Cell-Mediated Immunity to NAGLU Transgene Following Intracerebral Gene Therapy in Children With Mucopolysaccharidosis Type IIIB Syndrome

Marie-Lise Gougeon, Béatrice Poirier-Beaudouin, Jérome Ausseil, Michel Zérah, Cécile Artaud, Jean-Michel Heard, Kumaran Deiva, Marc Tardieu, Marie-Lise Gougeon, Béatrice Poirier-Beaudouin, Jérome Ausseil, Michel Zérah, Cécile Artaud, Jean-Michel Heard, Kumaran Deiva, Marc Tardieu

Abstract

Mucopolysaccharidosis type IIIB syndrome (Sanfilippo disease) is a rare autosomic recessif disorder caused by mutations in the α-N-acetylglucosaminidase (NAGLU) gene coding for a lysosomal enzyme, leading to neurodegeneration and progressive deterioration of cognitive abilities in affected children. To supply the missing enzyme, several recent human gene therapy trials relied on the deposit of adeno-associated virus (AAV) vectors directly into the brain. We reported safety and efficacy of an intracerebral therapy in a phase 1/2 clinical trial (https://ichgcp.net/clinical-trials-registry/NCT03300453), with a recombinant AAV serotype 2/5 (rAAV2/5) coding human NAGLU in four children with MPS IIIB syndrome receiving immunosuppression. It was reported that AAV-mediated gene therapies might elicit a strong host immune response resulting in decreased transgene expression. To address this issue, we performed a comprehensive analysis of cellular immunity and cytokine patterns generated against the therapeutic enzyme in the four treated children over 5.5 years of follow-up. We report the emergence of memory and polyfunctional CD4+ and CD8+ T lymphocytes sensitized to the transgene soon after the start of therapy, and appearing in peripheral blood in waves throughout the follow-up. However, this response had no apparent impact on CNS transgene expression, which remained stable 66 months after surgery, possibly a consequence of the long-term immunosuppressive treatment. We also report that gene therapy did not trigger neuroinflammation, evaluated through the expression of cytokines and chemokines in patients' CSF. Milder disease progression in the youngest patient was found associated with low level and less differentiated circulating NAGLU-specific T cells, together with the lack of proinflammatory cytokines in the CSF. Findings in this study support a systematic and comprehensive immunomonitoring approach for understanding the impact immune reactions might have on treatment safety and efficacy of gene therapies.

Keywords: AAV; CNS; MPS IIIB; NAGLU; T cells; cellular immunity; cytokines.

Conflict of interest statement

MT has received consulting fees from UniQure. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Gougeon, Poirier-Beaudouin, Ausseil, Zérah, Artaud, Heard, Deiva and Tardieu.

Figures

Graphical Abstract
Graphical Abstract
Figure 1
Figure 1
Time course distribution of lymphocyte subsets and expression of activation markers. (A) Lymphocyte subset enumeration was performed at indicated time points on whole blood by polychromatic flowcytometry. Cell subsets were characterized on gated CD45+ cells through the expression of CD3 (CD3+ T cells), coexpression of CD3 and CD4 (CD4+ T cells) or CD3 and CD8 (CD8+ T cells), CD3neg CD19+ (B cells) and CD3neg CD16+ (NK cells). Individual values of each patient (P1, P2, P3, P4) are shown. (B) The expression of the activation markers HLA-DR and CD38 on indicated T cell subsets was assessed on whole blood by polychromatic flowcytometry at indicated time points after gene therapy. Individual values of each patient (P1, P2, P3, P4) are shown.
Figure 2
Figure 2
Time course distribution and activation state of memory/effector CD4+ and CD8+ T cells. (A) Whole blood distribution of naïve/memory/effector subsets in CD4+ and CD8+ T cells. The percentage of cell subsets within CD4+ and CD8+ T cells are shown at indicated time points. (B) HLA-DR expression in indicated subsets within CD4+ and CD8+ T cells. (C) CD38 expression in indicated subsets within CD4+ and CD8+ T cells. N, naïve, CM, Central Memory; EM, Effector Memory; TD, Terminally Differentiated, identified through CD45RA/CCR7 expression. Mean value ± SD are shown. Statistical comparisons for each time point vs baseline are indicated. *0.01 <p< 0.05, **0.001<p< 0.01, ***0.0001<p<0.001.
Figure 3
Figure 3
Ex-vivo NAGLU-specific proliferative response.(A) Representative flow charts showing the proliferation of CD4+ and CD8+ T cells on gated CD3+ T cells in response to NAGLU (1 μg/ml), NAGLU peptide (1 μg/ml), SEB (1 μg/ml), or unstimulated (medium), assessed with the CSFE assay. Freshly isolated PBMCs were stained with CFSE and stimulated for 4 days with the indicated antigens. Percentage of proliferating CFSElow cells is indicated in the quadrants. (B) Time course evolution of NAGLU catalytic activity, assessed in concentrated CSF as described in “Patients and Methods”. (C) The ex-vivo detection of NAGLU-specific proliferating CD4+ and CD8+ T cells was assessed at baseline (BL) and 1, 3, 6, 12, 30, 48 and 66 months after surgery using the CFSE assay on freshly isolated PBMCs, stimulated for 4 days with the indicated concentrations of NAGLU or the peptide. The percentage of CFSElow proliferating cells within CD4+ and CD8+ T cells, obtained after removing the background in non-stimulated cultures, is indicated for each patient over the longitudinal follow-up. (D) Overlay of patients’ CD4+ and CD8+ T cell proliferating responses to NAGLU and peptide at 1 μg/ml.
Figure 4
Figure 4
Time course evolution of ex-vivo NAGLU-specific cytokine expressing CD4+ and CD8+ T cells. Whole blood was stimulated for 6 h at indicated concentrations of NAGLU or the peptide, and specific CD4+ and CD8+ T-cell responses were measured using an intracellular cytokine staining assay. For each patient, the percentage of NAGLU-specific CD69+ CD4+ or CD8+ T cells expressing TNF-α, IFN-γ or IL-2 is plotted at baseline (BL) and 1, 3, 6, 12, 30, 48 and 66 months after surgery.
Figure 5
Figure 5
Functional profiles of NAGLU-specific circulating CD4+ and CD8+ T cells. (A) Overlay of ex-vivo proliferation and TNF-α, IFN-γ or IL-2 expression by CD4+ and CD8+ T cells after exposure of patients’ PBMCs to NAGLU (0.5 μg/ml) or peptide (1 μg/ml) measured at baseline (BL) and 1, 3, 6, 12, 30, 48 and 66 months after surgery. Mean percentage of NAGLU-specific CFSElow cells or cytokine-expressing cells within CD4+ or CD8+ T cells is plotted for each time point. (B) CD4+ and CD8+ T-cell polyfunctionality analysis after whole blood stimulation for 6 h with NAGLU (0.5 μg/ml) or peptide (1 μg/ml). Bar charts represent the proportion of NAGLU-specific CD4+ or CD8+ T-cells displaying each particular cytokine or combination of cytokines at indicated time points of the follow-up. Data are presented as mean ± SD of specific responses from the four patients.
Figure 6
Figure 6
Baseline CSF and plasma cytokine/chemokine profiles. Boxplot diagrams of cytokine (A) and chemokine (B) concentrations in CSF and plasma from the four patients at baseline, assessed by multianalyte profiling of 27 cytokines and chemokines. Red star highlights cytokines or chemokines for which the values are markedly higher than those for healthy children (29). Data are presented as mean ± SD of cytokine/chemokine concentrations.
Figure 7
Figure 7
Impact of gene therapy on neuroinflammation. Heat map representing the broad array of cytokines (A) and chemokines (B) assessed by multianalyte profiling of 27 metabolites in patients’ CSF and plasma, at indicated time-points. The colored scale bar shows the range of concentrations expressed in pg/ml.

References

    1. Heron B, Mikaeloff Y, Froissart R, Caridade G, Maire I, Caillaud C, et al. . Incidence and Natural History of Mucopolysaccharidosis Type III in France and Comparison With United Kingdom and Greece. Am J Med Genet A (2011) 155A:58–68. 10.1002/ajmg.a.33779
    1. Valstar MJ, Bruggenwirth HT, Olmer R, Wevers RA, Verheijen FW, Poorthuis BJ, et al. . Mucopolysaccharidosis Type IIIB may Predominantly Present With an Attenuated Clinical Phenotype. J Inherit Metab Dis (2010) 33:759–67. 10.1007/s10545-010-9199-y
    1. Weber B, Guo XH, Kleijer WJ, van de Kamp JJ, Poorthuis BJ, Hopwood JJ. Sanfilippo Type B Syndrome (Mucopolysaccharidosis III B): Allelic Heterogeneity Corresponds to the Wide Spectrum of Clinical Phenotypes. Eur J Hum Genet (1999) 7:34–44. 10.1038/sj.ejhg.5200242
    1. Beneto N, Vilageliu L, Grinberg D, Canals I. Sanfilippo Syndrome: Molecular Basis, Disease Models and Therapeutic Approaches. Int J Mol Sci (2020) 21:7819–39. 10.3390/ijms21217819
    1. Cheng SH. Gene Therapy for the Neurological Manifestations in Lysosomal Storage Disorders. J Lipid Res (2014) 55:1827–38. 10.1194/jlr.R047175
    1. Mijanovic O, Brankovic A, Borovjagin A, Butnaru DV, Bezrukov EA, Sukhanov RB, et al. . Battling Neurodegenerative Diseases With Adeno-Associated Virus-Based Approaches. Viruses (2020) 12:460–85. 10.3390/v12040460
    1. Tardieu M, Zerah M, Husson B, de Bournonville S, Deiva K, Adamsbaum C, et al. . Intracerebral Administration of Adeno-Associated Viral Vector Serotype rh.10 Carrying Human SGSH and SUMF1 cDNAs in Children With Mucopolysaccharidosis Type IIIA Disease: Results of a Phase I/II Trial. Hum Gene Ther (2014) 25:506–16. 10.1089/hum.2013.238
    1. Cressant A, Desmaris N, Verot L, Brejot T, Froissart R, Vanier MT, et al. . Improved Behavior and Neuropathology in the Mouse Model of Sanfilippo Type IIIB Disease After Adeno-Associated Virus-Mediated Gene Transfer in the Striatum. J Neurosci (2004) 24:10229–39. 10.1523/JNEUROSCI.3558-04.2004
    1. Haurigot V, Marco S, Ribera A, Garcia M, Ruzo A, Villacampa P, et al. . Whole Body Correction of Mucopolysaccharidosis IIIA by Intracerebrospinal Fluid Gene Therapy. J Clin Invest (2013) 123:3254–71. 10.1172/JCI66778
    1. Ellinwood NM, Wang P, Skeen T, Sharp NJ, Cesta M, Decker S, et al. . A Model of Mucopolysaccharidosis IIIB (Sanfilippo Syndrome Type IIIB): N-acetyl-alpha-D-glucosaminidase Deficiency in Schipperke Dogs. J Inherit Metab Dis (2003) 26:489–504. 10.1023/a:1025177411938
    1. Ellinwood NM, Ausseil J, Desmaris N, Bigou S, Liu S, Jens JK, et al. . Safe, Efficient, and Reproducible Gene Therapy of the Brain in the Dog Models of Sanfilippo and Hurler Syndromes. Mol Ther (2011) 19:251–9. 10.1038/mt.2010.265
    1. Ferreira V, Twisk J, Kwikkers K, Aronica E, Brisson D, Methot J, et al. . Immune Responses to Intramuscular Administration of Alipogene Tiparvovec (AAV1-LPL(S447X)) in a Phase II Clinical Trial of Lipoprotein Lipase Deficiency Gene Therapy. Hum Gene Ther (2014) 25:180–8. 10.1089/hum.2013.169
    1. Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, et al. . Successful Transduction of Liver in Hemophilia by AAV-Factor IX and Limitations Imposed by the Host Immune Response. Nat Med (2006) 12:342–7. 10.1038/nm1358
    1. Ertl HCJ, High KA. Impact of AAV Capsid-Specific T-Cell Responses on Design and Outcome of Clinical Gene Transfer Trials With Recombinant Adeno-Associated Viral Vectors: An Evolving Controversy. Hum Gene Ther (2017) 28:328–37. 10.1089/hum.2016.172
    1. Mendell JR, Al-Zaidy S, Shell R, Arnold WD, Rodino-Klapac LR, Prior TW, et al. . Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med (2017) 377:1713–22. 10.1056/NEJMoa1706198
    1. McPhee SW, Janson CG, Li C, Samulski RJ, Camp AS, Francis J, et al. . Immune Responses to AAV in a Phase I Study for Canavan Disease. J Gene Med (2006) 8:577–88. 10.1002/jgm.885
    1. Hordeaux J, Hinderer C, Goode T, Katz N, Buza EL, Bell P, et al. . Toxicology Study of Intra-Cisterna Magna Adeno-Associated Virus 9 Expressing Human Alpha-L-Iduronidase in Rhesus Macaques. Mol Ther Methods Clin Dev (2018) 10:79–88. 10.1016/j.omtm.2018.06.003
    1. Ciron C, Desmaris N, Colle MA, Raoul S, Joussemet B, Verot L, et al. . Gene Therapy of the Brain in the Dog Model of Hurler’s Syndrome. Ann Neurol (2006) 60:204–13. 10.1002/ana.20870
    1. Ransohoff RM, Brown MA. Innate Immunity in the Central Nervous System. J Clin Invest (2012) 122:1164–71. 10.1172/JCI58644
    1. Tardieu M, Zerah M, Gougeon ML, Ausseil J, de Bournonville S, Husson B, et al. . Intracerebral Gene Therapy in Children With Mucopolysaccharidosis Type IIIB Syndrome: An Uncontrolled Phase 1/2 Clinical Trial. Lancet Neurol (2017) 16:712–20. 10.1016/S1474-4422(17)30169-2
    1. Lecoeur H, de Oliveira-Pinto LM, Gougeon ML. Multiparametric Flow Cytometric Analysis of Biochemical and Functional Events Associated With Apoptosis and Oncosis Using the 7-Aminoactinomycin D Assay. J Immunol Methods (2002) 265:81–96. 10.1016/s0022-1759(02)00072-8
    1. Tosato F, Bucciol G, Pantano G, Putti MC, Sanzari MC, Basso G, et al. . Lymphocytes Subsets Reference Values in Childhood. Cytometry A (2015) 87:81–5. 10.1002/cyto.a.22520
    1. Wells AD, Gudmundsdottir H, Turka LA. Following the Fate of Individual T Cells Throughout Activation and Clonal Expansion. Signals From T Cell Receptor and CD28 Differentially Regulate the Induction and Duration of a Proliferative Response. J Clin Invest (1997) 100:3173–83. 10.1172/JCI119873
    1. Boyd A, Almeida JR, Darrah PA, Sauce D, Seder RA, Appay V, et al. . Pathogen-Specific T Cell Polyfunctionality is a Correlate of T Cell Efficacy and Immune Protection. PLoS One (2015) 10:e0128714. 10.1371/journal.pone.0128714
    1. Casazza JP, Betts MR, Price DA, Precopio ML, Ruff LE, Brenchley JM, et al. . Acquisition of Direct Antiviral Effector Functions by CMV-specific CD4+ T Lymphocytes With Cellular Maturation. J Exp Med (2006) 203:2865–77. 10.1084/jem.20052246
    1. Ciuffreda D, Comte D, Cavassini M, Giostra E, Buhler L, Perruchoud M, et al. . Polyfunctional HCV-specific T-Cell Responses are Associated With Effective Control of HCV Replication. Eur J Immunol (2008) 38:2665–77. 10.1002/eji.200838336
    1. Qiu Z, Zhang M, Zhu Y, Zheng F, Lu P, Liu H, et al. . Multifunctional CD4 T Cell Responses in Patients With Active Tuberculosis. Sci Rep (2012) 2:216. 10.1038/srep00216
    1. Perez BA, Shutterly A, Chan YK, Byrne BJ, Corti M. Management of Neuroinflammatory Responses to AAV-Mediated Gene Therapies for Neurodegenerative Diseases. Brain Sci (2020) 10:119–35. 10.3390/brainsci10020119
    1. Pranzatelli MR, Tate ED, McGee NR, Colliver JA. Pediatric Reference Ranges for Proinflammatory and Anti-Inflammatory Cytokines in Cerebrospinal Fluid and Serum by Multiplexed Immunoassay. J Interferon Cytokine Res (2013) 33:523–8. 10.1089/jir.2012.0132
    1. Mingozzi F, Maus MV, Hui DJ, Sabatino DE, Murphy SL, Rasko JE, et al. . CD8(+) T-cell Responses to Adeno-Associated Virus Capsid in Humans. Nat Med (2007) 13:419–22. 10.1182/blood-2013-01-306647
    1. Hudry E, Vandenberghe LH. Therapeutic AAV Gene Transfer to the Nervous System: A Clinical Reality. Neuron (2019) 102:263. 10.1016/j.neuron.2019.03.020
    1. Basner-Tschakarjan E, Mingozzi F. Cell-Mediated Immunity to AAV Vectors, Evolving Concepts and Potential Solutions. Front Immunol (2014) 5:350. 10.3389/fimmu.2014.00350
    1. Barnes C, Scheideler O, Schaffer D. Engineering the AAV Capsid to Evade Immune Responses. Curr Opin Biotechnol (2019) 60:99–103. 10.1016/j.copbio.2019.01.002
    1. Kaplitt MG, Feigin A, Tang C, Fitzsimons HL, Mattis P, Lawlor PA, et al. . Safety and Tolerability of Gene Therapy With an Adeno-Associated Virus (AAV) Borne GAD Gene for Parkinson’s Disease: An Open Label, Phase I Trial. Lancet (2007) 369:2097–105. 10.1016/S0140-6736(07)60982-9
    1. Marks WJ, Jr., Bartus RT, Siffert J, Davis CS, Lozano A, Boulis N, et al. . Gene Delivery of AAV2-neurturin for Parkinson’s Disease: A Double-Blind, Randomised, Controlled Trial. Lancet Neurol (2010) 9:1164–72. 10.1016/S1474-4422(10)70254-4
    1. Christine CW, Starr PA, Larson PS, Eberling JL, Jagust WJ, Hawkins RA, et al. . Safety and Tolerability of Putaminal AADC Gene Therapy for Parkinson Disease. Neurology (2009) 73:1662–9. 10.1212/WNL.0b013e3181c29356
    1. Mingozzi F, High KA. Immune Responses to AAV Vectors: Overcoming Barriers to Successful Gene Therapy. Blood (2013) 122:23–36. 10.1038/nm1549
    1. Dickson P, Peinovich M, McEntee M, Lester T, Le S, Krieger A, et al. . Immune Tolerance Improves the Efficacy of Enzyme Replacement Therapy in Canine Mucopolysaccharidosis I. J Clin Invest (2008) 118:2868–76. 10.1172/JCI34676
    1. Sun B, Bird A, Young SP, Kishnani PS, Chen YT, Koeberl DD. Enhanced Response to Enzyme Replacement Therapy in Pompe Disease After the Induction of Immune Tolerance. Am J Hum Genet (2007) 81:1042–9. 10.1086/522236
    1. Kishnani PS, Corzo D, Nicolino M, Byrne B, Mandel H, Hwu WL, et al. . Recombinant Human Acid [Alpha]-Glucosidase: Major Clinical Benefits in Infantile-Onset Pompe Disease. Neurology (2007) 68:99–109. 10.1212/01.wnl.0000251268.41188.04
    1. Rastall DP, Amalfitano A. Recent Advances in Gene Therapy for Lysosomal Storage Disorders. Appl Clin Genet (2015) 8:157–69. 10.2147/TACG.S57682
    1. Rosenberg M, Kingma W, Fitzpatrick MA, Richards SM. Immunosurveillance of Alglucerase Enzyme Therapy for Gaucher Patients: Induction of Humoral Tolerance in Seroconverted Patients After Repeat Administration. Blood (1999) 93:2081–8.
    1. Zhao H, Bailey LA, Grabowski GA. Enzyme Therapy of Gaucher Disease: Clinical and Biochemical Changes During Production of and Tolerization for Neutralizing Antibodies. Blood Cells Mol Dis (2003) 30:90–6. 10.1016/s1079-9796(03)00012-3
    1. Lowenstein PR, Kroeger K, Castro MG. Immunology of Neurological Gene Therapy: How T Cells Modulate Viral Vector-Mediated Therapeutic Transgene Expression Through Immunological Synapses. Neurotherapeutics (2007) 4:715–24. 10.1016/j.nurt.2007.07.010
    1. Mendell JR, Campbell K, Rodino-Klapac L, Sahenk Z, Shilling C, Lewis S, et al. . Dystrophin Immunity in Duchenne’s Muscular Dystrophy. N Engl J Med (2010) 363:1429–37. 10.1056/NEJMoa1000228
    1. Calcedo R, Somanathan S, Qin Q, Betts MR, Rech AJ, Vonderheide RH, et al. . Class I-restricted T-Cell Responses to a Polymorphic Peptide in a Gene Therapy Clinical Trial for alpha-1-antitrypsin Deficiency. Proc Natl Acad Sci USA (2017) 114:1655–9. 10.1073/pnas.1617726114
    1. Dobrzynski E, Mingozzi F, Liu YL, Bendo E, Cao O, Wang L, et al. . Induction of Antigen-Specific CD4+ T-Cell Anergy and Deletion by In Vivo Viral Gene Transfer. Blood (2004) 104:969–77. 10.1182/blood-2004-03-0847
    1. Kumar SRP, Hoffman BE, Terhorst C, de Jong YP, Herzog RW. The Balance Between CD8(+) T Cell-Mediated Clearance of AAV-Encoded Antigen in the Liver and Tolerance is Dependent on the Vector Dose. Mol Ther (2017) 25:880–91. 10.1016/j.ymthe.2017.02.014
    1. Poupiot J, Costa Verdera H, Hardet R, Colella P, Collaud F, Bartolo L, et al. . Role of Regulatory T Cell and Effector T Cell Exhaustion in Liver-Mediated Transgene Tolerance in Muscle. Mol Ther Methods Clin Dev (2019) 15:83–100. 10.1016/j.omtm.2019.08.012
    1. Hoffman BE, Martino AT, Sack BK, Cao O, Liao G, Terhorst C, et al. . Nonredundant Roles of IL-10 and TGF-beta in Suppression of Immune Responses to Hepatic AAV-factor IX Gene Transfer. Mol Ther (2011) 19:1263–72. 10.1038/mt.2011.33
    1. Mueller C, Chulay JD, Trapnell BC, Humphries M, Carey B, Sandhaus RA, et al. . Human Treg Responses Allow Sustained Recombinant Adeno-Associated Virus-Mediated Transgene Expression. J Clin Invest (2013) 123:5310–8. 10.1172/JCI70314
    1. Ransohoff RM. How Neuroinflammation Contributes to Neurodegeneration. Science (2016) 353:777–83. 10.1126/science.aag2590
    1. Woodcock T, Morganti-Kossmann MC. The Role of Markers of Inflammation in Traumatic Brain Injury. Front Neurol (2013) 4:18. 10.3389/fneur.2013.00018
    1. Heon-Roberts R, Nguyen ALA, Pshezhetsky AV. Molecular Bases of Neurodegeneration and Cognitive Decline, the Major Burden of Sanfilippo Disease. J Clin Med (2020) 9:344–69. 10.3390/jcm9020344
    1. Ohmi K, Greenberg DS, Rajavel KS, Ryazantsev S, Li HH, Neufeld EF. Activated Microglia in Cortex of Mouse Models of Mucopolysaccharidoses I and IIIB. Proc Natl Acad Sci USA (2003) 100:1902–7. 10.1073/pnas.252784899
    1. DiRosario J, Divers E, Wang C, Etter J, Charrier A, Jukkola P, et al. . Innate and Adaptive Immune Activation in the Brain of MPS IIIB Mouse Model. J Neurosci Res (2009) 87:978–90. 10.1002/jnr.21912
    1. Ausseil J, Desmaris N, Bigou S, Attali R, Corbineau S, Vitry S, et al. . Early Neurodegeneration Progresses Independently of Microglial Activation by Heparan Sulfate in the Brain of Mucopolysaccharidosis IIIB Mice. PLoS One (2008) 3:e2296. 10.1371/journal.pone.0002296

Source: PubMed

3
Abonner