, Recife, PE, Brazil.,Universidade de Pernambuco, Recife, PE, Brazil
| | | | - Miriam V Flor-Park
- ITACI, Unidade de Onco-Hematologia, Instituto da Criança, HCFMUSP, São Paulo, Brazil
| | | | - Mina Cintho Ozahata
- Department of Computer Science - DCC, University of São Paulo, São Paulo, Brazil
| | - Rosimere Afonso Mota
- Centro de Tecidos Biológicos de Minas Gerais, Fundação Hemominas, Rua das Goiabeiras, 779, Lagoa Santa, MG, 33400-000, Brazil
| | | | | | - Brian Custer
- Vitalant Research Institute, San Francisco, CA, USA.,Department of Laboratory Medicine, UCSF, San Francisco, CA, USA
| | | |
Collapse
|
166
|
Emerging Strategies in Proteolysis-Targeting Chimeras (PROTACs): Highlights from 2022. Int J Mol Sci 2023; 24:ijms24065190. [PMID: 36982263 PMCID: PMC10049114 DOI: 10.3390/ijms24065190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Targeted protein degradation (TPD) is a promising therapeutic modality that has garnered attention in academic, industrial, and pharmaceutical research for treating diseases such as cancer, neurodegenerative disorders, inflammation, and viral infections. In this context, proteolysis-targeting chimeras (PROTACs) present a reliable technology for degrading disease-causing proteins. PROTACs complement small-molecule inhibitors, which primarily rely on direct protein regulation. From concept-to-clinic, PROTACs have evolved from cell impermeable peptide molecules to orally bioavailable drugs. Despite their potential in medicinal chemistry, certain aspects regarding PROTACs remain unclear. The clinical significance of PROTACs is primarily limited owing to their lack of selectivity and drug-like properties. This review focused on recently reported PROTAC strategies, particularly in 2022. It aimed to address and overcome the challenges posed by classical PROTACs by correlating them with emerging approaches with improved selectivity and controllability, cell permeability, linker flexibility, druggability, and PROTAC-based approaches, developed in 2022. Furthermore, recently reported PROTAC-based approaches are discussed, highlighting each of their advantages and limitations. We predict that several improved PROTAC molecules will be accessible for treating patients exhibiting various conditions, including cancer, neurodegenerative disorders, inflammation, and viral infections.
Collapse
|
167
|
In Humanized Sickle Cell Mice, Imatinib Protects Against Sickle Cell-Related Injury. Hemasphere 2023; 7:e848. [PMID: 36874380 PMCID: PMC9977487 DOI: 10.1097/hs9.0000000000000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/19/2023] [Indexed: 03/06/2023] Open
Abstract
Drug repurposing is a valuable strategy for rare diseases. Sickle cell disease (SCD) is a rare hereditary hemolytic anemia accompanied by acute and chronic painful episodes, most often in the context of vaso-occlusive crisis (VOC). Although progress in the knowledge of pathophysiology of SCD have allowed the development of new therapeutic options, a large fraction of patients still exhibits unmet therapeutic needs, with persistence of VOCs and chronic disease progression. Here, we show that imatinib, an oral tyrosine kinase inhibitor developed for the treatment of chronic myelogenous leukemia, acts as multimodal therapy targeting signal transduction pathways involved in the pathogenesis of both anemia and inflammatory vasculopathy of humanized murine model for SCD. In addition, imatinib inhibits the platelet-derived growth factor-B-dependent pathway, interfering with the profibrotic response to hypoxia/reperfusion injury, used to mimic acute VOCs. Our data indicate that imatinib might be considered as possible new therapeutic tool for chronic treatment of SCD.
Collapse
|
168
|
Hoyt CR, Hurwitz S, Varughese TE, Yaeger LH, King AA. Individual-level behavioral interventions to support optimal development of children with sickle cell disease: A systematic review. Pediatr Blood Cancer 2023; 70:e30178. [PMID: 36583467 PMCID: PMC10416609 DOI: 10.1002/pbc.30178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/15/2022] [Accepted: 12/05/2022] [Indexed: 12/31/2022]
Abstract
This review aimed to identify and describe individual-level behavioral interventions for children 0-18 years of age with sickle cell disease (SCD). PRISMA guidelines were followed at each stage of this review. Twenty-seven studies were included, representing six intervention types: disease knowledge (n = 7), self-management (n = 7), pain management (n = 4), school functioning (n = 4), cognitive health (n = 4), and mental health (n = 2). Most interventions targeted older children (5+ years), while only two examined interventions for children 0-3 years. This review suggests that offering education about disease knowledge, self-management, and pain management interventions can be beneficial for this population. Future research is needed to understand interventions to support young children and the impact of SCD on development.
Collapse
Affiliation(s)
- Catherine R Hoyt
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sadie Hurwitz
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Taniya E Varughese
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lauren H Yaeger
- Becker Medical Library, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Allison A King
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Education, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
169
|
Pincez T, Lo KS, D'Orengiani ALPHD, Garrett ME, Brugnara C, Ashley-Koch AE, Telen MJ, Galacteros F, Joly P, Bartolucci P, Lettre G. Variation and impact of polygenic hematologic traits in monogenic sickle cell disease. Haematologica 2023; 108:870-881. [PMID: 36226494 PMCID: PMC9973495 DOI: 10.3324/haematol.2022.281180] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
Several of the complications observed in sickle cell disease (SCD) are influenced by variation in hematologic traits (HT), such as fetal hemoglobin (HbF) level and neutrophil count. Previous large-scale genome-wide association studies carried out in largely healthy individuals have identified thousands of variants associated with HT, which have then been used to develop multi-ancestry polygenic trait scores (PTS). Here, we tested whether these PTS associate with HT in SCD patients and if they can improve statistical models associated with SCD-related complications. In 2,056 SCD patients, we found that the PTS predicted less HT variance than in non-SCD individuals of African ancestry. This was particularly striking at the Duffy/DARC locus, where we observed an epistatic interaction between the SCD genotype and the Duffy null variant (rs2814778) that led to a two-fold weaker effect on neutrophil count. PTS for these HT which are measured as part of routine practice were not associated with complications in SCD. In contrast, we found that a simple PTS for HbF that includes only six variants explained a large fraction of the phenotypic variation (20.5-27.1%), associated with acute chest syndrome and stroke risk, and improved the statistical modeling of the vaso-occlusive crisis rate. Using Mendelian randomization, we found that increasing HbF by 4.8% reduces stroke risk by 39% (P=0.0006). Taken together, our results highlight the importance of validating PTS in large diseased populations before proposing their implementation in the context of precision medicine initiatives.
Collapse
Affiliation(s)
- Thomas Pincez
- Montreal Heart Institute, Montreal, Quebec, Canada; Department of Pediatrics, Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, Universite de Montreal, Montreal, Quebec
| | - Ken Sin Lo
- Montreal Heart Institute, Montreal, Quebec
| | | | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC
| | - Carlo Brugnara
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA
| | | | - Marilyn J Telen
- Department of Medicine, Division of Hematology, Duke University Medical Center, Durham, NC
| | - Frederic Galacteros
- Red Cell Genetic Disease Unit, Hopital Henri-Mondor, Assistance Publique-Hopitaux de Paris (AP-HP), Universite Paris Est, IMRB - U955 - Equipe no 2, Creteil
| | - Philippe Joly
- Unite Fonctionnelle 34445 'Biochimie des Pathologies Erythrocytaires', Laboratoire de Biochimie et Biologie Moleculaire Grand-Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France; Laboratoire Inter-Universitaire de Biologie de la Motricite (LIBM) EA7424, Equipe 'Biologie Vasculaire et du Globule Rouge', Universite Claude Bernard Lyon 1, Comite d'Universites et d'Etablissements (COMUE), Lyon
| | - Pablo Bartolucci
- Red Cell Genetic Disease Unit, Hopital Henri-Mondor, Assistance Publique-Hopitaux de Paris (AP-HP), Universite Paris Est, IMRB - U955 - Equipe no 2, Creteil
| | - Guillaume Lettre
- Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Faculty of Medicine, Universite de Montreal, Montreal, Quebec.
| |
Collapse
|
170
|
Kardynska M, Kogut D, Pacholczyk M, Smieja J. Mathematical modeling of regulatory networks of intracellular processes - Aims and selected methods. Comput Struct Biotechnol J 2023; 21:1523-1532. [PMID: 36851915 PMCID: PMC9958294 DOI: 10.1016/j.csbj.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Regulatory networks structure and signaling pathways dynamics are uncovered in time- and resource consuming experimental work. However, it is increasingly supported by modeling, analytical and computational techniques as well as discrete mathematics and artificial intelligence applied to to extract knowledge from existing databases. This review is focused on mathematical modeling used to analyze dynamics and robustness of these networks. This paper presents a review of selected modeling methods that facilitate advances in molecular biology.
Collapse
Affiliation(s)
- Malgorzata Kardynska
- Dept. of Biosensors and Processing of Biomedical Signals, Silesian University of Technology, Gliwice, Poland
| | - Daria Kogut
- Dept. of Biosensors and Processing of Biomedical Signals, Silesian University of Technology, Gliwice, Poland.,Dept. of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Marcin Pacholczyk
- Dept. of Biosensors and Processing of Biomedical Signals, Silesian University of Technology, Gliwice, Poland.,Dept. of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Jaroslaw Smieja
- Dept. of Biosensors and Processing of Biomedical Signals, Silesian University of Technology, Gliwice, Poland.,Dept. of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
171
|
Abstract
While neutrophils are the main effectors of protective innate immune responses, they are also key players in inflammatory pathologies. Sickle cell disease (SCD) is a genetic blood disorder in which red blood cells (RBCs) are constantly destroyed in the circulation which generates a highly inflammatory environment that culminates in vascular occlusions. Vaso-occlusion is the hallmark of SCD and a predictor of disease severity. Neutrophils initiate and propagate SCD-related vaso-occlusion through adhesive interactions with the activated and dysfunctional endothelium, sickle RBCs, and platelets, leading to acute and chronic complications that progress to irreversible organ damage and ultimately death. The use of SCD humanized mouse models, in combination with in vivo imaging techniques, has emerged as a fundamental tool to understand the dynamics of neutrophils under complex inflammatory contexts and their contribution to vascular injury in SCD. In this review, we discuss the various mechanisms by which circulating neutrophils sense and respond to the wide range of stimuli present in the blood of SCD patients and mice. We argue that the central role of neutrophils in SCD can be rationalized to develop targets for the management of clinical complications in SCD patients.
Collapse
Affiliation(s)
- Lidiane S Torres
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
172
|
Agouti I, Masson E, Loundou A, Jean E, Arnaud L, Abdili E, Berenger P, Lavoipierre V, Séguier J, Dignat-George F, Lacroix R, Bernit E. Plasma levels of E-selectin are associated with retinopathy in sickle cell disease. Eur J Haematol 2023; 110:271-279. [PMID: 36409296 PMCID: PMC10100354 DOI: 10.1111/ejh.13902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND The vascular endothelium is markedly disrupted in sickle cell disease (SCD) and is the converging cascade of the complex pathophysiologic processes linked to sickle cell vasculopathy. Circulating endothelial activation and/or apoptotic markers may reflect this endothelial activation/damage that contributes to the pathophysiology of the SCD vascular complications. METHODS Plasmatic levels of circulating endothelial cells (CECs), E-selectin, progenitor's endothelial cells (EPCs), and circulating extracellular vesicles (EVs) were evaluated in 50 SCD patients, 16 with vasculopathy. The association between these markers and the occurrence of disease-related microvascular injuries of the eye (retinopathy), kidney (nephropathy), and skin (chronic active ulcers) was explored. RESULTS Among the endothelial activation markers studied, only higher plasma levels of E-selectin were found in SCD patients with vasculopathy (p = .015). Increased E-selectin levels were associated with retinopathy (p < .001) but not with nephropathy or leg ulcers. All patients, at steady state, with or without vasculopathy, did not display a high count of CEC and EPC, markers of endothelial injury and repair. We did not show any significant differences in EVs levels between vasculopathy and not vasculopathy SCD patients. CONCLUSIONS Further studies will be required to determine whether the E-selectin could be used as an early biomarker of retinopathy sickle cell development.
Collapse
Affiliation(s)
- Imane Agouti
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Elodie Masson
- Département de médecine interne, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Anderson Loundou
- Centre d'Etudes et de Recherche sur les services de santé et la qualité de vie. Unité de recherche EA 3279. Faculté de médecine, université Aix Marseille, Marseille, France
| | - Estelle Jean
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,Département de médecine interne, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Laurent Arnaud
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Evelyne Abdili
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Patricia Berenger
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Virginie Lavoipierre
- Département de médecine interne, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Julie Séguier
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,Département de médecine interne, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Françoise Dignat-George
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,C2VN, INSERM, INRAE, université Aix Marseille, Marseille, France
| | - Romaric Lacroix
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,C2VN, INSERM, INRAE, université Aix Marseille, Marseille, France
| | - Emmanuelle Bernit
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,Unité transversale de la drépanocytose, centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, CHU de la Guadeloupe, Guadeloupe, France
| |
Collapse
|
173
|
Samaee S, Samaee S, Mihalca D, Fitzgerald L, Ahmed A, Hall J, Tsitsikas DA. Mortality Rates and autopsy findings in fat embolism syndrome complicating sickle cell disease. J Clin Pathol 2023:jcp-2023-208763. [PMID: 36849230 DOI: 10.1136/jcp-2023-208763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/02/2023] [Indexed: 03/01/2023]
Abstract
Fat embolism syndrome is a rare but underdiagnosed complication of sickle cell disease associated with high morbidity and mortality. It affects predominantly patients with a previously mild course of their illness and those of non-SS genotypes while there is possibly an association with infection with human parvovirus B19 (HPV B19). Here, we present the mortality rates and autopsy findings of all reported cases to date. A systematic review has revealed 99 published cases in the world literature with a mortality rate of 46%. Mortality varied greatly according to the time of reported cases with no survivors in the 1940s, 1950s or 1960s and no deaths since 2020. 35% of cases had previously undiagnosed sickle cell disease and the latter was only identified at autopsy after developing fat embolism with a fatal outcome. 20% of cases reported after 1986 tested positive for HPV B19 with an associated mortality of 63% whereas in cases that have not documented HPV B19 infection the mortality was 32%. The organs most often staining positive for fat were the kidneys, lungs, brain and heart whereas ectopic haematopoietic tissue was found in 45% of the examined lung specimens.
Collapse
Affiliation(s)
- Sayna Samaee
- Haematology, Homerton Healthcare NHS Foundation Trust, London, UK
| | - Sepideh Samaee
- Haematology, Homerton Healthcare NHS Foundation Trust, London, UK
| | - Diana Mihalca
- Haematology, Homerton Healthcare NHS Foundation Trust, London, UK
| | | | - Adeel Ahmed
- Haematology, Homerton Healthcare NHS Foundation Trust, London, UK
| | - John Hall
- Haematology, Homerton Healthcare NHS Foundation Trust, London, UK
| | | |
Collapse
|
174
|
Couette M, Forté S, Oudin Doglioni D, Mekontso-Dessap A, Calvet D, Kuo KHM, Bartolucci P. Early Strokes Are Associated with More Global Cognitive Deficits in Adults with Sickle Cell Disease. J Clin Med 2023; 12:1615. [PMID: 36836150 PMCID: PMC9967394 DOI: 10.3390/jcm12041615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
This study sought to link neurocognitive profiles in sickle cell disease (SCD) patients with clinical characteristics. We conducted a prospective cohort study of adults with SCD who underwent comprehensive neuropsychological assessment at the UMGGR clinic at Henri Mondor Hospital, Créteil (France). A cluster analysis was performed based on neuropsychological testing scores. The association between clusters and clinical profiles was assessed. Between 2017 and 2021, 79 patients with a mean age of 36 [range 19-65] years were included. On principal component analysis, a 5-factor model presented the best fit (Bartlett's sphericity test [χ2 (171) = 1345; p < 0.001]), explaining 72% of the variance. The factors represent distinct cognitive domains and anatomical regions. On hierarchical classification, three clusters emerged. Cluster 1 (n = 24) presented deficits in all five factors compared to Cluster 3 (n = 33). Cluster 2 (n = 22) had deficits in all factors, but to a lesser extent than Cluster 1. MoCA scores mirrored the severity of these cognitive deficits. Age, genotype and stroke prevalence did not differ significantly between clusters. However, the time of first stroke occurrence differed significantly between Cluster 1 and 2-3: 78% of strokes occurred during childhood, whereas 80% and 83% occurred during adulthood in Clusters 2 and 3, respectively. Educational attainment was also reduced in Cluster 1. SCD patients with childhood stroke seem to be at increased risk of a global cognitive deficit profile. In addition to existing methods of primary and secondary stroke prevention, early neurorehabilitation should be prioritized in order to reduce the long-term cognitive morbidity of SCD.
Collapse
Affiliation(s)
- Maryline Couette
- Sickle Cell Referral Centre–UMGGR, University of Paris Est Créteil, Henri Mondor APHP, 94010 Créteil, France
- CARMAS (Cardiovascular and Respiratory Manifestations of Acute Lung Injury and Sepsis), University of Paris Est Créteil, 94010 Créteil, France
- IMRB, INSERM, University of Paris Est Créteil, 94010 Créteil, France
| | - Stéphanie Forté
- Department of Medecine, Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 0C1, Canada
- Department of Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada
| | - Damien Oudin Doglioni
- Sickle Cell Referral Centre–UMGGR, University of Paris Est Créteil, Henri Mondor APHP, 94010 Créteil, France
- Laboratoire Inter-Universitaire de Psychologie—Personnalité, Cognition, Changement Social (LIP/PC2S), Université Grenoble Alpes, 38058 Saint-Martin-d′Hères, France
| | - Armand Mekontso-Dessap
- CARMAS (Cardiovascular and Respiratory Manifestations of Acute Lung Injury and Sepsis), University of Paris Est Créteil, 94010 Créteil, France
- IMRB, INSERM, University of Paris Est Créteil, 94010 Créteil, France
| | - David Calvet
- INSERM, UMR 1266, Psychiatry and Neurosciences Institute of Paris, Paris-Descartes University, Department of Neurology and Stroke Unit, Sainte-Anne Hospital, 75014 Paris, France
| | - Kevin H. M. Kuo
- Department of Medicine, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Pablo Bartolucci
- Sickle Cell Referral Centre–UMGGR, University of Paris Est Créteil, Henri Mondor APHP, 94010 Créteil, France
- IMRB, INSERM, University of Paris Est Créteil, 94010 Créteil, France
- INSERM-U955, Equipe 2, Laboratoire d’Excellence, GRex, Institut Mondor, 94000 Créteil, France
| |
Collapse
|
175
|
Gbotosho OT, Gollamudi J, Hyacinth HI. The Role of Inflammation in The Cellular and Molecular Mechanisms of Cardiopulmonary Complications of Sickle Cell Disease. Biomolecules 2023; 13:381. [PMID: 36830749 PMCID: PMC9953727 DOI: 10.3390/biom13020381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Cardiopulmonary complications remain the major cause of mortality despite newer therapies and improvements in the lifespan of patients with sickle cell disease (SCD). Inflammation has been identified as a major risk modifier in the pathogenesis of SCD-associated cardiopulmonary complications in recent mechanistic and observational studies. In this review, we discuss recent cellular and molecular mechanisms of cardiopulmonary complications in SCD and summarize the most recent evidence from clinical and laboratory studies. We emphasize the role of inflammation in the onset and progression of these complications to better understand the underlying pathobiological processes. We also discuss future basic and translational research in addressing questions about the complex role of inflammation in the development of SCD cardiopulmonary complications, which may lead to promising therapies and reduce morbidity and mortality in this vulnerable population.
Collapse
Affiliation(s)
- Oluwabukola T. Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267-0525, USA
| | - Jahnavi Gollamudi
- Division of Hematology & Oncology, Department of Internal Medicine, 3125 Eden Avenue, ML 0562, Cincinnati, OH 45219-0562, USA
| | - Hyacinth I. Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267-0525, USA
| |
Collapse
|
176
|
Suwito BE, Adji AS, Widjaja JS, Angel SCS, Al Hajiri AZZ, Salamy NFW, Choirotussanijjah C. A Review of CRISPR Cas9 for SCA: Treatment Strategies and Could Target β-globin Gene and BCL11A Gene using CRISPR Cas9 Prevent the Patient from Sickle Cell Anemia? Open Access Maced J Med Sci 2023. [DOI: 10.3889/oamjms.2023.11435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND: Sickle cell anemia is a hereditary globin chain condition that leads to hemolysis and persistent organ damage. Chronic hemolytic anemia, severe acute and chronic pain, and end-organ destruction occur throughout the lifespan of sickle cell anemia. SCD is associated with a higher risk of mortality. Genome editing with CRISPR-associated regularly interspersed short palindromic repeats (CRISPR/Cas9) have therapeutic potential for sickle cell anemia thala.
AIM: This research aimed to see if using CRISPR/Cas9 to target β-globin gene is an effective therapeutic and if it has a long-term effect on Sickle Cell Anemia.
METHODS: The method used in this study summarizes the article by looking for keywords that have been determined in the title and abstract. The authors used official guidelines from Science Direct, PubMed, Google Scholar, and Journal Molecular Biology to select full-text articles published within the last decade, prioritizing searches within the past 10 years.
RESULTS: CRISPR/Cas9-mediated genome editing in clinical trials contributes to α-globin gene deletion correcting β-thalassemia through balanced α- and β-globin ratios and inhibiting disease progression.
CONCLUSION: HBB and BCL11A targeting by CRISPR/Cas9 deletion effectively inactivate BCL11A, a repressor of fetal hemoglobin production. However, further research is needed to determine its side effects and safety.
Collapse
|
177
|
Adigwe OP. Access to healthcare for people with sickle cell disease: Views of healthcare professionals on policies and practices. Mol Genet Genomic Med 2023; 11:e2142. [PMID: 36727575 PMCID: PMC10178791 DOI: 10.1002/mgg3.2142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/23/2022] [Accepted: 01/10/2023] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Sickle cell disease is a genetic disorder with its highest prevalence in Nigeria. The condition causes red blood cells to turn rigid, and consequently, results to several complications including organ damage. This study aimed at assessing views of health practitioners in Nigeria on policies and practices in the area of access to healthcare services for sickle cell disease. METHODS A cross-sectional study was undertaken amongst healthcare professionals in Nigeria. Data were collected using a self-administered questionnaire, and analyses were undertaken using Statistical Package for Social Sciences software. RESULTS A significant proportion of the participants (42.8%) disagreed that relevant legislative framework exists to facilitate optimal access to high-quality healthcare services for persons with sickle cell disorder in Nigeria. Two-thirds of the study cohort were of the opinion that public health surveillance towards sickle cell disease was suboptimal (61.2%). Also, more than three-quarters of the respondents (78.7%) indicated that the cost of managing sickle cell disease was not affordable to majority of affected Nigerians. CONCLUSION This study provides critical insights into access to healthcare services for sickle cell disease. As such, challenges preventing access to healthcare services for sickle cell patients which have been identified in this study can underpin the development of contextual policies to address them.
Collapse
Affiliation(s)
- Obi Peter Adigwe
- National Institute for Pharmaceutical Research and Development, Abuja, Nigeria
| |
Collapse
|
178
|
Abstract
Sickle cell disease (SCD) is the most-common monogenic recessive disease in humans, annually affecting almost 300,000 newborns worldwide, 75% of whom live in Africa. Genomics research can accelerate the development of curative therapies for SCD in three ways. First, research should explore the missing heritability of foetal haemoglobin (HbF) - the strongest known modifier of SCD clinical expression - among highly genetically heterogenous and understudied African populations, to provide novel therapeutics targets for HbF induction. Second, SCD research should invest in RNA therapies, either by using microRNA to target the production of HbF proteins by binding to the transcription machinery in a cell, or by directly mediating production of HbF or adult haemoglobin through injection of messenger RNA. Third, investigators should aim to identify currently unknown genetic risk factors for SCD cardiovascular complications, which will address mortality, particularly in adults. Now is the time for global research programs to uncover genomic keys to unlock SCD therapeutics.
Collapse
Affiliation(s)
- Ambroise Wonkam
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
179
|
Sy SKB, Tanaka C, Grosch K. Population Pharmacokinetics and Pharmacodynamics of Crizanlizumab in Healthy Subjects and Patients with Sickle Cell Disease. Clin Pharmacokinet 2023; 62:249-266. [PMID: 36529836 DOI: 10.1007/s40262-022-01193-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVES Crizanlizumab is a humanized monoclonal antibody against P-selectin for the prevention of vaso-occlusive crises in sickle cell disease (SCD). The objective of this study was to investigate crizanlizumab population pharmacokinetics (PK) and pharmacodynamics (PD), as well as influential covariates. METHODS A population PK model for crizanlizumab was developed from healthy volunteer and SCD patient data, using a two-compartment intravenous infusion model utilizing a target-mediated drug disposition (TMDD) approach. The relationship between crizanlizumab concentration and ex vivo P-selectin inhibition was fitted to a non-linear sigmoidal Emax model. Covariate selection was performed in a stepwise manner. RESULTS Crizanlizumab exhibits nonlinear pharmacokinetics in the wide dose range of 0.2-8 mg/kg body weight. The population pharmacokinetic base model incorporated body weight as covariate in the form of allometric scaling wherein the exponents were fixed to 0.8. SCD patients had higher baseline soluble P-selectin concentration, resulting in a higher estimated initial target concentration. The typical individual in the model is a 70 kg SCD patient with normal renal function and a baseline albumin of 43 g/L; CL was 0.012 L/h while Vss was 5.2 L. For the population PD model, none of the identified additional factors beyond PD assay and covariates, such as body weight at baseline nor patient type differences, led to relevant differences in P-selectin % inhibition. CONCLUSIONS Renal and hepatic impairments, concomitant hydroxyurea usage, and presence of anti-drug antibody are not expected to impact the exposure of crizanlizumab. The model allows for extrapolating the PK of crizanlizumab to pediatric population and evaluation of alternative regimens and route of administration. TRIAL REGISTRATION NUMBER [DATE OF REGISTRATION]: SUSTAIN (CSEG101A2201 Phase 2), ClinicalTrials.gov identifier: NCT01895361 [10 July 2013]; CSEG101A2202 (Phase 2), ClinicalTrials.gov identifier: NCT03264989 [29 August 2017].
Collapse
Affiliation(s)
- Sherwin K B Sy
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA.
| | - Chiaki Tanaka
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | - Kai Grosch
- Novartis Pharma AG, Forum 1, Novartis Campus, 4056, Basel, Switzerland
| |
Collapse
|
180
|
Alabed HBR, Gorello P, Pellegrino RM, Lancioni H, La Starza R, Taddei AA, Urbanelli L, Buratta S, Fernandez AGL, Matteucci C, Caniglia M, Arcioni F, Mecucci C, Emiliani C. Comparison between Sickle Cell Disease Patients and Healthy Donors: Untargeted Lipidomic Study of Erythrocytes. Int J Mol Sci 2023; 24:ijms24032529. [PMID: 36768849 PMCID: PMC9917006 DOI: 10.3390/ijms24032529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Sickle cell disease (SCD) is one of the most common severe monogenic disorders in the world caused by a mutation on HBB gene and characterized by hemoglobin polymerization, erythrocyte rigidity, vaso-occlusion, chronic anemia, hemolysis, and vasculopathy. Recently, the scientific community has focused on the multiple genetic and clinical profiles of SCD. However, the lipid composition of sickle cells has received little attention in the literature. According to recent studies, changes in the lipid profile are strongly linked to several disorders. Therefore, the aim of this study is to dig deeper into lipidomic analysis of erythrocytes in order to highlight any variations between healthy and patient subjects. 241 lipid molecular species divided into 17 classes have been annotated and quantified. Lipidomic profiling of SCD patients showed that over 24% of total lipids were altered most of which are phospholipids. In-depth study of significant changes in lipid metabolism can give an indication of the enzymes and genes involved. In a systems biology scenario, these variations can be useful to improve the understanding of the biochemical basis of SCD and to try to make a score system that could be predictive for the severity of clinical manifestations.
Collapse
Affiliation(s)
- Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy
| | - Paolo Gorello
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy
- Correspondence:
| | - Hovirag Lancioni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy
| | - Roberta La Starza
- Hematology and Bone Marrow Transplantation Unit, Laboratory of Molecular Medicine (CREO), Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Anna Aurora Taddei
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy
| | - Anair Graciela Lema Fernandez
- Hematology and Bone Marrow Transplantation Unit, Laboratory of Molecular Medicine (CREO), Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Caterina Matteucci
- Hematology and Bone Marrow Transplantation Unit, Laboratory of Molecular Medicine (CREO), Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Maurizio Caniglia
- Pediatric Oncology-Hematology, Azienda Ospedaliera di Perugia, 06100 Perugia, Italy
| | - Francesco Arcioni
- Pediatric Oncology-Hematology, Azienda Ospedaliera di Perugia, 06100 Perugia, Italy
| | - Cristina Mecucci
- Hematology and Bone Marrow Transplantation Unit, Laboratory of Molecular Medicine (CREO), Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| |
Collapse
|
181
|
Abstract
Sickle cell disease (SCD) results from a single base pair change in the sixth codon of the β-globin chain of hemoglobin, which promotes aggregation of deoxyhemoglobin, increasing rigidity of red blood cells and causing vaso-occlusive and hemolytic complications. Allogeneic transplant of hematopoietic stem cells (HSCs) can eliminate SCD manifestations but is limited by absence of well-matched donors and immune complications. Gene therapy with transplantation of autologous HSCs that are gene-modified may provide similar benefits without the immune complications. Much progress has been made, and patients are realizing significant clinical improvements in multiple trials using different approaches with lentiviral vector-mediated gene addition to inhibit hemoglobin aggregation. Gene editing approaches are under development to provide additional therapeutic opportunities. Gene therapy for SCD has advanced from an attractive concept to clinical reality.
Collapse
Affiliation(s)
- Shanna L White
- Department of Pediatrics, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, USA;
| | - Kevyn Hart
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Donald B Kohn
- Department of Pediatrics, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, USA;
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, USA
- The Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, David Geffen School of Medicine, University of California, Los Angeles, USA
| |
Collapse
|
182
|
Chiang KC, Gupta A, Sundd P, Krishnamurti L. Thrombo-Inflammation in COVID-19 and Sickle Cell Disease: Two Faces of the Same Coin. Biomedicines 2023; 11:338. [PMID: 36830874 PMCID: PMC9953430 DOI: 10.3390/biomedicines11020338] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/26/2023] Open
Abstract
People with sickle cell disease (SCD) are at greater risk of severe illness and death from respiratory infections, including COVID-19, than people without SCD (Centers for Disease Control and Prevention, USA). Vaso-occlusive crises (VOC) in SCD and severe SARS-CoV-2 infection are both characterized by thrombo-inflammation mediated by endothelial injury, complement activation, inflammatory lipid storm, platelet activation, platelet-leukocyte adhesion, and activation of the coagulation cascade. Notably, lipid mediators, including thromboxane A2, significantly increase in severe COVID-19 and SCD. In addition, the release of thromboxane A2 from endothelial cells and macrophages stimulates platelets to release microvesicles, which are harbingers of multicellular adhesion and thrombo-inflammation. Currently, there are limited therapeutic strategies targeting platelet-neutrophil activation and thrombo-inflammation in either SCD or COVID-19 during acute crisis. However, due to many similarities between the pathobiology of thrombo-inflammation in SCD and COVID-19, therapies targeting one disease may likely be effective in the other. Therefore, the preclinical and clinical research spurred by the COVID-19 pandemic, including clinical trials of anti-thrombotic agents, are potentially applicable to VOC. Here, we first outline the parallels between SCD and COVID-19; second, review the role of lipid mediators in the pathogenesis of these diseases; and lastly, examine the therapeutic targets and potential treatments for the two diseases.
Collapse
Affiliation(s)
| | - Ajay Gupta
- KARE Biosciences, Orange, CA 89128, USA
- Division of Nephrology, Hypertension and Kidney Transplantation, Department of Medicine, University of California Irvine (UCI) School of Medicine, Irvine, CA 92868, USA
| | - Prithu Sundd
- Vascular Medicine Institute and Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Lakshmanan Krishnamurti
- Division of Pediatric Hematology-Oncology, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
183
|
Cordovil K, Tassinari W, Oliveira RDVCD, Hökerberg Y. Social inequalities in the temporal trend of mortality from sickle cell disease in Brazil, 1996-2019. CAD SAUDE PUBLICA 2023; 39:e00256421. [PMID: 36651378 DOI: 10.1590/0102-311xen256421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 10/27/2022] [Indexed: 01/18/2023] Open
Abstract
Contrary to international trends, the mortality rate of sickle cell disease increased in Brazil after the implementation of the neonatal screening program, probably due to improving access to diagnosis. This study aimed to assess differences in the temporal trend of the mortality rate and median age at death from sickle cell disease in Brazil, considering implemented measures to expand diagnosis, and improve health care access in-country and in the international scenario. Time series were extracted from the Brazilian Mortality Information System from 1996 to 2019. Changes in the mortality rate and median age at death were verified via segmented regression models, which were stratified by sex, region of residence, and age. Most deaths occurred in non-white people, young adults, and the Southeast and Northeast population. Sickle cell disease mortality rate increased until 2010 (13.31%; 95%CI: 6.37; 20.70), particularly in individuals aged 30 years or more (12.78%; 95%CI: 2.98; 23.53) and in the Northeast (12.27%; 95%CI: 8.92; 15.72). Most deaths occurred in the second decade of life (3.01 deaths/million), with a 59% increase in the median age of death in Brazil, from 27.6 to 30.3 years, more pronounced in females and the North Region. The observed gain in the survival of sickle cell disease in Brazil is still much lower than in developed countries and presents regional disparities, probably due to the lack of access to health care and recent treatments, such as hydroxyurea, still restricted to hematological referral centers in Brazilian capitals.
Collapse
Affiliation(s)
- Karen Cordovil
- Escola Nacional de Saúde Pública Sergio Arouca, Fundação Oswaldo Cruz, Rio de Janeiro, Brasil
| | - Wagner Tassinari
- Instituto de Ciências Exatas, Universidade Federal Rural do Rio de Janeiro, Seropédica, Brasil
| | | | - Yara Hökerberg
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brasil
| |
Collapse
|
184
|
Li H, Kazmi JS, Lee S, Zhang D, Gao X, Maryanovich M, Torres L, Verma D, Kelly L, Ginzburg YZ, Frenette PS, Manwani D. Dietary iron restriction protects against vaso-occlusion and organ damage in murine sickle cell disease. Blood 2023; 141:194-199. [PMID: 36315910 PMCID: PMC10023724 DOI: 10.1182/blood.2022016218] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/07/2022] Open
Abstract
Sickle cell disease (SCD) is an inherited disorder resulting from a β-globin gene mutation, and SCD patients experience erythrocyte sickling, vaso-occlusive episodes (VOE), and progressive organ damage. Chronic hemolysis, inflammation, and repeated red blood cell transfusions in SCD can disrupt iron homeostasis. Patients who receive multiple blood transfusions develop iron overload, and another subpopulation of SCD patients manifest iron deficiency. To elucidate connections between dietary iron, the microbiome, and SCD pathogenesis, we treated SCD mice with an iron-restricted diet (IRD). IRD treatment reduced iron availability and hemolysis, decreased acute VOE, and ameliorated chronic organ damage in SCD mice. Our results extend previous studies indicating that the gut microbiota regulate disease in SCD mice. IRD alters microbiota load and improves gut integrity, together preventing crosstalk between the gut microbiome and inflammatory factors such as aged neutrophils, dampening VOE, and organ damage. These findings provide strong evidence for the therapeutic potential of manipulating iron homeostasis and the gut microbiome to ameliorate SCD pathophysiology. Many treatments, which are under development, focus on lowering the systemic iron concentration to relieve disease complications, and our data suggest that iron-induced changes in microbiota load and gut integrity are related- and novel-therapeutic targets.
Collapse
Affiliation(s)
- Huihui Li
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Jacob S. Kazmi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Sungkyun Lee
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Dachuan Zhang
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Xin Gao
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Lidiane Torres
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Divij Verma
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Libusha Kelly
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY
| | - Yelena Z. Ginzburg
- The Tisch Cancer Institute, Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Paul S. Frenette
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Deepa Manwani
- The Children’s Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
185
|
A randomized clinical trial of the efficacy and safety of rivipansel for sickle cell vaso-occlusive crisis. Blood 2023; 141:168-179. [PMID: 35981565 DOI: 10.1182/blood.2022015797] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/25/2022] [Accepted: 07/31/2022] [Indexed: 01/17/2023] Open
Abstract
The efficacy and safety of rivipansel, a predominantly E-selectin antagonist, were studied in a phase 3, randomized, controlled trial for vaso-occlusive crisis (VOC) requiring hospitalization (RESET). A total of 345 subjects (204 adults and 141 children) were randomized and 320 were treated (162 with rivipansel, 158 with placebo) with an IV loading dose, followed by up to 14 additional 12-hourly maintenance doses of rivipansel or placebo, in addition to standard care. Rivipansel was similarly administered during subsequent VOCs in the Open-label Extension (OLE) study. In the full analysis population, the median time to readiness for discharge (TTRFD), the primary end point, was not different between rivipansel and placebo (-5.7 hours, P = .79; hazard ratio, 0.97), nor were differences seen in secondary end points of time to discharge (TTD), time to discontinuation of IV opioids (TTDIVO), and cumulative IV opioid use. Mean soluble E-selectin decreased 61% from baseline after the loading dose in the rivipansel group, while remaining unchanged in the placebo group. In a post hoc analysis, early rivipansel treatment within 26.4 hours of VOC pain onset (earliest quartile of time from VOC onset to treatment) reduced median TTRFD by 56.3 hours, reduced median TTD by 41.5 hours, and reduced median TTDIVO by 50.5 hours, compared with placebo (all P < .05). A similar subgroup analysis comparing OLE early-treatment with early-treatment RESET placebo showed a reduction in TTD of 23.1 hours (P = .062) and in TTDIVO of 30.1 hours (P = .087). Timing of rivipansel administration after pain onset may be critical to achieving accelerated resolution of acute VOC. Trial Registration: Clinicaltrials.gov, NCT02187003 (RESET), NCT02433158 (OLE).
Collapse
|
186
|
Dutra VDF, Biassi TP, Figueiredo MS. Sickle cell anemia: hierarchical cluster analysis and clinical profile in a cohort in Brazil. Hematol Transfus Cell Ther 2023; 45:45-51. [PMID: 34930711 PMCID: PMC9938484 DOI: 10.1016/j.htct.2021.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/04/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Sickle cell anemia is a monogenic disorder caused by a mutation in the β-hemoglobin gene, resulting in sickle hemoglobin that can polymerize. Presentation and clinical course have significant inter-individual variability and classifying these patients for severity is a challenge. METHODS We applied hierarchical clusters with 10 routine laboratory tests to understand if this grouping could be associated with clinical manifestations. We included 145 adult homozygous patients (SS) at an outpatient clinic in a retrospective study. RESULTS We found five clusters by counting those that had been differentiated by unconjugated bilirubin, reticulocytes, LDH, leukocytes, lymphocytes and monocytes. When comparing groups to clinical findings, the clusters were different only for liver abnormality. Cluster 3 had the lower median of reticulocytes, LDH, leukocytes, lymphocytes and monocytes and a higher percentage of patients under treatment. Clusters 4 and 5 had higher frequencies of liver impairment and higher medians of reticulocytes, LDH, leukocytes, lymphocytes and monocytes. Hemolysis and inflammation seemed to influence the grouping. CONCLUSION In our study, cluster analysis showed five groups that exhibited different degrees of inflammation and hemolysis. When comparing clinical data, the result was different only for the criteria of liver abnormality.
Collapse
Affiliation(s)
- Valéria de Freitas Dutra
- Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil.
| | | | | |
Collapse
|
187
|
Siransy LK, Dasse RS, Adou H, Kouacou P, Kouamenan S, Sekongo Y, Yeboah R, Memel C, Assi-Sahoin A, Moussa SY, Oura D, Seri J. Are IL-1 family cytokines important in management of sickle cell disease in Sub-Saharan Africa patients? Front Immunol 2023; 14:954054. [PMID: 36969226 PMCID: PMC10034065 DOI: 10.3389/fimmu.2023.954054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction Sickle cell disease (SCD) is the most common genetic disease found in Africa and throughout the world. It is responsible for a high rate of hemolysis, systemic inflammation, and modulation of the immune system with the involvement of immunological molecules, such as cytokines. IL-1β is a major inflammatory cytokine. IL-18 and IL-33, members of IL-1 family, also exhibit characteristics of inflammation-related cytokines. Thus, in order to contribute to the evaluation of the severity and prognosis of SCD in Africa, this study aimed to estimate the cytokine response, in particular the levels of cytokines of the IL-1 family, in sickle cell patients living in a Sub-Saharan country. Methods Ninety patients with a diagnosis of SCD were recruited with different hemoglobin types. Samples were assessed for cytokine levels using the Human Inflammation Panel assay from BioLegend. The assay allows the simultaneous quantification of 13 human inflammatory cytokines/chemokines, i.e., IL-1β, IFN-α2, IFN-γ, TNFα, MCP-1 (CCL2), IL-6, IL-8 (CXCL8), IL-10, IL-12p70, IL-17A, IL-18, IL-23, and IL-33. Results and discussion the assessment of plasma cytokines in SCD patients revealed significantly increased levels of IL-1 family cytokines in crisis compared to steady state, suggesting a substantial involvement of these cytokines in clinical exacerbation. This suggests the possibility of a causal effect in the SCD pathology and can open the way to define better care, pointing toward new therapeutic avenues for sickle disease in Sub-Saharan Africa.
Collapse
Affiliation(s)
- Liliane K. Siransy
- Immunology–Allergology Department, Medical Sciences, Felix Houphouet Boigny University, Abidjan, Côte d’Ivoire
- Transfusional therapeutic department, National Blood Transfusion Center, Abidjan, Côte d’Ivoire
- *Correspondence: Liliane K. Siransy, ,
| | - Romuald S. Dasse
- Immunology–Allergology Department, Medical Sciences, Felix Houphouet Boigny University, Abidjan, Côte d’Ivoire
| | - Honoré Adou
- Immunology–Allergology Department, Medical Sciences, Felix Houphouet Boigny University, Abidjan, Côte d’Ivoire
| | - Patricia Kouacou
- Immunology–Allergology Department, Medical Sciences, Felix Houphouet Boigny University, Abidjan, Côte d’Ivoire
| | - Sidonie Kouamenan
- Transfusional therapeutic department, National Blood Transfusion Center, Abidjan, Côte d’Ivoire
| | - Yassongui Sekongo
- Transfusional therapeutic department, National Blood Transfusion Center, Abidjan, Côte d’Ivoire
| | - Richard Yeboah
- Immunology–Allergology Department, Medical Sciences, Felix Houphouet Boigny University, Abidjan, Côte d’Ivoire
| | - Charlene Memel
- Immunology Department, CHU Bouake, Alassane Ouattara University, Bouake, Côte d’Ivoire
| | - Aniella Assi-Sahoin
- Immunology–Allergology Department, Medical Sciences, Felix Houphouet Boigny University, Abidjan, Côte d’Ivoire
| | - Salimata Y. Moussa
- Transfusional therapeutic department, National Blood Transfusion Center, Abidjan, Côte d’Ivoire
| | - Doris Oura
- Transfusional therapeutic department, National Blood Transfusion Center, Abidjan, Côte d’Ivoire
| | - Jocelyne Seri
- Immunology–Allergology Department, Medical Sciences, Felix Houphouet Boigny University, Abidjan, Côte d’Ivoire
| |
Collapse
|
188
|
Shen F, Zheng G, Setegne M, Tenglin K, Izadi M, Xie H, Zhai L, Orkin SH, Dassama LMK. A Cell-Permeant Nanobody-Based Degrader That Induces Fetal Hemoglobin. ACS CENTRAL SCIENCE 2022; 8:1695-1703. [PMID: 36589886 PMCID: PMC9801508 DOI: 10.1021/acscentsci.2c00998] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Indexed: 06/13/2023]
Abstract
Proximity-based strategies to degrade proteins have enormous therapeutic potential in medicine, but the technologies are limited to proteins for which small molecule ligands exist. The identification of such ligands for therapeutically relevant but "undruggable" proteins remains challenging. Herein, we employed yeast surface display of synthetic nanobodies to identify a protein ligand selective for BCL11A, a critical repressor of fetal globin gene transcription. Fusion of the nanobody to a cell-permeant miniature protein and an E3 adaptor creates a degrader that depletes cellular BCL11A in differentiated primary erythroid precursor cells, thereby inducing the expression of fetal hemoglobin, a modifier of clinical severity of sickle cell disease and β-thalassemia. Our strategy provides a means of fetal hemoglobin induction through reversible, temporal modulation of BCL11A. Additionally, it establishes a new paradigm for the targeted degradation of previously intractable proteins.
Collapse
Affiliation(s)
- Fangfang Shen
- Department
of Chemistry and Sarafan ChEM-H, Stanford
University, Stanford, California 94305, United States
| | - Ge Zheng
- Dana
Farber Boston Children’s Cancer and Blood Disorders Center
and Howard Hughes Medical Institute, Boston, Massachusetts 02215, United States
- Department
of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Mekedlawit Setegne
- Department
of Chemistry and Sarafan ChEM-H, Stanford
University, Stanford, California 94305, United States
| | - Karin Tenglin
- Dana
Farber Boston Children’s Cancer and Blood Disorders Center
and Howard Hughes Medical Institute, Boston, Massachusetts 02215, United States
- Department
of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Manizheh Izadi
- Dana
Farber Boston Children’s Cancer and Blood Disorders Center
and Howard Hughes Medical Institute, Boston, Massachusetts 02215, United States
| | - Henry Xie
- Dana
Farber Boston Children’s Cancer and Blood Disorders Center
and Howard Hughes Medical Institute, Boston, Massachusetts 02215, United States
- Department
of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Liting Zhai
- Department
of Chemistry and Sarafan ChEM-H, Stanford
University, Stanford, California 94305, United States
| | - Stuart H. Orkin
- Dana
Farber Boston Children’s Cancer and Blood Disorders Center
and Howard Hughes Medical Institute, Boston, Massachusetts 02215, United States
- Department
of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Laura M. K. Dassama
- Department
of Chemistry and Sarafan ChEM-H, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
189
|
Lumbala PK, Mbayabo G, Ngole MN, Lumaka A, Race V, Matthijs G, Van Geet C, Lukusa PT, Devriendt K, Mikobi TM. Clinical and laboratory characterization of adult sickle cell anemia patients in Kinshasa. PLoS One 2022; 17:e0278478. [PMID: 36525434 PMCID: PMC9757547 DOI: 10.1371/journal.pone.0278478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Sickle cell anemia (SCA) is a monogenic hemoglobinopathy associated with severe acute and chronic complications, with the highest incidence worldwide in Sub-Saharan Africa. The wide variability in clinical manifestations suggest that a uniform response to hydroxurea may not be attained. In view of a potential treatment with hydroxyurea (HU), we assessed the variability of clinical and hematological manifestations in a cohort of adults with SCA in Kinshasa, capital of the DR Congo in Central Africa. METHODS A cross-sectional study was conducted in a hospital dedicated to SCA management in Kinshasa. Clinical history of patients was recorded, a complete physical examination performed. The diagnosis was confirmed by means of DNA analysis. A full blood count and hemolysis markers were measured. The severity of the disease was evaluated by means of a previously reported score. RESULTS The study group consisted of 166 genetically confirmed SCA patients. The SCA severity was mild in 28.9%, moderate in 64.5% and severe in 6.6%. The disease severity score increased with patient's age (p ≤ 0.001). The severity was higher in males compared to females (p = 0.012). In males, the severity score was correlated with the presence of priapism (p = 0.045), a manifestation not previously incorporated in the severity score. The severity score was inversely correlated with the fetal hemoglobin (HbF) rate (p = 0.005). Malnutrition (BMI <18.5 kg/m2) was present in 47% of patients and was related to the male sex, hip disease (aOR 3.11; p = 0.019) and severe phenotype (aOR 3.53; p = 0.012). Leg ulcers were more frequent in males than in females (p = 0.001; OR 24.3) and were correlated with the number of days of hospitalization (p = 0.029). Hip disease was related to the increasing age (p = 0.008). CONCLUSION In this selected, hospital-based populations of adults with SCA, severe disease was rare, which may be due to survival bias. However, two thirds had moderate severity of the disease, mostly with a low HbF, and they may benefit from HU treatment. In the Central-African setting the separation between vaso-occlusive and hyperhemolytic sub-phenotypes was not applicable.
Collapse
Affiliation(s)
| | - Gloire Mbayabo
- Department of Pediatrics, University of Kinshasa, Kinshasa, DRC
| | - Mamy Nzita Ngole
- Department of Clinical Biology, University of Kinshasa, Kinshasa, DRC
| | - Aimé Lumaka
- Faculty of Medicine, Center of Human Genetics, University of Kinshasa, Kinshasa, DRC
| | - Valerie Race
- Center for Human Genetics, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Gert Matthijs
- Center for Human Genetics, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Chris Van Geet
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | | | - Koenraad Devriendt
- Center for Human Genetics, KU Leuven and University Hospitals Leuven, Leuven, Belgium
- * E-mail: (KD); (TMM)
| | - Tite Minga Mikobi
- Faculty of Medicine, Center of Human Genetics, University of Kinshasa, Kinshasa, DRC
- Faculty of Medicine, Molecular Biology and Human Genetics Department of fundamental sciences, University of Kinshasa, Kinshasa, DRC
- * E-mail: (KD); (TMM)
| |
Collapse
|
190
|
Elagooz R, Dhara AR, Gott RM, Adams SE, White RA, Ghosh A, Ganguly S, Man Y, Owusu-Ansah A, Mian OY, Gurkan UA, Komar AA, Ramamoorthy M, Gnanapragasam MN. PUM1 mediates the posttranscriptional regulation of human fetal hemoglobin. Blood Adv 2022; 6:6016-6022. [PMID: 35667093 PMCID: PMC9699939 DOI: 10.1182/bloodadvances.2021006730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/30/2022] [Indexed: 12/14/2022] Open
Abstract
The fetal-to-adult hemoglobin switching at about the time of birth involves a shift in expression from γ-globin to β-globin in erythroid cells. Effective re-expression of fetal γ-globin can ameliorate sickle cell anemia and β-thalassemia. Despite the physiological and clinical relevance of this switch, its posttranscriptional regulation is poorly understood. Here, we identify Pumilo 1 (PUM1), an RNA-binding protein with no previously reported functions in erythropoiesis, as a direct posttranscriptional regulator of β-globin switching. PUM1, whose expression is regulated by the erythroid master transcription factor erythroid Krüppel-like factor (EKLF/KLF1), peaks during erythroid differentiation, binds γ-globin messenger RNA (mRNA), and reduces γ-globin (HBG1) mRNA stability and translational efficiency, which culminates in reduced γ-globin protein levels. Knockdown of PUM1 leads to a robust increase in fetal hemoglobin (∼22% HbF) without affecting β-globin levels in human erythroid cells. Importantly, targeting PUM1 does not limit the progression of erythropoiesis, which provides a potentially safe and effective treatment strategy for sickle cell anemia and β-thalassemia. In support of this idea, we report elevated levels of HbF in the absence of anemia in an individual with a novel heterozygous PUM1 mutation in the RNA-binding domain (p.(His1090Profs∗16); c.3267_3270delTCAC), which suggests that PUM1-mediated posttranscriptional regulation is a critical player during human hemoglobin switching.
Collapse
Affiliation(s)
- Reem Elagooz
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH
| | - Anita R. Dhara
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH
| | - Rose M. Gott
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH
| | - Sarah E. Adams
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH
| | - Rachael A. White
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH
| | - Arnab Ghosh
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH
| | - Shinjini Ganguly
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Yuncheng Man
- Department of Mechanical and Aerospace Engineering, University Hospitals Rainbow Babies and Children’s Hospital, Case Western Reserve University, Cleveland, OH
| | - Amma Owusu-Ansah
- Department of Pediatrics, Division of Hematology and Oncology, University Hospitals Rainbow Babies and Children’s Hospital, Case Western Reserve University, Cleveland, OH
| | - Omar Y. Mian
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Umut A. Gurkan
- Department of Mechanical and Aerospace Engineering, University Hospitals Rainbow Babies and Children’s Hospital, Case Western Reserve University, Cleveland, OH
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Anton A. Komar
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, OH
| | - Mahesh Ramamoorthy
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH
| | - Merlin Nithya Gnanapragasam
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
191
|
Identification and characterization of RBM12 as a novel regulator of fetal hemoglobin expression. Blood Adv 2022; 6:5956-5968. [PMID: 35622975 PMCID: PMC9678958 DOI: 10.1182/bloodadvances.2022007904] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/21/2022] [Indexed: 02/01/2023] Open
Abstract
The fetal-to-adult hemoglobin transition is clinically relevant because reactivation of fetal hemoglobin (HbF) significantly reduces morbidity and mortality associated with sickle cell disease (SCD) and β-thalassemia. Most studies on the developmental regulation of the globin genes, including genome-wide genetics screens, have focused on DNA binding proteins, including BCL11A and ZBTB7A/LRF and their cofactors. Our understanding of RNA binding proteins (RBPs) in this process is much more limited. Two RBPs, LIN28B and IGF2BP1, are known posttranscriptional regulators of HbF production, but a global view of RBPs is still lacking. Here, we carried out a CRISPR/Cas9-based screen targeting RBPs harboring RNA methyltransferase and/or RNA recognition motif (RRM) domains and identified RNA binding motif 12 (RBM12) as a novel HbF suppressor. Depletion of RBM12 induced HbF expression and attenuated cell sickling in erythroid cells derived from patients with SCD with minimal detrimental effects on cell maturation. Transcriptome and proteome profiling revealed that RBM12 functions independently of major known HbF regulators. Enhanced cross-linking and immunoprecipitation followed by high-throughput sequencing revealed strong preferential binding of RBM12 to 5' untranslated regions of transcripts, narrowing down the mechanism of RBM12 action. Notably, we pinpointed the first of 5 RRM domains as essential, and, in conjunction with a linker domain, sufficient for RBM12-mediated HbF regulation. Our characterization of RBM12 as a negative regulator of HbF points to an additional regulatory layer of the fetal-to-adult hemoglobin switch and broadens the pool of potential therapeutic targets for SCD and β-thalassemia.
Collapse
|
192
|
Chu Y, Talano JA, Baxter-Lowe LA, Verbsky JW, Morris E, Mahanti H, Ayello J, Keever-Taylor C, Johnson B, Weinberg RS, Shi Q, Moore TB, Fabricatore S, Grossman B, van de Ven C, Shenoy S, Cairo MS. Donor chimerism and immune reconstitution following haploidentical transplantation in sickle cell disease. Front Immunol 2022; 13:1055497. [PMID: 36569951 PMCID: PMC9780682 DOI: 10.3389/fimmu.2022.1055497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction We previously reported the initial results of a phase II multicenter transplant trial using haploidentical parental donors for children and aolescents with high-risk sickle cell disease achieving excellent survival with exceptionally low rates of graft-versus-host disease and resolution of sickle cell disease symptoms. To investigate human leukocyte antigen (HLA) sensitization, graft characteristics, donor chimerism, and immune reconstitution in these recipients. Methods CD34 cells were enriched using the CliniMACS® system with a target dose of 10 x 106 CD34+ cells/kg with a peripheral blood mononuclear cell (PBMNC) addback dose of 2x105 CD3/kg in the final product. Pre-transplant HLA antibodies were characterized. Donor chimerism was monitored 1-24 months post-transplant. Comprehensive assessment of immune reconstitution included lymphocyte subsets, plasma cytokines, complement levels, anti-viral T-cell responses, activation markers, and cytokine production. Infections were monitored. Results HLA antibodies were detected in 7 of 11 (64%) evaluable patients but rarely were against donor antigens. Myeloid engraftment was rapid (100%) at a median of 9 days. At 30 days, donor chimerism was 93-99% and natural killer cell levels were restored. By 60 days, CD19 B cells were normal. CD8 and CD4 T-cells levels were normal by 279 and 365 days, respectively. Activated CD4 and CD8 T-cells were elevated at 100-365 days post-transplant while naïve cells remained below baseline. Tregs were elevated at 100-270 days post-transplant, returning to baseline levels at one year. At one year, C3 and C4 levels were above baseline and CH50 levels were near baseline. At one year, cytokine levels were not significantly different from baseline. Discussion These results suggest that haploidentical transplantation with CD34-enriched cells and peripheral blood mononuclear cell addback results in rapid engraftment, sustained donor chimerism and broad-based immune reconstitution.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Julie-An Talano
- Department of Pediatrics, Hematology/Oncology and BMT, Children’s Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lee Ann Baxter-Lowe
- Department of Pathology, Children’s Hospital of Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - James W. Verbsky
- Department of Pediatrics, Hematology/Oncology and BMT, Children’s Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Erin Morris
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Harshini Mahanti
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - Carolyn Keever-Taylor
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bryon Johnson
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Qiuhu Shi
- Department of Epidemiology and Community Health, New York Medical College, Valhalla, NY, United States
| | - Theodore B. Moore
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Sandra Fabricatore
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Brenda Grossman
- Department of Pathology and Immunology, Washington University, St Louis, MO, United States
| | - Carmella van de Ven
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Shalini Shenoy
- Department of Pediatrics and Transfusion Medicine, Washington University, St Louis, MO, United States
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, New York Medical College, Valhalla, NY, United States,Department of Medicine, New York Medical College, Valhalla, NY, United States,Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, United States,Department of Cell Biology, New York Medical College, Valhalla, NY, United States,Department of Anatomy, New York Medical College, Valhalla, NY, United States,*Correspondence: Mitchell S. Cairo,
| |
Collapse
|
193
|
Drakopoulou E, Georgomanoli M, Lederer CW, Panetsos F, Kleanthous M, Voskaridou E, Valakos D, Papanikolaou E, Anagnou NP. The Optimized γ-Globin Lentiviral Vector GGHI-mB-3D Leads to Nearly Therapeutic HbF Levels In Vitro in CD34 + Cells from Sickle Cell Disease Patients. Viruses 2022; 14:v14122716. [PMID: 36560719 PMCID: PMC9783242 DOI: 10.3390/v14122716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/13/2022] [Accepted: 11/30/2022] [Indexed: 12/11/2022] Open
Abstract
We have previously demonstrated that both the original γ-globin lentiviral vector (LV) GGHI and the optimized GGHI-mB-3D LV, carrying the novel regulatory elements of the 3D HPFH-1 enhancer and the 3' β-globin UTR, can significantly increase HbF production in thalassemic CD34+ cells and ameliorate the disease phenotype in vitro. In the present study, we investigated whether the GGHI-mB-3D vector can also exhibit an equally therapeutic effect, following the transduction of sickle cell disease (SCD) CD34+ cells at MOI 100, leading to HbF increase coupled with HbS decrease, and thus, to phenotype improvement in vitro. We show that GGHI-mB-3D LV can lead to high and potentially therapeutic HbF levels, reaching a mean 2-fold increase to a mean value of VCN/cell of 1.0 and a mean transduction efficiency of 55%. Furthermore, this increase was accompanied by a significant 1.6-fold HbS decrease, a beneficial therapeutic feature for SCD. In summary, our data demonstrate the efficacy of the optimized γ-globin lentiviral vector to improve the SCD phenotype in vitro, and highlights its potential use in future clinical SCD trials.
Collapse
Affiliation(s)
- Ekati Drakopoulou
- Laboratory of Cell and Gene Therapy, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Georgomanoli
- Laboratory of Cell and Gene Therapy, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Carsten W. Lederer
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | | | - Marina Kleanthous
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Ersi Voskaridou
- Thalassemia and Sickle Cell Disease Centre, Laiko General Hospital, 11527 Athens, Greece
| | - Dimitrios Valakos
- Laboratory of Molecular Biology, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
| | - Eleni Papanikolaou
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nicholas P. Anagnou
- Laboratory of Cell and Gene Therapy, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Correspondence:
| |
Collapse
|
194
|
Knight-Madden JM, Hambleton IR. Inhaled bronchodilators for acute chest syndrome in people with sickle cell disease. Cochrane Database Syst Rev 2022; 12:CD003733. [PMID: 36458811 PMCID: PMC9717338 DOI: 10.1002/14651858.cd003733.pub5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND Bronchodilators are used to treat bronchial hyper-responsiveness in asthma. Bronchial hyper-responsiveness may be a component of acute chest syndrome in people with sickle cell disease. Therefore, bronchodilators may be useful in the treatment of acute chest syndrome. This is an update of a previously published Cochrane Review. OBJECTIVES The aim of the review is to determine whether the use of inhaled, short-acting bronchodilators for acute chest syndrome reduces morbidity and mortality in people with sickle cell disease and to assess whether this treatment causes adverse effects. SEARCH METHODS We searched the Cochrane Cystic Fibrosis and Genetic Disorders Group's Trials Register comprising references identified from comprehensive electronic database searches, handsearches of relevant journals and abstract books of conference proceedings. Additional searches were carried out on MEDLINE (1966 to 2004) and Embase (1981 to 2004) and ongoing trial registries (28 September 2022). Date of the most recent search of the Group's Haemoglobinopathies Trials Register: 25 July 2022. SELECTION CRITERIA Randomised or quasi-randomised controlled trials. Trials using quasi-randomisation methods will be included in future updates of this review if there is sufficient evidence that the treatment and control groups are similar at baseline. DATA COLLECTION AND ANALYSIS We found no trials investigating the use of bronchodilators for acute chest syndrome in people with sickle cell disease. MAIN RESULTS We found no trials investigating the use of bronchodilators for acute chest syndrome in people with sickle cell disease. AUTHORS' CONCLUSIONS If bronchial hyper-responsiveness is an important component of some episodes of acute chest syndrome in people with sickle cell disease, the use of inhaled bronchodilators may be indicated. There is need for a well-designed, adequately-powered randomised controlled trial to assess the benefits and risks of the addition of inhaled bronchodilators to established therapies for acute chest syndrome in people with sickle cell disease.
Collapse
Affiliation(s)
- Jennifer M Knight-Madden
- Caribbean Institute for Health Research - Sickle Cell Unit, The University of the West Indies, Kingston, Jamaica
| | - Ian R Hambleton
- Caribbean Institute for Health Research - George Alleyne Chronic Disease Research Centre, The University of the West Indies, Bridgetown, Barbados
| |
Collapse
|
195
|
Winer JC, Yee ME, Ataga KI, Lebensburger JD, Zahr RS. Patients with sickle cell disease who develop end-stage kidney disease continue to experience poor survival - A 19-year United States Renal Data System study. Br J Haematol 2022; 199:e43-e47. [PMID: 36191963 PMCID: PMC10739628 DOI: 10.1111/bjh.18494] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Jeffrey C Winer
- Division of Academic Hospital Medicine, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Marianne E Yee
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kenneth I Ataga
- Center for Sickle Cell Disease, University of Tennessee Health Science Center Memphis, Tennessee, USA
| | - Jeffrey D Lebensburger
- Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rima S Zahr
- Division of Pediatric Nephrology and Hypertension, University of Tennessee Health Science Center Memphis, Tennessee, USA
| |
Collapse
|
196
|
Khalaf Z, Mahmood M. Acute chest syndrome in post-operative sickle cell disease patients: A systematic review of predisposing factors and interventions. SURGERY IN PRACTICE AND SCIENCE 2022; 11:100143. [PMID: 39845168 PMCID: PMC11749942 DOI: 10.1016/j.sipas.2022.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background Acute chest syndrome (ACS) is a life-threatening complication of sickle cell disease (SCD). ACS can be precipitated by surgery. This review studies the clinical profiles of ACS following abdominal surgeries to identify risk factors and the impact of different interventions. Methods A systematic review using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines was performed in Medline, Embase, Cochrane, and Web of Science. The search terms related to "sickle cell disease", "acute chest syndrome", and "abdominal surgery". Two researchers independently assessed studies for eligibility. Data was reported as a thematic qualitative synthesis. Results Twenty-four articles were included. ACS occurs in 4% to 23% of SCD patients following abdominal surgeries. Conflicting views exist on the link between age and ACS. Sex is not associated with ACS. Most studies found no association between SCD severity and ACS. One study linked severe SCD with a likelihood of serious complications. Two studies found no association between comorbidities and ACS rates and one study found that pulmonary comorbidities were linked to higher rates of postoperative ACS. ACS is associated with postoperative desaturation. Patients developed leukocytosis, sterile blood and sputum cultures and pulmonary infiltrates. Conservative and risk-based transfusions reduce ACS rates. There is no consensus on whether using an open or laparoscopic approach affects the rates of ACS. Conclusion Conflicting views exist on the correlations between age and comorbidities with ACS. Findings in ACS patients included postoperative desaturation, sterile blood and sputum, and pulmonary infiltrates ipsilateral to the side of the surgery. It has been suggested that postoperative pain may be an early indicator of ACS. Future studies should address the impact of intraoperative complications on postoperative ACS rates. Individualised risk-based transfusions confer benefit. Interventions such as prophylactic cholecystectomies, prophylactic postoperative CPAP, and minimally invasive laparoscopy need further exploration.Systematic review registration: PROSPERO CRD42022337302; https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=337302.
Collapse
Affiliation(s)
- Zahra Khalaf
- Department of Postgraduate Surgical Studies, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Maria Mahmood
- Clinical Skills Laboratories, Trinity Center for Health Sciences, Tallaght University Hospital, Dublin, D24 NR0A, Ireland
| |
Collapse
|
197
|
Dayer LE, Wagner R, King D, Lakkad M, Wilson LA, Montgomery C, Painter JT. Impact of Hydroxyurea Starting Dose on Pain Outcomes in Patients with Sickle Cell Disease. J Pain Palliat Care Pharmacother 2022; 36:223-227. [PMID: 36688614 DOI: 10.1080/15360288.2022.2128154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In patients with sickle cell disease, hydroxyurea decreases the number of pain crises experienced. This study aimed to evaluate the difference in pain outcomes between patients started on a guideline concordant, weight-based starting dose of at least 15 mg/kg/day of hydroxyurea and those not. The first prescription of hydroxyurea was the baseline date, follow-up was a visit 60-120 days after baseline. The primary outcome was the change in opioid prescribing between baseline and follow-up. 138 patients met inclusion criteria; of these, 55 were started on a guideline concordant dose of hydroxyurea. Greater white blood cell count (9.5 vs 12.0; p < 0.01) was statistically associated with subtherapeutic dosing. Greater actual body weight (68.0 vs 72.1 kg; p = 0.16) also appeared higher in the non-guideline concordant group. No statistically significant difference in opioid prescribing was observed between those started on a guideline concordant dose of hydroxyurea and those who were not. In the guideline concordant starting dose group, 42% had a reduction in pain scores at first follow up, compared to 35% with a non-guideline recommended starting dose. (p = 0.41). While this difference is in the direction that would be expected based on the guidelines, the difference does not appear to be clinically meaningful.
Collapse
|
198
|
Nero A, Bozzo J. Economics of Sickle Cell Disease and Evidence to Support Comprehensive Care. Hematol Oncol Clin North Am 2022; 36:1125-1135. [DOI: 10.1016/j.hoc.2022.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
199
|
Desai AA, Machado RF, Cohen RT. The Cardiopulmonary Complications of Sickle Cell Disease. Hematol Oncol Clin North Am 2022; 36:1217-1237. [PMID: 36400540 PMCID: PMC10323820 DOI: 10.1016/j.hoc.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Sickle cell disease (SCD) is a genetic hemoglobinopathy associated with extensive morbidity and early mortality. While there have been recent improvements in available disease-modifying therapies for SCD, cardiopulmonary complications remain a major risk factor for death in this population. We provide an overview of current knowledge regarding several of the major acute and chronic cardiopulmonary complications in SCD, including: acute chest syndrome, airway disease, lung function abnormalities, nocturnal hypoxemia and sleep disordered breathing, pulmonary vascular disease, and sickle cell cardiomyopathy.
Collapse
Affiliation(s)
- Ankit A Desai
- Department of Medicine, Indiana School of Medicine, Indianapolis, IN, USA; Indiana University, 950 W. Walnut Street R2 Building, Room 466, Indianapolis, IN 46202, USA
| | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Room C400, Walther Hall, R3 980 W. Walnut Street, Indianapolis, IN 46202, USA
| | - Robyn T Cohen
- Department of Pediatrics, Boston Medical Center/Boston University School of Medicine, 801 Albany Street 4th Floor, Boston, MA 02118, USA.
| |
Collapse
|
200
|
Sickle Cell Disease Pathophysiology and Related Molecular and Biophysical Biomarkers. Hematol Oncol Clin North Am 2022; 36:1077-1095. [DOI: 10.1016/j.hoc.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|