Elsevier

Blood Cells, Molecules, and Diseases

Volume 67, September 2017, Pages 155-168
Blood Cells, Molecules, and Diseases

Curative approaches for sickle cell disease: A review of allogeneic and autologous strategies

https://doi.org/10.1016/j.bcmd.2017.08.014Get rights and content

Abstract

Despite sickle cell disease (SCD) first being reported > 100 years ago and molecularly characterized > 50 years ago, patients continue to experience severe morbidity and early mortality. Although there have been substantial clinical advances with immunizations, penicillin prophylaxis, hydroxyurea treatment, and transfusion therapy, the only cure that can be offered is hematopoietic stem cell transplantation (HSCT). In this work, we summarize the various allogeneic curative approaches reported to date and discuss open and upcoming clinical research protocols. Then we consider gene therapy and gene editing strategies that may enable cure based on autologous HSCs.

Introduction

All curative therapies depend upon engraftment of hematopoietic cells. In this review we use “cure” to mean durable modification of hematopoietic stem cells in a manner to prevent clinical manifestations of SCD. Cure could be achieved by transplant of HSCs from a healthy donor, addition of an anti-sickling globin gene to patient HSCs, repair of the sickle cell mutation itself, or induction of fetal hemoglobin (HbF) to levels that prevent sickling. We discuss each of these approaches in detail below (Fig. 1). Various intensities of pre-transplantation conditioning therapies may be given to promote hematopoietic cell engraftment accompanied by an array of regimens to limit the risk of graft-versus-host disease (GvHD) that is mediated by allogeneic immune cells. With myeloablative protocols, total body irradiation (TBI) and/or chemotherapy is given at doses which would not allow autologous hematopoietic recovery. Nonmyeloablative regimens rely on intensive immunosuppressive conditioning along with high numbers of donor CD34 + cells and donor T lymphocytes to enable engraftment. Reduced intensity regimens deliver an intermediate level of conditioning. Investigational targeted therapies suggest the future potential to “make space” in the bone marrow for HSC engraftment without the risks of conventional chemotherapy or radiotherapy [1], [2], [3].

Section snippets

HLA-matched sibling donors

The first successful report of HSCT in SCD was in 1984, a patient transplanted for AML [4]. Since then, myeloablative conditioning, including busulfan and cyclophosphamide, have become established as the standard approach for pediatric patients with severe disease [5]. The French group reported a decrease in graft rejection rate from 22.6% to 2.9% with the addition of anti-thymocyte globulin (ATG) and an impressive event-free survival (EFS) of 95% [6]. ATG use is now commonplace. Published data

Hematopoietic stem cell based gene therapy

Since the first successful gene therapy was reported more than a decade ago, the field has rapidly evolved [44]. During the first decade of the millennium, hematopoietic cell based gene therapies for several life threatening immunodeficiencies using γ-retroviral vectors led to mixed results (for X-linked severe combined immunodeficiency, adenosine deaminase-deficient severe combined immunodeficiency, Wiskott-Aldrich syndrome, and X-linked chronic granulomatous disease) [44], [45], [46], [47].

Gene editing

Genome editing technology enables the production of specified changes to DNA sequences within cells. Diseases with a genetic etiology like SCD could be fundamentally addressed by genome editing. In this section we consider the current landscape of genome editing technologies and review therapeutic approaches relevant to SCD, including key parameters for success, areas of challenge, and anticipated future directions for the field.

Since the discovery of the molecular basis of SCD as a single

Conclusion

As curative therapies for SCD advance, a moral imperative will be to extend these gains to as many patients as possible, including those in Africa, where the worldwide burden of SCD is greatest. Major areas of future research will be how to simplify the procedures for allogeneic HSCT, viral gene therapy, and genome editing. Advances in HSC procurement, ex vivo manipulation of pluripotent and hematopoietic stem cells, and conditioning therapy could help extend curative therapies. Alternatively,

Acknowledgements

D.E.B. was supported by NIDDK (K08DK093705, R03DK109232), NHLBI (DP2OD022716), Burroughs Wellcome Fund (1014696), American Society of Hematology, Harvard Stem Cell Institute, and the Doris Duke Charitable Foundation (2015134), Charles H. Hood Foundation, and Cooley's Anemia Foundation. C.B. was supported by NIH grant HL117720-03. C.D.F. was supported by the intramural program of the National Heart, Lung, and Blood Institute, NIH.

References (146)

  • F. Locatelli et al.

    Outcome of patients with hemoglobinopathies given either cord blood or bone marrow transplantation from an HLA-identical sibling

    Blood

    (Aug 8 2013)
  • F. Locatelli et al.

    Related umbilical cord blood transplantation in patients with thalassemia and sickle cell disease

    Blood

    (Mar 15 2003)
  • A. Ruggeri et al.

    Umbilical cord blood transplantation for children with thalassemia and sickle cell disease

    Biol. Blood Marrow Transplant.

    (Sep 2011)
  • N.R. Kamani 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.

    (Aug 2012)
  • S. Shenoy et al.

    A BMT CTN phase II trial of unrelated donor marrow transplantation for children with severe sickle cell disease

    Blood

    (Sep 13 2016)
  • C.T. Quinn et al.

    Improved survival of children and adolescents with sickle cell disease

    Blood

    (Apr 29 2010)
  • J. Bolanos-Meade et al.

    HLA-haploidentical bone marrow transplantation with posttransplant cyclophosphamide expands the donor pool for patients with sickle cell disease

    Blood

    (Nov 22 2012)
  • A.T. Munchel et al.

    Treatment of hematological malignancies with nonmyeloablative, HLA-haploidentical bone marrow transplantation and high dose, post-transplantation cyclophosphamide

    Best Pract. Res. Clin. Haematol.

    (Sep 2011)
  • L. Luznik et al.

    HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide

    Biol. Blood Marrow Transplant.

    (Jun 2008)
  • M.H. Dallas et al.

    Long-term outcome and evaluation of organ function in pediatric patients undergoing haploidentical and matched related hematopoietic cell transplantation for sickle cell disease

    Biol. Blood Marrow Transplant.

    (May 2013)
  • C.D. Fitzhugh et al.

    Granulocyte colony-stimulating factor (G-CSF) administration in individuals with sickle cell disease: time for a moratorium?

    Cytotherapy

    (2009)
  • U. Modlich et al.

    Insertional transformation of hematopoietic cells by self-inactivating lentiviral and gammaretroviral vectors

    Mol. Ther.

    (Nov 2009)
  • D. Zychlinski et al.

    Physiological promoters reduce the genotoxic risk of integrating gene vectors

    Mol. Ther.

    (Apr 2008)
  • P.W. Hargrove et al.

    Globin lentiviral vector insertions can perturb the expression of endogenous genes in beta-thalassemic hematopoietic cells

    Mol. Ther.

    (Mar 2008)
  • G. Maruggi et al.

    Transcriptional enhancers induce insertional gene deregulation independently from the vector type and design

    Mol. Ther.

    (May 2009)
  • P.I. Arumugam et al.

    Genotoxic potential of lineage-specific lentivirus vectors carrying the beta-globin locus control region

    Mol. Ther.

    (Nov 2009)
  • F.R. Santoni de Sio et al.

    Proteasome activity restricts lentiviral gene transfer into hematopoietic stem cells and is down-regulated by cytokines that enhance transduction

    Blood

    (Jun 1 2006)
  • E.S. Rosler et al.

    An in vivo competitive repopulation assay for various sources of human hematopoietic stem cells

    Blood

    (Nov 15 2000)
  • N. Psatha et al.

    Optimizing autologous cell grafts to improve stem cell gene therapy

    Exp. Hematol.

    (Jul 2016)
  • D.A. Williams

    Broadening the indications for hematopoietic stem cell genetic therapies

    Cell Stem Cell

    (Sep 5 2013)
  • D.N. Levasseur et al.

    Correction of a mouse model of sickle cell disease: lentiviral/antisickling beta-globin gene transduction of unmobilized, purified hematopoietic stem cells

    Blood

    (Dec 15 2003)
  • F. Grosveld et al.

    Position-independent, high-level expression of the human beta-globin gene in transgenic mice

    Cell

    (1987)
  • J. Kanter et al.

    Initial results from study Hgb-206: a phase 1 study evaluating gene therapy by transplantation of autologous CD34 + stem cells transduced ex vivo with the lentiglobin BB305 lentiviral vector in subjects with severe sickle cell disease

    Blood

    (2015)
  • P. Leboulch

    Bench to bedside-gene therapy for thalassemia and sickle cell disease

    Blood

    (2015)
  • M. Cavazzana et al.

    Outcomes of gene therapy for severe sickle disease and beta-thalassemia major via transplantation of autologous hematopoietic stem cells transduced ex vivo with a lentiviral beta AT87Q-globin vector

    Blood

    (2015)
  • F. Urbinati et al.

    Potentially therapeutic levels of anti-sickling globin gene expression following lentivirus-mediated gene transfer in sickle cell disease bone marrow CD34 + cells

    Exp. Hematol.

    (May 2015)
  • A. Perumbeti et al.

    A novel human gamma-globin gene vector for genetic correction of sickle cell anemia in a humanized sickle mouse model: critical determinants for successful correction

    Blood

    (Aug 6 2009)
  • P. Malik

    Gene therapy for hemoglobinopathies: tremendous successes and remaining caveats

    Mol. Ther.

    (Apr 2016)
  • A.P. Funnell et al.

    2p15–p16.1 microdeletions encompassing and proximal to BCL11A are associated with elevated HbF in addition to neurologic impairment

    Blood

    (Jul 2 2015)
  • C. Dias et al.

    BCL11A haploinsufficiency causes an intellectual disability syndrome and dysregulates transcription

    Am. J. Hum. Genet.

    (Aug 4 2016)
  • A. Czechowicz et al.

    Efficient transplantation via antibody-based clearance of hematopoietic stem cell niches

    Science

    (Nov 23 2007)
  • R. Palchaudhuri et al.

    Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin

    Nat. Biotechnol.

    (Jul 2016)
  • F.L. Johnson et al.

    3rd. Bone-marrow transplantation in a patient with sickle-cell anemia

    N. Engl. J. Med.

    (Sep 20 1984)
  • A.A. King et al.

    Successful matched sibling donor marrow transplantation following reduced intensity conditioning in children with hemoglobinopathies

    Am. J. Hematol.

    (Sep 8 2015)
  • M. Bhatia et al.

    Reduced toxicity, myeloablative conditioning with BU, fludarabine, alemtuzumab and SCT from sibling donors in children with sickle cell disease

    Bone Marrow Transplant.

    (May 5 2014)
  • S. Matthes-Martin et al.

    Stem cell transplantation after reduced-intensity conditioning for sickle cell disease

    Eur. J. Haematol.

    (Apr 2013)
  • M.M. Hsieh et al.

    Nonmyeloablative hla-matched sibling allogeneic hematopoietic stem cell transplantation for severe sickle cell phenotype

    JAMA

    (2014)
  • S.L. Saraf 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.

    (Sep 5 2015)
  • M.C. Walters et al.

    Barriers to bone marrow transplantation for sickle cell anemia

    Biol. Blood Marrow Transplant.

    (1996)
  • E. Gluckman et al.

    Hematopoietic reconstitution in a patient with Fanconi's anemia by means of umbilical-cord blood from an HLA-identical sibling

    N. Engl. J. Med.

    (Oct 26 1989)
  • Cited by (10)

    • In Vivo Outcome of Homology-Directed Repair at the HBB Gene in HSC Using Alternative Donor Template Delivery Methods

      2019, Molecular Therapy Nucleic Acids
      Citation Excerpt :

      Patients with SCD have increased morbidity and a reduced lifespan.1–4 While curative treatment can be achieved through human leukocyte antigen (HLA)-matched allogeneic transplant from a healthy donor, the availability of HLA-matched donors is limited, and the outcomes are complicated by the possibility of graft-versus-host disease (GvHD) and short-term and long-term impacts following higher intensity myelo-ablative conditioning.5–8 Gene editing in autologous stem cells could circumvent the limitation of HLA-matched donor availability, directly correcting the disease-causing mutation in self-renewing stem cells and mitigating the historical risk of random integration posed by early viral vectors.9–13

    • The in vitro growth of a cord blood–derived cell population enriched for CD34<sup>+</sup> cells is influenced by its cell cycle status and treatment with hydroxyurea

      2018, Cytotherapy
      Citation Excerpt :

      It is noteworthy that positive effects of HU on the E arm of hematopoiesis have long been described. In fact, HU has been widely used for the treatment of sickle cell disease [41], and it has been shown to inhibit parvovirus B19 infection of E cells [42]. Interestingly, in the present study, HU also increased CFC frequency as well as the proliferation and expansion capacities of CD34+ cells in cultures under M conditions.

    View all citing articles on Scopus
    View full text