Correction of beta-thalassemia major by gene transfer in haematopoietic progenitors of pediatric patients

Emanuela Anna Roselli, Riccardo Mezzadra, Marta Claudia Frittoli, Giulietta Maruggi, Erika Biral, Fulvio Mavilio, Fabrizio Mastropietro, Antonio Amato, Giovanni Tonon, Chiara Refaldi, Maria Domenica Cappellini, Marco Andreani, Guido Lucarelli, Maria Grazia Roncarolo, Sarah Marktel, Giuliana Ferrari, Emanuela Anna Roselli, Riccardo Mezzadra, Marta Claudia Frittoli, Giulietta Maruggi, Erika Biral, Fulvio Mavilio, Fabrizio Mastropietro, Antonio Amato, Giovanni Tonon, Chiara Refaldi, Maria Domenica Cappellini, Marco Andreani, Guido Lucarelli, Maria Grazia Roncarolo, Sarah Marktel, Giuliana Ferrari

Abstract

Beta-thalassemia is a common monogenic disorder due to mutations in the beta-globin gene and gene therapy, based on autologous transplantation of genetically corrected haematopoietic stem cells (HSCs), holds the promise to treat patients lacking a compatible bone marrow (BM) donor. We recently showed correction of murine beta-thalassemia by gene transfer in HSCs with the GLOBE lentiviral vector (LV), expressing a transcriptionally regulated human beta-globin gene. Here, we report successful correction of thalassemia major in human cells, by studying a large cohort of pediatric patients of diverse ethnic origin, carriers of different mutations and all candidates to BM transplantation. Extensive characterization of BM-derived CD34(+) cells before and following gene transfer shows the achievement of high frequency of transduction, restoration of haemoglobin A synthesis, rescue from apoptosis and correction of ineffective erythropoiesis. The procedure does not significantly affect the differentiating potential and the relative proportion of haematopoietic progenitors. Analysis of vector integrations shows preferential targeting of transcriptionally active regions, without bias for cancer-related genes. Overall, these results provide a solid rationale for a future clinical translation.

Figures

Figure 1. Characterization of thalassemia major BM-CD34…
Figure 1. Characterization of thalassemia major BM-CD34+ cells
  1. Expression of erythroid lineage markers in BM-CD34+ progenitors: cumulative FACS data of the percentage of CD34+GpA+, CD34+CD36+ and CD34+CD71+ cells plotted for normal (ND, n = 5, 5 and 3, respectively) and thalassemic (THAL, n = 17, 8 and 9, respectively) samples.

  2. CFU activity of CD34+ cells analysed as total number of colonies/1000 cells plated and as proportion of the different type of colonies (BFU-E, CFU-GM and CFU-GEMM) in ND (n = 12) and THAL (n = 20) samples. Values are given as mean ± SEM. Asterisks indicate values significantly different between groups (***p < 0.001).

Figure 2. Effect of cytokines treatment on…
Figure 2. Effect of cytokines treatment on clonogenic activity and progenitors subpopulations of CD34+ cells
  1. CFU activity of CD34+ cells and proportion of the different type of colonies (BFU-E, CFU-GM and CFU-GEMM) in thalassemia major samples untreated (THAL, n = 20), activated with cytokines (THAL-act, n = 19) and transduced with the GLOBE vector (THAL-GLOBE, n = 18).

  2. Cumulative data from FACS analysis of multipotent (CD34+CD38−CD50+), lymphoid (CD34+CD10+) and myeloid (CD34+CD33+) progenitors subpopulations in thalassemia major untreated (THAL, black bars, n = 3, 6 and 6, respectively) and activated (THAL-act, white bars, n = 3) samples. Values represent the mean ± SEM. Asterisks indicate parameters significantly different between groups (*p < 0.05; **p < 0.01).

Figure 3. Genome expression profiling of thalassemic…
Figure 3. Genome expression profiling of thalassemic CD34+ cells and molecular response to cytokine activation
  1. Unsupervised cluster analysis of transcripts from thalassemic and normal cells untreated (THAL, n = 9; ND, n = 8) and activated with cytokines (THAL-act, n = 11; ND-act, n = 9).

  2. Unsupervised cluster analysis of transcripts from thalassemic and normal cells, untreated and activated with cytokines, including normal donor pediatric cells (ND-P, n = 5). In red overexpressed probe sets, in blue downregulated ones.

Figure 4. Transduction efficiency of thalassemia major…
Figure 4. Transduction efficiency of thalassemia major CD34+ cells by GLOBE vector and correction of HbA deficiency in transduced erythroblasts
  1. Schematic representation of the percentage of transduction as determined by PCR on CFUs derived from transduced CD34+ cells, and of the average VCN/cell, as determined by qPCR on bulk erythroid culture. Single dots and triangles represent individual experiments.

  2. Left: Representative FACS analysis for HbA expression at 2 weeks of erythroid culture from normal (ND1), thalassemic (THAL 106, 51) and thalassemic GLOBE-transduced (THAL 106-GLOBE, 51-GLOBE) CD34+ cells after intracellular HbA staining. Right: Cumulative data from FACS analysis for HbA expression in erythroid cells. In the upper panel, the mean proportion ± SEM of HbA+ cells in normal (ND, black bar, n = 5), thalassemia-β0 (THAL-β0, white bar, n = 7) and thalassemia-β0-GLOBE-transduced (THAL-β0-GLOBE, grey bar, n = 7) cultures. In the lower panel, the MFI of HbA-expressing cells in the normal (ND, black bar, n = 5), thalassemic (THAL-β0 and -β+, white bars, n = 7 and 3) and thalassemic GLOBE-transduced (THAL-β0 and -β+, grey bars, n = 7 and 3) cultures. Asterisks indicate parameters significantly different between groups (*p < 0.05; **p < 0.01; ***p < 0.001).

Figure 5. Reverse phase HPLC analysis of…
Figure 5. Reverse phase HPLC analysis of radiolabelled globin chains synthetized in erythroblastic culture
HPLC profile of globin chains obtained from lysates of 15 × 106 erythroid cells after 14 days of liquide culture started from normal (ND1 and ND2), thalassemic (THAL 35 = THAL-β0 and THAL 51 = THAL-β+) and thalassemic GLOBE-transduced (THAL β0-GLOBE or THAL β+-GLOBE) BM CD34+ cells after metabolic incorporation of radiolabelled leucine. Neo-synthetized single globin chains (α, β, δ, Aγ and Gγ) are indicated on each corresponding peak.
Figure 6. Correction of ineffective erythropoiesis
Figure 6. Correction of ineffective erythropoiesis
  1. Photographs of May Grünwald–Giemsa-stained cytospin of erythroid cells from normal donor (ND), β-thalassemia (THAL) and GLOBE-transduced β-thalassemia (THAL-GLOBE) samples after 2 weeks of erythroid liquid culture (Microscope: Olympus Provis AX70. Objective: 100×/1.30 oil).

  2. Percentage cell number for each erythroblasts population after differential counting. ND n = 4, THAL n = 7, THAL-GLOBE n = 7.

  3. Representative FACS analysis of GpA+ erythroid cells for apoptotic/dead cells using Annexin V or propidium iodide labelling. Percentage of double positive cells on the total is reported in the panel.

  4. Percentage of apoptotic (GpA+ Annexin V+) or late apoptotic/dead (GpA+ PI+) erythroid cells at day 14 of culture. ND n = 2, THAL n = 3, THAL-GLOBE n = 3 THAL vs. THAL-GLOBE; *p < 0.05.

Figure 7. Distribution of the GLOBE vector…
Figure 7. Distribution of the GLOBE vector integration sites in the human genome and correlation with gene activity in CD34+ cells
  1. GLOBE vector integration sites (red bars) in the genome of BM-CD34+ cells from thalassemic patients. LV-CMV-GFP integrations (LV-GFP) and random genomic sequences originated by LM-PCR (RANDOM), are used as control (green and blue bars, respectively). Intergenic, intragenic and TSS-proximal integrations are defined as represented in the scheme. Asterisks indicate frequency significantly different between groups (*p < 0.05; ***p < 0.001).

  2. Distribution of target genes in the different expression level categories. The first two bars (CHIP THAL and CHIP CD34+) show the distribution of the genes on the microarray of thalassemic and normal CD34+ cells activated with cytokines, the third bar (RANDOM) represents random control sequences, the fourth and fifth bars (LV-GFP and GLOBE) report the expression values of genes targeted by control LV-CMV-GFP and GLOBE integrations, respectively. The N-value indicates the number of probesets, the n values indicate the number of genes analysed.

References

    1. Aiuti A, Cattaneo F, Galimberti S, Benninghoff U, Cassani B, Callegaro L, Scaramuzza S, Andolfi G, Mirolo M, Brigida I, et al. Gene therapy for immunodeficiency due to adenosine deaminase deficiency. N Engl J Med. 2009;360:447–458.
    1. Andreani M, Nesci S, Lucarelli G, Tonucci P, Rapa S, Angelucci E, Persini B, Agostinelli F, Donati M, Manna M. Long-term survival of ex-thalassemic patients with persistent mixed chimerism after bone marrow transplantation. Bone Marrow Transplant. 2000;25:401–404.
    1. Andreani M, Testi M, Battarra M, Indigeno P, Guagnano A, Polchi P, Federici G, Lucarelli G. Relationship between mixed chimerism and rejection after bone marrow transplantation in thalassaemia. Blood Transfus. 2008;6:143–149.
    1. Arumugam PI, Urbinati F, Velu CS, Higashimoto T, Grimes HL, Malik P. The 3′ region of the chicken hypersensitive site-4 insulator has properties similar to its core and is required for full insulator activity. PLoS One. 2009;4:e6995.
    1. Bohmer RM. Flow cytometry of erythropoiesis in culture: bivariate profiles of fetal and adult hemoglobin. Methods Cell Biol. 2001;64:139–152.
    1. Borgna-Pignatti C, Cappellini MD, De Stefano P, Del Vecchio GC, Forni GL, Gamberini MR, Ghilardi R, Origa R, Piga A, Romeo MA, et al. Survival and complications in thalassemia. Ann N Y Acad Sci. 2005;1054:40–47.
    1. Bradford GB, Williams B, Rossi R, Bertoncello I. Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment. Exp Hematol. 1997;25:445–453.
    1. Buckner CD, Clift RA, Sanders JE, Stewart P, Bensinger WI, Doney KC, Sullivan KM, Witherspoon RP, Deeg HJ, Appelbaum FR, et al. Marrow harvesting from normal donors. Blood. 1984;64:630–634.
    1. Bushman F, Lewinski M, Ciuffi A, Barr S, Leipzig J, Hannenhalli S, Hoffmann C. Genome-wide analysis of retroviral DNA integration. Nat Rev Microbiol. 2005;3:848–858.
    1. Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G, Schmidt M, Kutschera I, Vidaud M, Abel U, Dal-Cortivo L, Caccavelli L, et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science. 2009;326:818–823.
    1. Cattoglio C, Facchini G, Sartori D, Antonelli A, Miccio A, Cassani B, Schmidt M, von Kalle C, Howe S, Thrasher AJ, et al. Hot spots of retroviral integration in human CD34+ hematopoietic cells. Blood. 2007;110:1770–1778.
    1. Cheshier SH, Morrison SJ, Liao X, Weissman IL. In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells. Proc Natl Acad Sci USA. 1999;96:3120–3125.
    1. Dai M, Wang P, Boyd AD, Kostov G, Athey B, Jones EG, Bunney WE, Myers RM, Speed TP, Akil H, et al. Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data. Nucleic Acids Res. 2005;33:e175.
    1. Felice B, Cattoglio C, Cittaro D, Testa A, Miccio A, Ferrari G, Luzi L, Recchia A, Mavilio F. Transcription factor binding sites are genetic determinants of retroviral integration in the human genome. PLoS One. 2009;4:e4571.
    1. Ferrari F, Bortoluzzi S, Coppe A, Sirota A, Safran M, Shmoish M, Ferrari S, Lancet D, Danieli GA, Bicciato S. Novel definition files for human GeneChips based on GeneAnnot. BMC Bioinformatics. 2007;8:446.
    1. Fritsch G, Stimpfl M, Kurz M, Printz D, Buchinger P, Fischmeister G, Hoecker P, Gadner H. The composition of CD34 subpopulations differs between bone marrow, blood and cord blood. Bone Marrow Transplant. 1996;17:169–178.
    1. Galanello R, Satta S, Pirroni MG, Travi M, Maccioni L. Globin chain synthesis analysis by high performance liquid chromatography in the screening of thalassemia syndromes. Hemoglobin. 1998;22:501–508.
    1. Gaspar HB, Parsley KL, Howe S, King D, Gilmour KC, Sinclair J, Brouns G, Schmidt M, Von Kalle C, Barington T, et al. Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector. Lancet. 2004;364:2181–2187.
    1. Giarratana MC, Kobari L, Lapillonne H, Chalmers D, Kiger L, Cynober T, Marden MC, Wajcman H, Douay L. Ex vivo generation of fully mature human red blood cells from hematopoietic stem cells. Nat Biotechnol. 2005;23:69–74.
    1. Giordano PC, Van Delft P, Batelaan D, Harteveld CL, Bernini LF. Haemoglobinopathy analyses in the Netherlands: a report of an in vitro globin chain biosynthesis survey using a rapid, modified method. Clin Lab Haematol. 1999;21:247–256.
    1. Hacein-Bey-Abina S, Le Deist F, Carlier F, Bouneaud C, Hue C, De Villartay JP, Thrasher AJ, Wulffraat N, Sorensen R, Dupuis-Girod S, et al. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. N Engl J Med. 2002;346:1185–1193.
    1. Hanawa H, Yamamoto M, Zhao H, Shimada T, Persons DA. Optimized lentiviral vector design improves titer and transgene expression of vectors containing the chicken beta-globin locus HS4 insulator element. Mol Ther. 2009;17:667–674.
    1. Kaiser J. β-Thalassemia treatment succeeds, with a caveat. Science. 2009;326:1468–1469.
    1. Kinniburgh D, Russell NH. Comparative study of CD34-positive cells and subpopulations in human umbilical cord blood and bone marrow. Bone Marrow Transplant. 1993;12:489–494.
    1. Li C, Wong WH. Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc Natl Acad Sci USA. 2001;98:31–36.
    1. Lucarelli G, Gaziev J. Advances in the allogeneic transplantation for thalassemia. Blood Rev. 2008;22:53–63.
    1. Malik P, Arumugam PI, Yee JK, Puthenveetil G. Successful correction of the human Cooley's anemia beta-thalassemia major phenotype using a lentiviral vector flanked by the chicken hypersensitive site 4 chromatin insulator. Ann N Y Acad Sci. 2005;1054:238–249.
    1. Mathias LA, Fisher TC, Zeng L, Meiselman HJ, Weinberg KI, Hiti AL, Malik P. Ineffective erythropoiesis in beta-thalassemia major is due to apoptosis at the polychromatophilic normoblast stage. Exp Hematol. 2000;28:1343–1353.
    1. Miccio A, Cesari R, Lotti F, Rossi C, Sanvito F, Ponzoni M, Routledge SJ, Chow CM, Antoniou MN, Ferrari G. In vivo selection of genetically modified erythroblastic progenitors leads to long-term correction of beta-thalassemia. Proc Natl Acad Sci USA. 2008;105:10547–10552.
    1. Migliaccio G, Di Pietro R, di Giacomo V, Di Baldassarre A, Migliaccio AR, Maccioni L, Galanello R, Papayannopoulou T. In vitro mass production of human erythroid cells from the blood of normal donors and of thalassemic patients. Blood Cells Mol Dis. 2002;28:169–180.
    1. Modell B, Darlison M. Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ. 2008;86:480–487.
    1. Nienhuis AW, Dunbar CE, Sorrentino BP. Genotoxicity of retroviral integration in hematopoietic cells. Mol Ther. 2006;13:1031–1049.
    1. Olweus J, Thompson PA, Lund-Johansen F. Granulocytic and monocytic differentiation of CD34hi cells is associated with distinct changes in the expression of the PU.1-regulated molecules, CD64 and macrophage colony-stimulating factor receptor. Blood. 1996;88:3741–3754.
    1. Puthenveetil G, Scholes J, Carbonell D, Qureshi N, Xia P, Zeng L, Li S, Yu Y, Hiti AL, Yee JK, et al. Successful correction of the human beta-thalassemia major phenotype using a lentiviral vector. Blood. 2004;104:3445–3453.
    1. Sadelain M, Boulad F, Galanello R, Giardina P, Locatelli F, Maggio A, Rivella S, Riviere I, Tisdale J. Therapeutic options for patients with severe beta-thalassemia: the need for globin gene therapy. Hum Gene Ther. 2007;18:1–9.
    1. Taher AT, Otrock ZK, Uthman I, Cappellini MD. Thalassemia and hypercoagulability. Blood Rev. 2008;22:283–292.
    1. Terstappen LW, Huang S, Safford M, Lansdorp PM, Loken MR. Sequential generations of hematopoietic colonies derived from single nonlineage-committed CD34+CD38-progenitor cells. Blood. 1991;77:1218–1227.
    1. Tjonnfjord GE, Steen R, Evensen SA, Thorsby E, Egeland T. Characterization of CD34+ peripheral blood cells from healthy adults mobilized by recombinant human granulocyte colony-stimulating factor. Blood. 1994;84:2795–2801.
    1. Urbinati F, Arumugam P, Higashimoto T, Perumbeti A, Mitts K, Xia P, Malik P. Mechanism of reduction in titers from lentivirus vectors carrying large inserts in the 3′LTR. Mol Ther. 2009;17:1527–1536.
    1. van Lochem EG, van der Velden VH, Wind HK, te Marvelde JG, Westerdaal NA, van Dongen JJ. Immunophenotypic differentiation patterns of normal hematopoiesis in human bone marrow: reference patterns for age-related changes and disease-induced shifts. Cytometry B Clin Cytom. 2004;60:1–13.
    1. Weatherall DJ, Clegg JB. Inherited haemoglobin disorders: an increasing global health problem. Bull World Health Organ. 2001;79:704–712.
    1. Williams DA. Gene therapy continues to mature and to face challenges. Mol Ther. 2009;17:1305–1306.
    1. Wu X, Luke BT, Burgess SM. Redefining the common insertion site. Virology. 2006;344:292–295.

Source: PubMed

3
S'abonner