Lentiviral haematopoietic stem-cell gene therapy for early-onset metachromatic leukodystrophy: long-term results from a non-randomised, open-label, phase 1/2 trial and expanded access

Francesca Fumagalli, Valeria Calbi, Maria Grazia Natali Sora, Maria Sessa, Cristina Baldoli, Paola Maria V Rancoita, Francesca Ciotti, Marina Sarzana, Maddalena Fraschini, Alberto Andrea Zambon, Serena Acquati, Daniela Redaelli, Vanessa Attanasio, Simona Miglietta, Fabiola De Mattia, Federica Barzaghi, Francesca Ferrua, Maddalena Migliavacca, Francesca Tucci, Vera Gallo, Ubaldo Del Carro, Sabrina Canale, Ivana Spiga, Laura Lorioli, Salvatore Recupero, Elena Sophia Fratini, Francesco Morena, Paolo Silvani, Maria Rosa Calvi, Marcella Facchini, Sara Locatelli, Ambra Corti, Stefano Zancan, Gigliola Antonioli, Giada Farinelli, Michela Gabaldo, Jesus Garcia-Segovia, Laetitia C Schwab, Gerald F Downey, Massimo Filippi, Maria Pia Cicalese, Sabata Martino, Clelia Di Serio, Fabio Ciceri, Maria Ester Bernardo, Luigi Naldini, Alessandra Biffi, Alessandro Aiuti, Francesca Fumagalli, Valeria Calbi, Maria Grazia Natali Sora, Maria Sessa, Cristina Baldoli, Paola Maria V Rancoita, Francesca Ciotti, Marina Sarzana, Maddalena Fraschini, Alberto Andrea Zambon, Serena Acquati, Daniela Redaelli, Vanessa Attanasio, Simona Miglietta, Fabiola De Mattia, Federica Barzaghi, Francesca Ferrua, Maddalena Migliavacca, Francesca Tucci, Vera Gallo, Ubaldo Del Carro, Sabrina Canale, Ivana Spiga, Laura Lorioli, Salvatore Recupero, Elena Sophia Fratini, Francesco Morena, Paolo Silvani, Maria Rosa Calvi, Marcella Facchini, Sara Locatelli, Ambra Corti, Stefano Zancan, Gigliola Antonioli, Giada Farinelli, Michela Gabaldo, Jesus Garcia-Segovia, Laetitia C Schwab, Gerald F Downey, Massimo Filippi, Maria Pia Cicalese, Sabata Martino, Clelia Di Serio, Fabio Ciceri, Maria Ester Bernardo, Luigi Naldini, Alessandra Biffi, Alessandro Aiuti

Abstract

Background: Effective treatment for metachromatic leukodystrophy (MLD) remains a substantial unmet medical need. In this study we investigated the safety and efficacy of atidarsagene autotemcel (arsa-cel) in patients with MLD.

Methods: This study is an integrated analysis of results from a prospective, non-randomised, phase 1/2 clinical study and expanded-access frameworks. 29 paediatric patients with pre-symptomatic or early-symptomatic early-onset MLD with biochemical and molecular confirmation of diagnosis were treated with arsa-cel, a gene therapy containing an autologous haematopoietic stem and progenitor cell (HSPC) population transduced ex vivo with a lentiviral vector encoding human arylsulfatase A (ARSA) cDNA, and compared with an untreated natural history (NHx) cohort of 31 patients with early-onset MLD, matched by age and disease subtype. Patients were treated and followed up at Ospedale San Raffaele, Milan, Italy. The coprimary efficacy endpoints were an improvement of more than 10% in total gross motor function measure score at 2 years after treatment in treated patients compared with controls, and change from baseline of total peripheral blood mononuclear cell (PBMC) ARSA activity at 2 years after treatment compared with values before treatment. This phase 1/2 study is registered with ClinicalTrials.gov, NCT01560182.

Findings: At the time of analyses, 26 patients treated with arsa-cel were alive with median follow-up of 3·16 years (range 0·64-7·51). Two patients died due to disease progression and one due to a sudden event deemed unlikely to be related to treatment. After busulfan conditioning, all arsa-cel treated patients showed sustained multilineage engraftment of genetically modified HSPCs. ARSA activity in PBMCs was significantly increased above baseline 2 years after treatment by a mean 18·7-fold (95% CI 8·3-42·2; p<0·0001) in patients with the late-infantile variant and 5·7-fold (2·6-12·4; p<0·0001) in patients with the early-juvenile variant. Mean differences in total scores for gross motor function measure between treated patients and age-matched and disease subtype-matched NHx patients 2 years after treatment were significant for both patients with late-infantile MLD (66% [95% CI 48·9-82·3]) and early-juvenile MLD (42% [12·3-71·8]). Most treated patients progressively acquired motor skills within the predicted range of healthy children or had stabilised motor performance (maintaining the ability to walk). Further, most displayed normal cognitive development and prevention or delay of central and peripheral demyelination and brain atrophy throughout follow-up; treatment benefits were particularly apparent in patients treated before symptom onset. The infusion was well tolerated and there was no evidence of abnormal clonal proliferation or replication-competent lentivirus. All patients had at least one grade 3 or higher adverse event; most were related to conditioning or to background disease. The only adverse event related to arsa-cel was the transient development of anti-ARSA antibodies in four patients, which did not affect clinical outcomes.

Interpretation: Treatment with arsa-cel resulted in sustained, clinically relevant benefits in children with early-onset MLD by preserving cognitive function and motor development in most patients, and slowing demyelination and brain atrophy.

Funding: Orchard Therapeutics, Fondazione Telethon, and GlaxoSmithKline.

Conflict of interest statement

Declaration of interests FFu, VC, MGNS, AA, CB, FB, FFe, MM, FT, VG, SR, ESF, MPC, and MEB are investigators of gene therapy clinical trials for MLD sponsored by Orchard Therapeutics, the licence holder of investigational medicinal product arsa-cel. FFu and VC have acted as ad-hoc consultants for an Orchard Therapeutics advisory board. The MLD gene therapy was licensed to GlaxoSmithKline in 2014, and then to Orchard Therapeutics in 2018. Telethon and Ospedale San Raffaele are entitled to receive milestone payments and royalties for such a therapy. MSe and AB left San Raffaele Hospital and their role as principal investigators of the pivotal study on November, 2014, and September, 2015, respectively. AA subsequently became study principal investigator and responsible physician for treatment under expanded access frameworks. AB is currently a member of the scientific advisory board of Orchard Therapeutics and holds stock in Orchard Therapeutics. PMVR and CDS have a contract with Orchard Therapeutics to perform statistical analyses of gene therapy clinical trials for MLD. SMa and FM have a service contract with Ospedale San Raffaele. SMa has a contract with Orchard Therapeutics to perform ARSA activity on CSF in the clinical trial NCT03392987. JG-S, LCS, and GFD are former employees and hold stock in Orchard Therapeutics, which sponsored the clinical trial. All other authors declare that they have no financial interest related to the work described in the manuscript.

Copyright © 2022 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
High-level engraftment of gene-corrected HSPCs (A) Assessed over time by disease subtype by percentage of lentiviral vector-transduced cells in bone marrow clonogenic progenitors. (B) Mean VCN in PBMCs. LLQ is 0·0037 VCN per cell. Zero values are plotted as 0·001. (C) Mean VCN in bone marrow-derived CD34+ cells. LLQ is 0·0037 vector copy number/cell. Zero values are plotted as 0·001. (D) Mean ARSA activity in PBMCs. LLQ is 25·79 nmol/mg/h. ARSA activity measured in PBMC in the intention-to-treat set after treatment at years 2 (coprimary endpoint) and 3 was compared with pre-treatment values using a mixed-model repeated measures model. (E) Mean ARSA activity in cerebrospinal fluid (CSF). LLQ is 0·0032 nmol/mg/h. Geometric means and 95% CIs are presented where there are at least three patients with non-missing data. ARSA=arylsulfatase A. GM=geometric mean. LLQ=lower limit of quantification. PBMCs=peripheral blood mononuclear cells. VCN=vector copy number. *95% CI for 60-month timepoint: 24·35–128·73. †95% CI for 72-month timepoint: 6·69–303·48. ‡From adult reference donors. §From paediatric reference donors. In all panels, values less than the LLQ were imputed as the LLQ as this represents a conservative approach to treatment evaluation in those cases.
Figure 2
Figure 2
GMFM scores and age at severe motor impairment or death (A) GMFM scores for patients with late-infantile and early-juvenile MLD compared with age-matched and disease subtype-matched untreated natural history controls at 2 and 3 years after treatment. Adjusted least-squares means, treatment difference (atidarsagene autotemcel [arsa-cel] minus natural history), and the associated 95% CI and p value were reported overall and by disease subtype (late infantile, early juvenile) for the null hypothesis of 10% or less difference in total GMFM scores between treated and natural history patients at years 2 and 3. (B) Kaplan-Meier plot showing age at severe motor impairment or death in patients with late-infantile MLD versus untreated natural history late-infantile MLD controls. (C) Kaplan-Meier plot showing age at severe motor impairment or death in patients with pre-symptomatic and early-symptomatic early-juvenile MLD versus untreated natural history early-juvenile MLD controls. Severe motor impairment-free survival is defined as the interval from birth to the earlier loss of locomotion and sitting without support (GMFC level 5 or 6) or death from any cause; otherwise severe motor impairment-free survival is censored at the last GMFC assessment date. Symptomatic status refers to arsa-cel treated patients at the time of treatment. GMFM=gross motor function measure. MLD=metachromatic leukodystrophy. *p values calculated using an unstratified log-rank test.
Figure 3
Figure 3
Cognitive performance and verbal age-equivalent profiles (A) Age-equivalent cognitive performance in late-infantile patients. (B) Age-equivalent cognitive performance in early-juvenile patients. (C) Verbal-age equivalent in late-infantile patients. (D) Verbal-age equivalent in early-juvenile patients. Age-equivalent corresponds to the chronological age at which, on average, typically developing children reach a given raw score. Cognitive age-equivalent for late-infantile (A) and early-juvenile (B) at each visit has been derived as follows. For Wechsler Preschool & Primary Scale of Intelligence (WPPSI) and Wechsler Intelligence Scale for Children (WISC): (development quotient performance x chronological age)/100 for which development quotient is derived by dividing the age-equivalent by the chronological age and then multiplying by 100. For Bayley III: cognitive raw scores have been compared with the tabulated values in the Bayley III manual to calculate cognitive age-equivalent. For Bayley II and in cases for which a neuropsychological assessment has been done but a questionnaire could not be completed because of severe clinical condition, cognitive age-equivalent is based on mental development age as reported on the case report form (CRF). Verbal age-equivalent for late-infantile (C) and early-juvenile (D) at each visit has been derived as follows. For WPPSI and WISC: (development quotient language x chronological age)/100. For Bayley III: expressive communication and receptive communication raw scores have each been compared with the tabulated values in the Bayley III manual and based on the average of the mental development ages for the two scores. For Bayley II and in cases for which a neuropsychological assessment has been done but a questionnaire could not be completed because of severe clinical condition, verbal age-equivalent is based on mental development age as reported on the CRF. Arsa-cel=atidarsagene autotemcel. MLD=metachromatic leukodystrophy.
Figure 4
Figure 4
Longitudinal evaluation of brain MRI of patients treated with arsa-cel vs natural history (A) Axial T2-weighted MRI obtained from a patient with late-infantile MLD (patient 3) over time showing, at baseline, a physiological T2 signal corresponding to myelin maturation, typical of the first year of life, followed by appearance of subtle posterior periventricular T2 hyperintensities that stabilised over time, with normal maturation of the remaining brain regions. (B) The comparison with the corresponding images obtained from his untreated sibling at the same age. (C) The comparison with an unrelated untreated patient with late-infantile MLD with similar age of onset, followed along disease course demonstrating white matter alterations (T2 hyperintensities) involving, in the earliest phases, periventricular areas and corpus callosum, spreading to subcortical regions, cerebellum and corticospinal tracts, and associated with progressive brain atrophy (enlargement of ventricular system and subarachnoid spaces). (D and E) Comparison of brain MRI total scores of patients with late-infantile (D) and early-juvenile by symptomatic status (E) treated with atidarsagene autotemcel [arsa-cel] versus natural history through an non-linear mixed-effects model based on a published methodology and strategy for model selection. MRI severity scoring system is a modified Loes’ score using methodology as previously described. The maximum total score is 31·5. MLD=metachromatic leukodystrophy.

References

    1. van Rappard DF, Boelens JJ, Wolf NI. Metachromatic leukodystrophy: disease spectrum and approaches for treatment. Best Pract Res Clin Endocrinol Metab. 2015;29:261–273.
    1. Gieselmann V, Krägeloh-Mann I. Metachromatic leukodystrophy–an update. Neuropediatrics. 2010;41:1–6.
    1. Von Figura K, Gieselmann V, Jaeken J. In: The metabolic and molecular bases of inherited diseases. Scriver CR BA, Sly WS, editors. McGraw-Hill; New York, NY: 2001. Metachromatic leukodystrophy.
    1. Elgun S, Waibel J, Kehrer C, et al. Phenotypic variation between siblings with metachromatic leukodystrophy. Orphanet J Rare Dis. 2019;14:136.
    1. Lorioli L, Cicalese MP, Silvani P, et al. Abnormalities of acid-base balance and predisposition to metabolic acidosis in metachromatic leukodystrophy patients. Mol Genet Metab. 2015;115:48–52.
    1. Kim J, Sun Z, Ezekian B, et al. Gallbladder abnormalities in children with metachromatic leukodystrophy. J Surg Res. 2017;208:187–191.
    1. Kehrer C, Blumenstock G, Gieselmann V, Krägeloh-Mann I, on behalf of the German Leukonet The natural course of gross motor deterioration in metachromatic leukodystrophy. Dev Med Child Neurol. 2011;53:850–885.
    1. Kehrer C, Elgun S, Raabe C, et al. Association of age at onset and first symptoms with disease progression in patients with metachromatic leukodystrophy. Neurology. 2021;96:e255–e266.
    1. Biffi A, Cesani M, Fumagalli F, et al. Metachromatic leukodystrophy – mutation analysis provides further evidence of genotype–phenotype correlation. Clin Genet. 2008;74:349–357.
    1. Bredius RG, Laan LA, Lankester AC, et al. Early marrow transplantation in a pre-symptomatic neonate with late infantile metachromatic leukodystrophy does not halt disease progression. Bone Marrow Transplant. 2007;39:309–310.
    1. Boucher AA, Miller W, Shanley R, et al. Long-term outcomes after allogeneic hematopoietic stem cell transplantation for metachromatic leukodystrophy: the largest single-institution cohort report. Orphanet J Rare Dis. 2015;10:94.
    1. Martin HR, Poe MD, Provenzale JM, Kurtzberg J, Mendizabal A, Escolar ML. Neurodevelopmental outcomes of umbilical cord blood transplantation in metachromatic leukodystrophy. Biol Blood Marrow Transplant. 2013;19:616–624.
    1. van den Broek BTA, Page K, Paviglianiti A, et al. Early and late outcomes after cord blood transplantation for pediatric patients with inherited leukodystrophies. Blood Adv. 2018;2:49–60.
    1. Dali C, Sevin C, Krageloh-Mann I, et al. Safety of intrathecal delivery of recombinant human arylsulfatase A in children with metachromatic leukodystrophy: results from a phase 1/2 clinical trial. Mol Genet Metab. 2020;131:235–244.
    1. Sevin C, Roujeau T, Cartier N, et al. Intracerebral gene therapy in children with metachromatic leukodystrophy: results of a phase I/II trial. Mol Genet Metab. 2018;123:S129.
    1. Biffi A, Montini E, Lorioli L, et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science. 2013;341
    1. Sessa M, Lorioli L, Fumagalli F, et al. 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. Fumagalli F, Zambon AA, Rancoita PMV, et al. Metachromatic leukodystrophy: a single-center longitudinal study of 45 patients. J Inherit Metab Dis. 2021;44:1151–1164.
    1. Russell D, Rosenbaum P, Wright M, Avery L, Lane M. MacKeith Press; London: 2002. Gross motor function measure (GMFM-66 & GMFM-88) user's manual. Clinics in developmental medicine no.159; p. 233.
    1. Morena F, Argentati C, Acquati S, et al. Toward reference intervals of ARSA activity in the cerebrospinal fluid: implication for the clinical practice of metachromatic leukodystrophy. J Appl Lab Med. 2020;6:354–366.
    1. Calbi V, Fumagalli F, Consiglieri G, et al. Use of defibrotide to help prevent post-transplant endothelial injury in a genetically predisposed infant with metachromatic leukodystrophy undergoing hematopoietic stem cell gene therapy. Bone Marrow Transplant. 2018;53:913–917.
    1. Calabria A, Spinozzi G, Benedicenti F, et al. Late phases of hematopoietic reconstitution in metachromatic leukodystrophy gene therapy patients characterized by lineage commitment of individual HSC clones. Mol Ther. 2020;28:234.
    1. Groeschel S, Kuhl JS, Bley AE, et al. Long-term outcome of allogeneic hematopoietic stem cell transplantation in patients with juvenile metachromatic leukodystrophy compared with nontransplanted control patients. JAMA Neurol. 2016;73:1133–1140.
    1. Tan EY, Boelens JJ, Jones SA, Wynn RF. Hematopoietic stem cell transplantation in inborn errors of metabolism. Front Pediatr. 2019;7:433.
    1. Capotondo A, Milazzo R, Politi LS, et al. Brain conditioning is instrumental for successful microglia reconstitution following hematopoietic stem cell transplantation. Proc Natl Acad Sci USA. 2012;109:15018–15023.
    1. Biffi A, De Palma M, Quattrini A, et al. Correction of metachromatic leukodystrophy in the mouse model by transplantation of genetically modified hematopoietic stem cells. J Clin Invest. 2004;113:1118–1129.
    1. Biffi A, Capotondo A, Fasano S, et al. Gene therapy of metachromatic leukodystrophy reverses neurological damage and deficits in mice. J Clin Invest. 2006;116:3070–3082.
    1. Staal FJT, Aiuti A, Cavazzana M. Autologous stem-cell-based gene therapy for inherited disorders: state of the art and perspectives. Front Pediatr. 2019;7:443.
    1. Hong X, Daiker J, Sadilek M, et al. Toward newborn screening of metachromatic leukodystrophy: results from analysis of over 27,000 newborn dried blood spots. Genet Med. 2021;23:555–561.
    1. Hong X, Kumar AB, Daiker J, et al. Leukocyte and dried blood spot arylsulfatase A assay by tandem mass spectrometry. Anal Chem. 2020;92:6341–6348.
    1. Capotondo A, Milazzo R, Garcia-Manteiga JM, et al. Intracerebroventricular delivery of hematopoietic progenitors results in rapid and robust engraftment of microglia-like cells. Sci Adv. 2017;3
    1. Meneghini V, Lattanzi A, Tiradani L, et al. Pervasive supply of therapeutic lysosomal enzymes in the CNS of normal and Krabbe-affected non-human primates by intracerebral lentiviral gene therapy. EMBO Mol Med. 2016;8:489–510.
    1. Audouard E, Oger V, Meha B, Cartier N, Sevin C, Piguet F. Complete correction of brain and spinal cord pathology in metachromatic leukodystrophy mice. Front Mol Neurosci. 2021;14
    1. Eichler FS, Cox TM, Crombez E, Dali CI, Kohlschutter A. Metachromatic leukodystrophy: an assessment of disease burden. J Child Neurol. 2016;31:1457–1463.

Source: PubMed

3
Se inscrever