Thirty-year clinical outcomes after haematopoietic stem cell transplantation in neuronopathic Gaucher disease

Aimee Donald, Cecilia Kämpe Björkvall, Ashok Vellodi, GAUCHERITE Consortium, Timothy M Cox, Derralyn Hughes, Simon A Jones, Robert Wynn, Maciej Machaczka, Aimee Donald, Cecilia Kämpe Björkvall, Ashok Vellodi, GAUCHERITE Consortium, Timothy M Cox, Derralyn Hughes, Simon A Jones, Robert Wynn, Maciej Machaczka

Abstract

Background: Neuronopathic Gaucher Disease (nGD) describes the condition of a subgroup of patients with the Lysosomal Storage Disorder (LSD), Gaucher disease with involvement of the central nervous system (CNS) which results from inherited deficiency of β-glucosylceramidase. Although systemic manifestations of disease are now corrected by augmentation with macrophage-targeted therapeutic enzyme (enzyme replacement therapy, ERT), neurological disease progresses unpredictably as a result of failure of therapeutic enzyme to cross the blood-brain barrier (BBB). Without therapy, the systemic and neurological effects of the disease progress and shorten life: investigators, principally in Sweden and the UK, pioneered bone marrow transplantation (BMT; Haematopoietic Stem Cell Transplantation HSCT) to supply healthy marrow-derived macrophages and other cells, to correct the peripheral disease. Here we report the first long-term follow-up (over 20 years in all cases) of nine patients in the UK and Sweden who underwent HSCT in the 1970s and 1980s. This retrospective, multicentre observational study was undertaken to determine whether there are neurological features of Gaucher disease that can be corrected by HSCT and the extent to which deterioration continues after the procedure. Since intravenous administration of ERT is approved for patients with the neuronopathic disease and ameliorates many of the important systemic manifestations but fails to correct the neurological features, we also consider the current therapeutic positioning of HSCT in this disorder.

Results: In the nine patients here reported, neurological disease continued to progress after transplantation, manifesting as seizures, cerebellar disease and abnormalities of tone and reflexes.

Conclusions: Although neurological disease progressed in this cohort of patients, there may be a future role for HSCT in the treatment of nGD. The procedure has the unique advantage of providing a life-long source of normally functioning macrophages in the bone marrow, and possibly other sites, after a single administration. HSCT moreover, clearly ameliorates systemic disease and this may be advantageous-especially where sustained provision of high-cost ERT cannot be guaranteed. Given the remaining unmet needs of patients with neuronopathic Gaucher disease and the greatly improved safety profile of the transplant procedure, HSCT could be considered to provide permanent correction of systemic disease, including bone disease not ameliorated by ERT, when combined with emerging therapies directed at the neurological manifestations of disease; this could include ex-vivo gene therapy approaches.

Keywords: BMT; HSCT; Neurology; Neuronopathic Gaucher disease; Outcomes; Type 3 Gaucher disease.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Modified Severity Score (mSST) by age at transplant; X-axis: Age at BMT in years; Y-axis: Post-transplant mSST
Fig. 2
Fig. 2
Age at Splenectomy vs mSST at follow-up; X-axis Age at splenectomy in years, Y-axis: Post-transplant mSST
Fig. 3
Fig. 3
ERT vs HSCT mSST outcomes by age of intervention; X-axis Age at intervention (years); Y-axis: Follow-up mSST; Legend by Intervention type: Blue marker—Bone Marrow Transplant; Green Marker—Enzyme Replacement Therapy; Orange marker: Enzyme Replacement Therapy and splenectomy

References

    1. Schiffmann R, Sevigny J, Rolfs A, Davies EH, Goker-Alpan O, Abdelwahab M, et al. The definition of neuronopathic Gaucher disease. J Inherit Metab Dis. 2020;43:1056–1059. doi: 10.1002/jimd.12235.
    1. Neumann J, Bras J, Deas E, O’Sullivan SS, Parkkinen L, Lachmann RH, et al. Glucocerebrosidase mutations in clinical and pathologically proven Parkinson’s disease. Brain. 2009;132:1783–1794. doi: 10.1093/brain/awp044.
    1. Joint Formulary Committee. British National Formulary (online). London: BMJ Group and Pharmaceutical Press; n.d.
    1. Cabrera-Salazar MA, O’Rourke E, Henderson N, Wessel H, Barranger JA. Correlation of surrogate markers of Gaucher disease Implications for long-term follow up of enzyme replacement therapy. Clin Chim Acta. 2004;344:101–107. doi: 10.1016/j.cccn.2004.02.018.
    1. Schiffmann R, Heyes MP, Aerts JM, Dambrosia JM, Patterson MC, DeGraba T, et al. Prospective study of neurological responses to treatment with macrophage-targeted glucocerebrosidase in patients with type 3 Gaucher’s disease. Ann Neurol. 1997;42:613–621. doi: 10.1002/ana.410420412.
    1. Altarescu G, Hill S, Wiggs E, Jeffries N, Kreps C, Parker CC, et al. The efficacy of enzyme replacement therapy in patients with chronic neuronopathic Gaucher’s disease. J Pediatr. 2001;138:539–547. doi: 10.1067/mpd.2001.112171.
    1. Cabrera-Salazar MA, DeRiso M, Bercury SD, Li L, Lydon JT, Weber W, et al. Systemic delivery of a glucosylceramide synthase inhibitor reduces CNS substrates and increases lifespan in a mouse model of type 2 gaucher disease. PLoS ONE. 2012;7:e43310. doi: 10.1371/journal.pone.0043310.
    1. Treiber A, Morand O, Clozel M. The pharmacokinetics and tissue distribution of the glucosylceramide synthase inhibitor miglustat in the rat. Xenobiotica. 2007;37:298–314. doi: 10.1080/00498250601094543.
    1. Schiffmann R, FitzGibbon EJ, Harris C, DeVile C, Davies EH, Abel L, et al. Randomized, controlled trial of miglustat in Gaucher’s disease type 3. Ann Neurol. 2008;64:514–522. doi: 10.1002/ana.21491.
    1. Marshall J, Sun Y, Bangari DS, Budman E, Park H, Nietupski JB, et al. CNS-accessible inhibitor of glucosylceramide synthase for substrate reduction therapy of neuronopathic gaucher disease. Mol Ther. 2016 doi: 10.1038/mt.2016.53.
    1. Blom S, Erikson A. Gaucher disease—Norrbottnian type. Eur J Pediatr. 1983;140:316–322. doi: 10.1007/BF00442672.
    1. Erikson A, Groth CG, Månsson J-E, Percy A, Ringdén O, Svennerholm L. Clinical and biochemical outcome of marrow transplantation for Gaucher disease of the Norrbottnian type. Acta Pædiatr. 1990;79:680–685. doi: 10.1111/j.1651-2227.1990.tb11535.x.
    1. Erikson A. Gaucher disease: Norrbottnian type (III)—neuropaediatric and neurobiological aspects of clinical patterns and treatment. Stockholm: Almqvist & Wiksell; 1986.
    1. Dahl N, Hillborg P-O, Olofsson A. Gaucher disease (Norrbottnian type III): probable founders identified by genealogical and molecular studies. Hum Genet. 1993;92:513–515. doi: 10.1007/BF00216461.
    1. Davies EH, Mengel E, Tylki-Szymanska A, Kleinotiene G, Reinke J, Vellodi A. Four-year follow-up of chronic neuronopathic Gaucher disease in Europeans using a modified severity scoring tool. J Inherit Metab Dis. 2011;34:1053–1059. doi: 10.1007/s10545-011-9347-z.
    1. den Dunnen JT, Dalgleish R, Maglott DR, Hart RK, Greenblatt MS, McGowan-Jordan J, et al. HGVS recommendations for the description of sequence variants: 2016 update. Hum Mutat. 2016;37:564–569. doi: 10.1002/humu.22981.
    1. Hobbs JR, Shaw PJ, Jones KH, Lindsay I, Hancock M. Beneficial effect of pre-transplant splenectomy on displacement bone marrow transplantation for Gaucher’s syndromE. The Lancet. 1987;329:1111–1115. doi: 10.1016/S0140-6736(87)91673-4.
    1. Ringden O, Groth CG, Erikson A, Granqvist S, Mansson JE, Sparrelid E. Ten years’ experience of bone marrow transplantation for Gaucher disease. Transplantation. 1995;59:864–870. doi: 10.1097/00007890-199503000-00011.
    1. Ringden O, Groth C-G, Erikson A, Backman L, Granqvist S, Marnsson J-E, et al. Long-term follow-up of the first successful bone marrow transplantation in Gaucher disease. Transplantation. 1988;46:66–69. doi: 10.1097/00007890-198807000-00011.
    1. Henig I, Zuckerman T. Hematopoietic stem cell transplantation—50 years of evolution and future perspectives. Rambam Maimonides Med J. 2014 doi: 10.5041/RMMJ.10162.
    1. Pavletic SZ, Fowler DH. Are we making progress in GVHD prophylaxis and treatment? Hematology. 2012;2012:251–264. doi: 10.1182/asheducation-2012.1.251.
    1. Steward AM, Wiggs E, Lindstrom T, Ukwuani S, Ryan E, Tayebi N, et al. Variation in cognitive function over time in Gaucher disease type 3. Neurology. 2019;93:e2272–e2283. doi: 10.1212/WNL.0000000000008618.
    1. Ringden O, Groth CG, Erikson A, Mansson JE, Svennerholm L. Bone Marrow transplantation in the Norrbottnian type of Gaucher’. Correction of Certain Genetic Diseases by Transplantation 1989;30–5.
    1. Tajes M, Ramos-Fernández E, Weng-Jiang X, Bosch-Morató M, Guivernau B, Eraso-Pichot A, et al. The blood-brain barrier: structure, function and therapeutic approaches to cross it. Mol Membr Biol. 2014;31:152–167. doi: 10.3109/09687688.2014.937468.
    1. Norman RM, Urich H, Lloyd OC. The neuropathology of infantile Gaucher’s disease. J Pathol. 1956;72:121–131. doi: 10.1002/path.1700720116.
    1. Wong K, Sidransky E, Verma A, Mixon T, Sandberg GD, Wakefield LK, et al. Neuropathology provides clues to the pathophysiology of Gaucher disease. Mol Genet Metab. 2004;82:192–207. doi: 10.1016/j.ymgme.2004.04.011.
    1. Faraco G, Park L, Anrather J, Iadecola C. Brain perivascular macrophages: characterization and functional roles in health and disease. J Mol Med (Berl) 2017;95:1143–1152. doi: 10.1007/s00109-017-1573-x.
    1. Vallieres L, Sawchenko PE. Bone marrow-derived cells that populate the adult mouse brain preserve their hematopoietic identity. J Neurosci. 2003;23:5197–5207. doi: 10.1523/JNEUROSCI.23-12-05197.2003.
    1. Capotondo A, Milazzo R, Politi LS, Quattrini A, Palini A, Plati T, et al. Brain conditioning is instrumental for successful microglia reconstitution following hematopoietic stem cell transplantation. Proc Natl Acad Sci. 2012;109:15018–15023. doi: 10.1073/pnas.1205858109.
    1. Derecki NC, Cronk JC, Lu Z, Xu E, Abbott SBG, Guyenet PG, et al. Wild-type microglia arrest pathology in a mouse model of Rett syndrome. Nature. 2012;484:105–109. doi: 10.1038/nature10907.
    1. Wilkinson FL, Sergijenko A, Langford-Smith KJ, Malinowska M, Wynn RF, Bigger BW. Busulfan conditioning enhances engraftment of hematopoietic donor-derived cells in the brain compared with irradiation. Mol Ther. 2013;21:868–876. doi: 10.1038/mt.2013.29.
    1. Shah AJ, Epport K, Azen C, Killen R, Wilson K, De Clerck D, et al. Progressive declines in neurocognitive function among survivors of hematopoietic stem cell transplantation for pediatric hematologic malignancies. J Pediatr Hematol Oncol. 2008;30:411–418. doi: 10.1097/MPH.0b013e318168e750.
    1. Aldenhoven M, Wynn RF, Orchard PJ, O’Meara A, Veys P, Fischer A, et al. Long-term outcome of Hurler syndrome patients after hematopoietic cell transplantation: an international multicenter study. Blood. 2015;125:2164–2172. doi: 10.1182/blood-2014-11-608075.
    1. Eichler F, Duncan C, Musolino PL, Orchard PJ, De Oliveira S, Thrasher AJ, et al. Hematopoietic stem-cell gene therapy for cerebral adrenoleukodystrophy. N Engl J Med. 2017;2017(377):1630–1638. doi: 10.1056/NEJMoa1700554.

Source: PubMed

3
Subscribe