CC BY 4.0 · Pharmaceutical Fronts 2023; 05(02): e57-e63
DOI: 10.1055/s-0043-1768918
Review Article

The Present Condition of Sickle Cell Disease: An Overview of Stem Cell Transplantation as a Cure

Md. Sadique Hussain
1   School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
,
Varunesh Chaturvedi
1   School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
› Author Affiliations

Abstract

Treatment of sickle cell disease (SCD) remains largely palliative. While it can enhance living standards, persons having SCD still suffer from extreme sickling crises, end-organ destruction, and reduced life expectancy. Increasing research has resulted in the recognition and advancement of stem cell transplantation and gene therapy as possible solutions for SCDs. However, there have been various factors that have hindered their clinical application. The more advantageous of the two, stem cell transplantation, is constrained by a small donor pool, transplant difficulties, and eligibility requirements. The current article reviewed the literature on SCDs, current treatment options, and more particularly the progress of stem cell transplants. It outlined various challenges of stem cell transplant and proposed ways to increase the donor pool using alternative strategies and modifications of regimen conditioning with minimal transplant-related toxicities and associated complications.



Publication History

Received: 28 January 2023

Accepted: 13 April 2023

Article published online:
24 May 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Herrick JB. Peculiar elongated and sickle-shaped red blood corpuscles in a case of severe anemia. JAMA 2014; 312 (10) 1063
  • 2 Chakravorty S, Williams TN. Sickle cell disease: a neglected chronic disease of increasing global health importance. Arch Dis Child 2015; 100 (01) 48-53
  • 3 Pauling L, Itano HA, Singer SJ, Wells IC. Sickle cell anemia a molecular disease. Science 1949; 110 (2865): 543-548
  • 4 Ingram VM. Abnormal human haemoglobins. I. The comparison of normal human and sickle-cell haemoglobins by fingerprinting. Biochim Biophys Acta 1958; 28 (03) 539-545
  • 5 Embury SH. The interaction of coexistent α-thalassemia and sickle cell anemia: a model for the clinical and cellular results of diminished polymerization?. Ann N Y Acad Sci 1985; 445 (01) 37-44
  • 6 Tanabe P, Spratling R, Smith D, Grissom P, Hulihan M. CE: Understanding the complications of sickle cell disease. Am J Nurs 2019; 119 (06) 26-35
  • 7 Trivedi K, Hussain MS, Mohapatra C. Role of glutamine as an ergogenic amino acid during fatigue. J Clinic Med Rev Rep 2022; 4 (02) 1-6
  • 8 Hussain MS, Tyagi S, Khatri H, Singh S. Stem cell therapy for myocardial infarction: a mini-review. Asian J Pharmaceut Res Dev 2022; 10 (02) 122-124
  • 9 Kaul DK, Finnegan E, Barabino GA. Sickle red cell-endothelium interactions. Microcirculation 2009; 16 (01) 97-111
  • 10 Pathare A, Kindi SA, Daar S, Dennison D. Cytokines in sickle cell disease. Hematology 2003; 8 (05) 329-337
  • 11 Knight-Perry J, DeBaun MR, Strunk RC, Field JJ. Leukotriene pathway in sickle cell disease: a potential target for directed therapy. Expert Rev Hematol 2009; 2 (01) 57-68
  • 12 McGann PT, Ware RE. Hydroxyurea therapy for sickle cell anemia. Expert Opin Drug Saf 2015; 14 (11) 1749-1758
  • 13 Dresler WF, Stein R. Ueber den hydroxylharnstoff. ustus Liebigs Annalen der Chemie 1869; 150 (02) 242-252
  • 14 Platt OS, Orkin SH, Dover G, Beardsley GP, Miller B, Nathan DG. Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia. J Clin Invest 1984; 74 (02) 652-656
  • 15 Habara A, Steinberg MH. Minireview: Genetic basis of heterogeneity and severity in sickle cell disease. Exp Biol Med (Maywood) 2016; 241 (07) 689-696
  • 16 Charache S, Terrin ML, Moore RD. et al; Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. N Engl J Med 1995; 332 (20) 1317-1322
  • 17 Ware RE, Eggleston B, Redding-Lallinger R. et al. Predictors of fetal hemoglobin response in children with sickle cell anemia receiving hydroxyurea therapy. Blood 2002; 99 (01) 10-14
  • 18 Strouse JJ, Lanzkron S, Beach MC. et al. Hydroxyurea for sickle cell disease: a systematic review for efficacy and toxicity in children. Pediatrics 2008; 122 (06) 1332-1342
  • 19 Chapman TR, Kinsella TJ. Ribonucleotide reductase inhibitors: a new look at an old target for radiosensitization. Front Oncol 2012; 1: 56
  • 20 Foong WC, Loh CK, Ho JJ, Lau DS. Foetal haemoglobin inducers for reducing blood transfusion in non-transfusion-dependent beta-thalassaemias. Cochrane Database Syst Rev 2023; 1 (01) CD013767
  • 21 Ho JA, Pickens CV, Gamcsik MP, Colvin OM, Ware RE. In vitro induction of fetal hemoglobin in human erythroid progenitor cells. Exp Hematol 2003; 31 (07) 586-591
  • 22 Ali MA, Ahmad A, Chaudry H. et al. Efficacy and safety of recently approved drugs for sickle cell disease: a review of clinical trials. Exp Hematol 2020; 92: 11-18.e1
  • 23 Pavan AR, Dos Santos JL. Advances in sickle cell disease treatments. Curr Med Chem 2021; 28 (10) 2008-2032
  • 24 Quinn CT. l-Glutamine for sickle cell anemia: more questions than answers. Blood 2018; 132 (07) 689-693
  • 25 Ballas SK. The evolving pharmacotherapeutic landscape for the treatment of sickle cell disease. Mediterr J Hematol Infect Dis 2020; 12 (01) e2020010
  • 26 Torres L, Conran N. Emerging pharmacotherapeutic approaches for the management of sickle cell disease. Expert Opin Pharmacother 2019; 20 (02) 173-186
  • 27 Ataga KI, Kutlar A, Kanter J. et al. Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med 2017; 376 (05) 429-439
  • 28 Gutsaeva DR, Parkerson JB, Yerigenahally SD. et al. Inhibition of cell adhesion by anti-P-selectin aptamer: a new potential therapeutic agent for sickle cell disease. Blood 2011; 117 (02) 727-735
  • 29 Matsui NM, Borsig L, Rosen SD, Yaghmai M, Varki A, Embury SH. P-selectin mediates the adhesion of sickle erythrocytes to the endothelium. Blood 2001; 98 (06) 1955-1962
  • 30 Hutchaleelaha A, Patel M, Washington C. et al. Pharmacokinetics and pharmacodynamics of voxelotor (GBT440) in healthy adults and patients with sickle cell disease. Br J Clin Pharmacol 2019; 85 (06) 1290-1302
  • 31 Chou ST. Transfusion therapy for sickle cell disease: a balancing act. Hematology Am Soc Hematol Educ Program 2013; 2013: 439-446
  • 32 Yawn BP, Buchanan GR, Afenyi-Annan AN. et al. Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. JAMA 2014; 312 (10) 1033-1048
  • 33 Adams RJ, McKie VC, Hsu L. et al. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography. N Engl J Med 1998; 339 (01) 5-11
  • 34 Bernaudin F, Socie G, Kuentz M. et al; SFGM-TC. Long-term results of related myeloablative stem-cell transplantation to cure sickle cell disease. Blood 2007; 110 (07) 2749-2756
  • 35 Vermylen C, Cornu G, Ferster A. et al. Haematopoietic stem cell transplantation for sickle cell anaemia: the first 50 patients transplanted in Belgium. Bone Marrow Transplant 1998; 22 (01) 1-6
  • 36 Gluckman E, Cappelli B, Bernaudin F. et al; Eurocord, the Pediatric Working Party of the European Society for Blood and Marrow Transplantation, and the Center for International Blood and Marrow Transplant Research. Sickle cell disease: an international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood 2017; 129 (11) 1548-1556
  • 37 Bhatia M, Sheth S. Hematopoietic stem cell transplantation in sickle cell disease: patient selection and special considerations. J Blood Med 2015; 6: 229-238
  • 38 Gluckman E. Allogeneic transplantation strategies including haploidentical transplantation in sickle cell disease. Hematology Am Soc Hematol Educ Program 2013; 2013: 370-376
  • 39 Shenoy S. Hematopoietic stem-cell transplantation for sickle cell disease: current evidence and opinions. Ther Adv Hematol 2013; 4 (05) 335-344
  • 40 Nickel RS, Flegel WA, Adams SD. et al. The impact of pre-existing HLA and red blood cell antibodies on transfusion support and engraftment in sickle cell disease after nonmyeloablative hematopoietic stem cell transplantation from HLA-matched sibling donors: a prospective, single-center, observational study. EClinicalMedicine 2020; 24: 100432
  • 41 Saraf SL, Oh AL, Patel PR. et al. Nonmyeloablative stem cell transplantation with alemtuzumab/low-dose irradiation to cure and improve the quality of life of adults with sickle cell disease. Biol Blood Marrow Transplant 2016; 22 (03) 441-448
  • 42 Guilcher GMT, Monagel DA, Nettel-Aguirre A. et al. Nonmyeloablative matched sibling donor hematopoietic cell transplantation in children and adolescents with sickle cell disease. Biol Blood Marrow Transplant 2019; 25 (06) 1179-1186
  • 43 McPherson ME, Anderson AR, Haight AE. et al. Transfusion management of sickle cell patients during bone marrow transplantation with matched sibling donor. Transfusion 2009; 49 (09) 1977-1986
  • 44 Kodish E, Lantos J, Stocking C, Singer PA, Siegler M, Johnson FL. Bone marrow transplantation for sickle cell disease. A study of parents' decisions. N Engl J Med 1991; 325 (19) 1349-1353
  • 45 Shenoy S, Eapen M, Panepinto JA. et al. A trial of unrelated donor marrow transplantation for children with severe sickle cell disease. Blood 2016; 128 (21) 2561-2567
  • 46 Krishnamurti L, Neuberg DS, Sullivan KM. et al. Bone marrow transplantation for adolescents and young adults with sickle cell disease: results of a prospective multicenter pilot study. Am J Hematol 2019; 94 (04) 446-454
  • 47 Kamani NR, Walters MC, Carter S. et al. Unrelated donor cord blood transplantation for children with severe sickle cell disease: results of one cohort from the phase II study from the Blood and Marrow Transplant Clinical Trials Network (BMT CTN). Biol Blood Marrow Transplant 2012; 18 (08) 1265-1272
  • 48 Abraham A, Cluster A, Jacobsohn D. et al. Unrelated umbilical cord blood transplantation for sickle cell disease following reduced-intensity conditioning: results of a phase I trial. Biol Blood Marrow Transplant 2017; 23 (09) 1587-1592
  • 49 Locatelli F, Kabbara N, Ruggeri A. et al; Eurocord and European Blood and Marrow Transplantation (EBMT) group. Outcome of patients with hemoglobinopathies given either cord blood or bone marrow transplantation from an HLA-identical sibling. Blood 2013; 122 (06) 1072-1078
  • 50 Fitzhugh CD, Hsieh MM, Taylor T. et al. Cyclophosphamide improves engraftment in patients with SCD and severe organ damage who undergo haploidentical PBSCT. Blood Adv 2017; 1 (11) 652-661
  • 51 Kurtzberg J. Update on umbilical cord blood transplantation. Curr Opin Pediatr 2009; 21 (01) 22-29
  • 52 Mehta RS, Dave H, Bollard CM, Shpall EJ. Engineering cord blood to improve engraftment after cord blood transplant. Stem Cell Investig 2017; 4 (06) 41
  • 53 Munoz J, Shah N, Rezvani K. et al. Concise review: umbilical cord blood transplantation: past, present, and future. Stem Cells Transl Med 2014; 3 (12) 1435-1443
  • 54 Foell J, Kleinschmidt K, Jakob M, Troeger A, Corbacioglu S. Alternative donor: αß/CD19 T-cell-depleted haploidentical hematopoietic stem cell transplantation for sickle cell disease. Hematol Oncol Stem Cell Ther 2020; 13 (02) 98-105
  • 55 de la Fuente J, Dhedin N, Koyama T. et al. Haploidentical bone marrow transplantation with post-transplantation cyclophosphamide plus thiotepa improves donor engraftment in patients with sickle cell anemia: results of an international learning collaborative. Biol Blood Marrow Transplant 2019; 25 (06) 1197-1209
  • 56 Gögel S, Gubernator M, Minger SL. Progress and prospects: stem cells and neurological diseases. Gene Ther 2011; 18 (01) 1-6
  • 57 Zhang GL, Zhu ZH, Wang YZ. Neural stem cell transplantation therapy for brain ischemic stroke: review and perspectives. World J Stem Cells 2019; 11 (10) 817-830
  • 58 Mcdonald-Hyman C, Turka LA, Blazar BR. Advances and challenges in immunotherapy for solid organ and bone marrow transplantation. Sci Transl Med 2015; 7 (280) 280rv2
  • 59 Strecker JK, Olk J, Hoppen M. et al. Combining growth factor and bone marrow cell therapy induces bleeding and alters immune response after stroke in mice. Stroke 2016; 47 (03) 852-862
  • 60 Broughton BR, Lim R, Arumugam TV, Drummond GR, Wallace EM, Sobey CG. Post-stroke inflammation and the potential efficacy of novel stem cell therapies: focus on amnion epithelial cells. Front Cell Neurosci 2013; 6: 66
  • 61 Carson MJ, Doose JM, Melchior B, Schmid CD, Ploix CC. CNS immune privilege: hiding in plain sight. Immunol Rev 2006; 213 (01) 48-65
  • 62 Galea I, Bechmann I, Perry VH. What is immune privilege (not)?. Trends Immunol 2007; 28 (01) 12-18
  • 63 Newman MB, Misiuta I, Willing AE. et al. Tumorigenicity issues of embryonic carcinoma-derived stem cells: relevance to surgical trials using NT2 and hNT neural cells. Stem Cells Dev 2005; 14 (01) 29-43
  • 64 Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced pluripotent stem cells. Nature 2011; 474 (7350): 212-215
  • 65 Ben-David U, Benvenisty N. The tumorigenicity of human embryonic and induced pluripotent stem cells. Nat Rev Cancer 2011; 11 (04) 268-277
  • 66 Blum B, Benvenisty N. The tumorigenicity of human embryonic stem cells. Adv Cancer Res 2008; 100: 133-158
  • 67 Menendez S, Camus S, Herreria A. et al. Increased dosage of tumor suppressors limits the tumorigenicity of iPS cells without affecting their pluripotency. Aging Cell 2012; 11 (01) 41-50
  • 68 Lawrenz B, Schiller H, Willbold E, Ruediger M, Muhs A, Esser S. Highly sensitive biosafety model for stem-cell-derived grafts. Cytotherapy 2004; 6 (03) 212-222