Plerixafor enables safe, rapid, efficient mobilization of hematopoietic stem cells in sickle cell disease patients after exchange transfusion

Chantal Lagresle-Peyrou, François Lefrère, Elisa Magrin, Jean-Antoine Ribeil, Oriana Romano, Leslie Weber, Alessandra Magnani, Hanem Sadek, Clémence Plantier, Aurélie Gabrion, Brigitte Ternaux, Tristan Félix, Chloé Couzin, Aurélie Stanislas, Jean-Marc Tréluyer, Lionel Lamhaut, Laure Joseph, Marianne Delville, Annarita Miccio, Isabelle André-Schmutz, Marina Cavazzana, Chantal Lagresle-Peyrou, François Lefrère, Elisa Magrin, Jean-Antoine Ribeil, Oriana Romano, Leslie Weber, Alessandra Magnani, Hanem Sadek, Clémence Plantier, Aurélie Gabrion, Brigitte Ternaux, Tristan Félix, Chloé Couzin, Aurélie Stanislas, Jean-Marc Tréluyer, Lionel Lamhaut, Laure Joseph, Marianne Delville, Annarita Miccio, Isabelle André-Schmutz, Marina Cavazzana

Abstract

Sickle cell disease is characterized by chronic anemia and vaso-occlusive crises, which eventually lead to multi-organ damage and premature death. Hematopoietic stem cell transplantation is the only curative treatment but it is limited by toxicity and poor availability of HLA-compatible donors. A gene therapy approach based on the autologous transplantation of lentiviral-corrected hematopoietic stem and progenitor cells was shown to be efficacious in one patient. However, alterations of the bone marrow environment and properties of the red blood cells hamper the harvesting and immunoselection of patients' stem cells from bone marrow. The use of Filgrastim to mobilize large numbers of hematopoietic stem and progenitor cells into the circulation has been associated with severe adverse events in sickle cell patients. Thus, broader application of the gene therapy approach requires the development of alternative mobilization methods. We set up a phase I/II clinical trial whose primary objective was to assess the safety of a single injection of Plerixafor in sickle cell patients undergoing red blood cell exchange to decrease the hemoglobin S level to below 30%. The secondary objective was to measure the efficiency of mobilization and isolation of hematopoietic stem and progenitor cells. No adverse events were observed. Large numbers of CD34+ cells were mobilized extremely quickly. Importantly, the mobilized cells contained high numbers of hematopoietic stem cells, expressed high levels of stemness genes, and engrafted very efficiently in immunodeficient mice. Thus, Plerixafor can be safely used to mobilize hematopoietic stem cells in sickle cell patients; this finding opens up new avenues for treatment approaches based on gene addition and genome editing. Clinicaltrials.gov identifier: NCT02212535.

Copyright © 2018 Ferrata Storti Foundation.

Figures

Figure 1.
Figure 1.
Plerixafor is highly efficient at mobilizing hematopoietic stem and progenitor cells from sickle cell disease patients. (A) Changes in white blood cell (WBC) and (B) CD34+ hematopoietic stem/progenitor cell (HSPC) counts over the 66 h following Plerixafor administration in SCD Pler 1 (red squares), SCD Pler 2 (blue circles) and SCD Pler 3 (green triangles). Arrows indicate time and duration of apheresis. (C) Number of hematopoietic stem cells (HSC, black bars) and multipotent progenitors (MPP, dotted bars) per 1,000 CD34+ cells in samples of various origins. HD: healthy donor, BM: bone marrow, SCD: sickle cell disease, Pler: Plerixafor, Filg: Filgrastim.
Figure 2.
Figure 2.
Analysis of the transcriptomic profiles of hematopoietic stem and progenitor cells from different sources (A) Hierarchical clustering analysis of HD BM, SCD BM, SCD Plerixafor-mobilized (Pler), HD Plerixafor-mobilized and HD Filgrastim-mobilized (Filg) HSPC (cluster method: average; distance: correlation). The color of the sample name indicates the classification. (B) Gene ontology analysis of differentially expressed genes. The most enriched biological process categories are shown on the y-axis. The x-axis shows sample comparisons, as defined in Table 3. The orange and green color gradients correspond to the statistical significance of the enrichment [expressed as –log10 (qvalue)] in up- and downregulated genes, respectively. The first color bar at the top indicates comparisons between HSPC from different types of source (dark red) or the same type of source (light red). The second color bar at the top indicates comparisons between HSPC from different types of donor (dark blue) or the same type of donor (light blue). (C) Heat map of genes involved in HSC and progenitor biology. A proportion of the HSC markers were highly expressed in SCD Plerixafor-mobilized HSPC compared with the other samples. The row Z-score is plotted on a red-blue color scale, where red indicates high expression and blue indicates low expression. The color bar at the top indicates the sample classification. HD: healthy donor; BM: bone marrow; HSPC: hematopoietic stem and progenitor cells; HSC: hematopoietic stem cells; SCD: sickle cell disease; Pler: Plerixafor; Filg: Filgrastim.
Figure 3.
Figure 3.
Plerixafor-mobilized CD34+ cells from sickle cell disease patients engraft to the same degree as Filgrastim-mobilized CD34+ cells from healthy donors in NSG mice. NSG mice were sacrificed 3 to 4 months after the injection of SCD (SCD Plerixafor, n=3) or HD (HD Filgrastim, n=2) CD34+ cells. (A) Bone marrow cells and (B) splenocytes were isolated, stained and analyzed by flow cytometry. The chimerism (defined as % human CD45+cells/total CD45+cells) and the numbers of human B lymphocytes (CD19+IgM+), granulocytes (CD11b+CD15+), and monocytes (CD11b+CD14+) were evaluated in each group of mice (red circles and red triangles SCD Pler1; blue circles and blue triangles SCD Pler2; green circles and green triangles SCD Pler3; the two HD Filg control are represented by gray squares/gray diamond and black squares/black diamonds, respectively). Each dot represents an individual mouse. HD: healthy donor; SCD: sickle cell disease; Pler: Plerixafor; Filg: Filgrastim.

References

    1. Ware RE, de Montalembert M, Tshilolo L, Abboud MR. Sickle cell disease. Lancet. 2017;390(10091):311–323.
    1. Walters MC, De Castro LM, Sullivan KM, et al. Indications and results of HLA-identical sibling hematopoietic cell transplantation for sickle cell disease. Biol Blood Marrow Transplant. 2016;22(2):207–211.
    1. Gluckman E, Cappelli B, Bernaudin F, et al. Sickle cell disease: an international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood. 2017;129(11):1548–1556.
    1. Ribeil JA, Hacein-Bey-Abina S, Payen E, et al. Gene therapy in a patient with sickle cell disease. N Engl J Med. 2017;376(9): 848–855.
    1. Cavazzana M, Ribeil JA, Lagresle-Peyrou C, Andre-Schmutz I. Gene therapy with hematopoietic stem cells: the diseased bone marrow’s point of view. Stem Cells Dev. 2017;26(2):71–76.
    1. Wang TF, Chen SH, Yang SH, Su YC, Chu SC, Li DK. Poor harvest of peripheral blood stem cell in donors with microcytic red blood cells. Transfusion. 2013;53(1):91–95.
    1. Constantinou VC, Bouinta A, Karponi G, et al. Poor stem cell harvest may not always be related to poor mobilization: lessons gained from a mobilization study in patients with beta-thalassemia major. Transfusion. 2017;57(4):1031–1039.
    1. Abboud M, Laver J, Blau CA. Granulocytosis causing sickle-cell crisis. Lancet. 1998;351(9107):959.
    1. Adler BK, Salzman DE, Carabasi MH, Vaughan WP, Reddy VV, Prchal JT. Fatal sickle cell crisis after granulocyte colony- stimulating factor administration. Blood. 2001;97(10):3313–3314.
    1. Grigg AP. Granulocyte colony-stimulating factor-induced sickle cell crisis and multiorgan dysfunction in a patient with compound heterozygous sickle cell/beta+ thalassemia. Blood. 2001;97(12):3998–3999.
    1. Onitilo AA, Lazarchick J, Brunson CY, Frei-Lahr D, Stuart RK. Autologous bone marrow transplant in a patient with sickle cell disease and diffuse large B-cell lymphoma. Transplant Proc. 2003;35(8):3089–3092.
    1. Kamble RT, Tin UC, Carrum G. Successful mobilization and transplantation of filgrastim mobilized hematopoietic stem cells in sickle cell-hemoglobin C disease. Bone Marrow Transplant. 2006;37(11):1065–1066.
    1. Rosenbaum C, Peace D, Rich E, Van Besien K. Granulocyte colony-stimulating factor- based stem cell mobilization in patients with sickle cell disease. Biol Blood Marrow Transplant. 2008;14(6):719–723.
    1. Tormey CA, Snyder EL, Cooper DL. Mobilization, collection, and transplantation of peripheral blood hematopoietic progenitor cells in a patient with multiple myeloma and hemoglobin SC disease. Transfusion. 2008;48(9):1930–1933.
    1. Liles WC, Broxmeyer HE, Rodger E, et al. Mobilization of hematopoietic progenitor cells in healthy volunteers by AMD3100, a CXCR4 antagonist. Blood. 2003;102(8): 2728–2730.
    1. Schroeder MA, Rettig MP, Lopez S, et al. Mobilization of allogeneic peripheral blood stem cell donors with intravenous plerixafor mobilizes a unique graft. Blood. 2017;129(19):2680–2692.
    1. Broxmeyer HE, Orschell CM, Clapp DW, et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med. 2005;201(8):1307–1318.
    1. Devine SM, Flomenberg N, Vesole DH, et al. Rapid mobilization of CD34+ cells following administration of the CXCR4 antagonist AMD3100 to patients with multiple myeloma and non-Hodgkin’s lymphoma. J Clin Oncol. 2004;22(6):1095–1102.
    1. Karponi G, Psatha N, Lederer CW, et al. Plerixafor+G-CSF-mobilized CD34+ cells represent an optimal graft source for thalassemia gene therapy. Blood. 2015;126(5): 616–619.
    1. Ferrari G, Cavazzana M, Mavilio F. Gene therapy approaches to hemoglo binopathies. Hematol Oncol Clin North Am. 2017;31(5): 835–852.
    1. Lionnet F, Arlet JB, Bartolucci P, et al. [Guidelines for management of adult sickle cell disease]. Rev Med Interne. 2009;30(Suppl 3):S162–223.
    1. Lamming CE, Augustin L, Blackstad M, Lund TC, Hebbel RP, Verfaillie CM. Spontaneous circulation of myeloid-lymphoid-initiating cells and SCID-repopulating cells in sickle cell crisis. J Clin Invest. 2003;111(6):811–819.
    1. Doulatov S, Notta F, Eppert K, Nguyen LT, Ohashi PS, Dick JE. Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development. Nat Immunol. 2010;11(7):585–593.
    1. Six EM, Bonhomme D, Monteiro M, et al. A human postnatal lymphoid progenitor capable of circulating and seeding the thymus. J Exp Med. 2007;204(13):3085–3093.
    1. Handgretinger R, Kuci S. CD133-positive hematopoietic stem cells: from biology to medicine. Adv Exp Med Biol. 2013;777:99–111.
    1. Fitzhugh CD, Hsieh MM, Bolan CD, Saenz C, Tisdale JF. Granulocyte colony-stimulating factor (G-CSF) administration in individuals with sickle cell disease: time for a moratorium? Cytotherapy. 2009;11(4):464–471.
    1. Yannaki E, Papayannopoulou T, Jonlin E, et al. Hematopoietic stem cell mobilization for gene therapy of adult patients with severe beta-thalassemia: results of clinical trials using G-CSF or plerixafor in splenectomized and nonsplenectomized subjects. Mol Ther. 2012;20(1):230–238.
    1. Richard RE, Siritanaratkul N, Jonlin E, Skarpidi E, Heimfeld S, Blau CA. Collection of blood stem cells from patients with sickle cell anemia. Blood Cells Mol Dis. 2005;35(3):384–388.
    1. Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med. 1995;332(20):1317–1322.
    1. Alvarez-Larran A, Perez-Encinas M, Ferrer- Marin F, et al. Risk of thrombosis according to need of phlebotomies in patients with polycythemia vera treated with hydroxyurea. Haematologica. 2017;102(1):103–109.
    1. Halter J, Kodera Y, Ispizua AU, et al. Severe events in donors after allogeneic hematopoietic stem cell donation. Haematologica. 2009;94(1):94–101.
    1. Lack NA, Green B, Dale DC, et al. A pharmacokinetic-pharmacodynamic model for the mobilization of CD34+ hematopoietic progenitor cells by AMD3100. Clin Pharmacol Ther. 2005;77(5):427–436.
    1. To LB, Levesque JP, Herbert KE. How I treat patients who mobilize hematopoietic stem cells poorly. Blood. 2011;118(17):4530–4540.
    1. Lefrere F, Mauge L, Rea D, et al. A specific time course for mobilization of peripheral blood CD34+ cells after plerixafor injection in very poor mobilizer patients: impact on the timing of the apheresis procedure. Transfusion. 2013;53(3):564–569.
    1. Pantin J, Purev E, Tian X, et al. Effect of high-dose plerixafor on CD34(+) cell mobilization in healthy stem cell donors: results of a randomized crossover trial. Haematologica. 2017;102(3):600–609.
    1. Weisser M, Demel UM, Stein S, et al. Hyperinflammation in patients with chronic granulomatous disease leads to impairment of hematopoietic stem cell functions. J Allergy Clin Immunol. 2016;138(1):219–228.e219.

Source: PubMed

3
Subscribe