Lentiviral haemopoietic stem/progenitor cell gene therapy for treatment of Wiskott-Aldrich syndrome: interim results of a non-randomised, open-label, phase 1/2 clinical study

Francesca Ferrua, Maria Pia Cicalese, Stefania Galimberti, Stefania Giannelli, Francesca Dionisio, Federica Barzaghi, Maddalena Migliavacca, Maria Ester Bernardo, Valeria Calbi, Andrea Angelo Assanelli, Marcella Facchini, Claudia Fossati, Elena Albertazzi, Samantha Scaramuzza, Immacolata Brigida, Serena Scala, Luca Basso-Ricci, Roberta Pajno, Miriam Casiraghi, Daniele Canarutto, Federica Andrea Salerio, Michael H Albert, Antonella Bartoli, Hermann M Wolf, Rossana Fiori, Paolo Silvani, Salvatore Gattillo, Anna Villa, Luca Biasco, Christopher Dott, Emily J Culme-Seymour, Koenraad van Rossem, Gillian Atkinson, Maria Grazia Valsecchi, Maria Grazia Roncarolo, Fabio Ciceri, Luigi Naldini, Alessandro Aiuti, Francesca Ferrua, Maria Pia Cicalese, Stefania Galimberti, Stefania Giannelli, Francesca Dionisio, Federica Barzaghi, Maddalena Migliavacca, Maria Ester Bernardo, Valeria Calbi, Andrea Angelo Assanelli, Marcella Facchini, Claudia Fossati, Elena Albertazzi, Samantha Scaramuzza, Immacolata Brigida, Serena Scala, Luca Basso-Ricci, Roberta Pajno, Miriam Casiraghi, Daniele Canarutto, Federica Andrea Salerio, Michael H Albert, Antonella Bartoli, Hermann M Wolf, Rossana Fiori, Paolo Silvani, Salvatore Gattillo, Anna Villa, Luca Biasco, Christopher Dott, Emily J Culme-Seymour, Koenraad van Rossem, Gillian Atkinson, Maria Grazia Valsecchi, Maria Grazia Roncarolo, Fabio Ciceri, Luigi Naldini, Alessandro Aiuti

Abstract

Background: Wiskott-Aldrich syndrome is a rare, life-threatening, X-linked primary immunodeficiency characterised by microthrombocytopenia, infections, eczema, autoimmunity, and malignant disease. Lentiviral vector-mediated haemopoietic stem/progenitor cell (HSPC) gene therapy is a potentially curative treatment that represents an alternative to allogeneic HSPC transplantation. Here, we report safety and efficacy data from an interim analysis of patients with severe Wiskott-Aldrich syndrome who received lentiviral vector-derived gene therapy.

Methods: We did a non-randomised, open-label, phase 1/2 clinical study in paediatric patients with severe Wiskott-Aldrich syndrome, defined by either WAS gene mutation or absent Wiskott-Aldrich syndrome protein (WASP) expression or a Zhu clinical score of 3 or higher. We included patients who had no HLA-identical sibling donor available or, for children younger than 5 years of age, no suitable 10/10 matched unrelated donor or 6/6 unrelated cord blood donor. After treatment with rituximab and a reduced-intensity conditioning regimen of busulfan and fludarabine, patients received one intravenous infusion of autologous CD34+ cells genetically modified with a lentiviral vector encoding for human WAS cDNA. The primary safety endpoints were safety of the conditioning regimen and safety of lentiviral gene transfer into HSPCs. The primary efficacy endpoints were overall survival, sustained engraftment of genetically corrected HSPCs, expression of vector-derived WASP, improved T-cell function, antigen-specific responses to vaccinations, and improved platelet count and mean platelet volume normalisation. This interim analysis was done when the first six patients treated had completed at least 3 years of follow-up. The planned analyses are presented for the intention-to-treat population. This trial is registered with ClinicalTrials.gov (number NCT01515462) and EudraCT (number 2009-017346-32).

Findings: Between April 20, 2010, and Feb 26, 2015, nine patients (all male) were enrolled of whom one was excluded after screening; the age range of the eight treated children was 1·1-12·4 years. At the time of the interim analysis (data cutoff April 29, 2016), median follow-up was 3·6 years (range 0·5-5·6). Overall survival was 100%. Engraftment of genetically corrected HSPCs was successful and sustained in all patients. The fraction of WASP-positive lymphocytes increased from a median of 3·9% (range 1·8-35·6) before gene therapy to 66·7% (55·7-98·6) at 12 months after gene therapy, whereas WASP-positive platelets increased from 19·1% (range 4·1-31·0) to 76·6% (53·1-98·4). Improvement of immune function was shown by normalisation of in-vitro T-cell function and successful discontinuation of immunoglobulin supplementation in seven patients with follow-up longer than 1 year, followed by positive antigen-specific response to vaccination. Severe infections fell from 2·38 (95% CI 1·44-3·72) per patient-year of observation (PYO) in the year before gene therapy to 0·31 (0·04-1·11) per PYO in the second year after gene therapy and 0·17 (0·00-0·93) per PYO in the third year after gene therapy. Before gene therapy, platelet counts were lower than 20 × 109 per L in seven of eight patients. At the last follow-up visit, the platelet count had increased to 20-50 × 109 per L in one patient, 50-100 × 109 per L in five patients, and more than 100 × 109 per L in two patients, which resulted in independence from platelet transfusions and absence of severe bleeding events. 27 serious adverse events in six patients occurred after gene therapy, 23 (85%) of which were infectious (pyrexia [five events in three patients], device-related infections, including one case of sepsis [four events in three patients], and gastroenteritis, including one case due to rotavirus [three events in two patients]); these occurred mainly in the first 6 months of follow-up. No adverse reactions to the investigational drug product and no abnormal clonal proliferation or leukaemia were reported after gene therapy.

Interpretation: Data from this study show that gene therapy provides a valuable treatment option for patients with severe Wiskott-Aldrich syndrome, particularly for those who do not have a suitable HSPC donor available.

Funding: Italian Telethon Foundation, GlaxoSmithKline, and Orchard Therapeutics.

Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Study design PBSC=peripheral blood stem cell.
Figure 2
Figure 2
Multilineage engraftment of genetically corrected haemopoietic stem cells and peripheral blood cells in eight patients with Wiskott-Aldrich syndrome treated with lentiviral vector gene therapy (A) In-vivo engraftment of transduced progenitor cells in bone marrow (LV-positive colonies). (B) Median VCN per cell in bone marrow cell lineages. (C) Median VCN per cell in peripheral blood cell lineages. No vector was detected in bone marrow total cells analysed before gene therapy. LV=lentiviral vector. VCN=vector copy number. GlyA=glycophorin A. NK=natural killer.
Figure 3
Figure 3
Immune reconstitution and clinical benefit Gene therapy was administered on day 1 (month 0). (A) Percentage of peripheral blood lymphocytes expressing WASP, as measured by flow cytometry. (B) T-cell proliferation response (expressed as SI) after incubation with increasing concentrations of anti-CD3i antigen, by observation period. Boxes represent upper and lower quartiles (outliers excluded). Horizontal line within the box is the median. Whiskers represent most extreme points ≤1·5 × IQR. Control data are from 12 healthy children. (C) Rate of severe infections (events per PYO) for each observation period. Error bars represent the 95% CI. (D) Number of days in hospital (days per PYO) for each observation period. Error bars represent the 95% CI. (E) Eczema scores are 1 (absent), 2 (mild), 3 (moderate), and 4 (severe). Six patients were followed up for at least 3 years after gene therapy. WASP=Wiskott-Aldrich syndrome protein. SI=stimulation index. Anti-CD3i=anti-immobilised CD3. PYO=patient-year of observation.
Figure 4
Figure 4
Platelets and bleeding events Gene therapy was administered on day 1 (month 0). (A) Percentage of platelets expressing WASP, as measured by flow cytometry. (B) Mean (SD) platelet count (× 109 cells per L). SD is not plotted for follow-up at 60 months because data were available for only two patients. (C) Moderate or severe bleeding events (per PYO) for each observation period. Error bars represent the 95% CI. (D) Distribution of all bleeding events for each observation period, by body site. Total number of events for each observation period is shown in the centre of each chart. WASP=Wiskott-Aldrich syndrome protein. PYO=patient-year of observation.

References

    1. Thrasher AJ, Burns SO. WASP: a key immunological multitasker. Nat Rev Immunol. 2010;10:182–192.
    1. Ochs HD, Thrasher AJ. The Wiskott-Aldrich syndrome. J Allergy Clin Immunol. 2006;117:725–738.
    1. Bosticardo M, Marangoni F, Aiuti A, Villa A, Roncarolo MG. Recent advances in understanding the pathophysiology of Wiskott-Aldrich syndrome. Blood. 2009;113:6288–6295.
    1. Sullivan KE, Mullen CA, Blaese RM, Winkelstein JA. A multi-institutional survey of the Wiskott-Aldrich syndrome. J Pediatr. 1994;125:876–885.
    1. Dupuis-Girod S, Medioni J, Haddad E. Autoimmunity in Wiskott-Aldrich syndrome: risk factors, clinical features, and outcome in a single-center cohort of 55 patients. Pediatrics. 2003;111:e622–e627.
    1. Filipovich AH, Stone JV, Tomany SC. Impact of donor type on outcome of bone marrow transplantation for Wiskott-Aldrich syndrome: collaborative study of the International Bone Marrow Transplant Registry and the National Marrow Donor Program. Blood. 2001;97:1598–1603.
    1. Kobayashi R, Ariga T, Nonoyama S. Outcome in patients with Wiskott-Aldrich syndrome following stem cell transplantation: an analysis of 57 patients in Japan. Br J Haematol. 2006;135:362–366.
    1. Ozsahin H, Cavazzana-Calvo M, Notarangelo LD. Long-term outcome following hematopoietic stem-cell transplantation in Wiskott-Aldrich syndrome: collaborative study of the European Society for Immunodeficiencies and European Group for Blood and Marrow Transplantation. Blood. 2008;111:439–445.
    1. Moratto D, Giliani S, Bonfim C. Long-term outcome and lineage-specific chimerism in 194 patients with Wiskott-Aldrich syndrome treated by hematopoietic cell transplantation in the period 1980–2009: an international collaborative study. Blood. 2011;118:1675–1684.
    1. Shin CR, Kim M-O, Li D. Outcomes following hematopoietic cell transplantation for Wiskott-Aldrich syndrome. Bone Marrow Transplant. 2012;47:1428–1435.
    1. Braun CJ, Boztug K, Paruzynski A. Gene therapy for Wiskott-Aldrich syndrome: long-term efficacy and genotoxicity. Sci Transl Med. 2014;6:227ra33.
    1. Dupré L, Trifari S, Follenzi A. Lentiviral vector-mediated gene transfer in T cells from Wiskott-Aldrich syndrome patients leads to functional correction. Mol Ther. 2004;10:903–915.
    1. Modlich U, Navarro S, Zychlinski D. Insertional transformation of hematopoietic cells by self-inactivating lentiviral and gammaretroviral vectors. Mol Ther. 2009;17:1919–1928.
    1. Scaramuzza S, Biasco L, Ripamonti A. Preclinical safety and efficacy of human CD34(+) cells transduced with lentiviral vector for the treatment of Wiskott-Aldrich syndrome. Mol Ther. 2013;21:175–184.
    1. Marangoni F, Bosticardo M, Charrier S. Evidence for long-term efficacy and safety of gene therapy for Wiskott-Aldrich syndrome in preclinical models. Mol Ther. 2009;17:1073–1082.
    1. Aiuti A, Biasco L, Scaramuzza S. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science. 2013;341:1233151.
    1. Malär R, Sjöö F, Rentsch K, Hassan M, Güngör T. Therapeutic drug monitoring is essential for intravenous busulfan therapy in pediatric hematopoietic stem cell recipients. Pediatr Transplant. 2011;15:580–588.
    1. Brigida I, Scaramuzza S, Lazarevic D. A novel genomic inversion in Wiskott-Aldrich-associated autoinflammation. J Allergy Clin Immunol. 2016;138:619–622.
    1. Sessa M, Lorioli L, Fumagalli F. Lentiviral haemopoietic stem-cell gene therapy in early-onset metachromatic leukodystrophy: an ad-hoc analysis of a non-randomised, open-label, phase 1/2 trial. Lancet. 2016;388:476–487.
    1. Lidonnici MR, Paleari Y, Tiboni F. Multiple integrated non-clinical studies predict the safety of lentivirus-mediated gene therapy for β-thalassemia. Mol Ther Methods Clin Dev. 2018;11:9–28.
    1. Comans-Bitter WM, de Groot R, van den Beemd R. Immunophenotyping of blood lymphocytes in childhood: reference values for lymphocyte subpopulations. J Pediatr. 1997;130:388–393.
    1. Shearer WT, Rosenblatt HM, Gelman RS. Lymphocyte subsets in healthy children from birth through 18 years of age: the Pediatric AIDS Clinical Trials Group P1009 study. J Allergy Clin Immunol. 2003;112:973–980.
    1. Mahlaoui N, Pellier I, Mignot C. Characteristics and outcome of early-onset, severe forms of Wiskott-Aldrich syndrome. Blood. 2013;121:1510–1516.
    1. Scala S, Basso-Ricci L, Dionisio F. Dynamics of genetically engineered hematopoietic stem and progenitor cells after autologous transplantation in humans. Nat Med. 2018;24:1683–1690.
    1. Castiello M, Scaramuzza S, Pala F. B-cell reconstitution after lentiviral vector-mediated gene therapy in patients with Wiskott-Aldrich syndrome. J Allergy Clin Immunol. 2015;136:692–702.
    1. Pala F, Morbach H, Castiello M. Lentiviral-mediated gene therapy restores B cell tolerance in Wiskott-Aldrich syndrome patients. J Clin Invest. 2015;125:3941–3951.
    1. Hacein-Bey Abina S, Gaspar H, Blondeau J. Outcomes following gene therapy in patients with severe Wiskott-Aldrich syndrome. JAMA. 2015;313:1550–1563.
    1. Cavazzana M, Ribeil J, Lagresle-Peyrou C, André-Schmutz I. Gene therapy with hematopoietic stem cells: the diseased bone marrow's point of view. Stem Cells Dev. 2017;26:71–76.
    1. Elfeky R, Furtado-Silva J, Chiesa R. One hundred percent survival after transplantation of 34 patients with Wiskott-Aldrich syndrome over 20 years. J Allergy Clin Immunol. 2018;142:1654–1656.
    1. Balashov D, Laberko A, Shcherbina A. A conditioning regimen with plerixafor is safe and improves the outcome of TCRαβ+ and CD19+ cell-depleted stem cell transplantation in patients with Wiskott-Aldrich syndrome. Biol Blood Marrow Transpl. 2018;24:1432–1440.
    1. Morris E, Fox T, Chakraverty R. Gene therapy for Wiskott-Aldrich syndrome in a severely affected adult. Blood. 2017;130:1327–1335.
    1. Astrakhan A, Sather BD, Ryu BY. Ubiquitous high-level gene expression in hematopoietic lineages provides effective lentiviral gene therapy of murine Wiskott-Aldrich syndrome. Blood. 2012;119:4395–4407.
    1. Teipel R, Oelschlägel U, Wetzko K. Differences in cellular composition of peripheral blood stem cell grafts from healthy stem cell donors mobilized with either G-CSF alone or G-CSF and plerixafor. Biol Blood Marrow Transpl. 2018;24:2171–2177.

Source: PubMed

3
購読する