Efficacy and long-term safety of alipogene tiparvovec (AAV1-LPLS447X) gene therapy for lipoprotein lipase deficiency: an open-label trial

D Gaudet, J Méthot, S Déry, D Brisson, C Essiembre, G Tremblay, K Tremblay, J de Wal, J Twisk, N van den Bulk, V Sier-Ferreira, S van Deventer, D Gaudet, J Méthot, S Déry, D Brisson, C Essiembre, G Tremblay, K Tremblay, J de Wal, J Twisk, N van den Bulk, V Sier-Ferreira, S van Deventer

Abstract

We describe the 2-year follow-up of an open-label trial (CT-AMT-011-01) of AAV1-LPL(S447X) gene therapy for lipoprotein lipase (LPL) deficiency (LPLD), an orphan disease associated with chylomicronemia, severe hypertriglyceridemia, metabolic complications and potentially life-threatening pancreatitis. The LPL(S447X) gene variant, in an adeno-associated viral vector of serotype 1 (alipogene tiparvovec), was administered to 14 adult LPLD patients with a prior history of pancreatitis. Primary objectives were to assess the long-term safety of alipogene tiparvovec and achieve a ≥40% reduction in fasting median plasma triglyceride (TG) at 3-12 weeks compared with baseline. Cohorts 1 (n=2) and 2 (n=4) received 3 × 10(11) gc kg(-1), and cohort 3 (n=8) received 1 × 10(12) gc kg(-1). Cohorts 2 and 3 also received immunosuppressants from the time of alipogene tiparvovec administration and continued for 12 weeks. Alipogene tiparvovec was well tolerated, without emerging safety concerns for 2 years. Half of the patients demonstrated a ≥40% reduction in fasting TG between 3 and 12 weeks. TG subsequently returned to baseline, although sustained LPL(S447X) expression and long-term changes in TG-rich lipoprotein characteristics were noted independently of the effect on fasting plasma TG.

Trial registration: ClinicalTrials.gov NCT01109498.

Conflict of interest statement

Conflict of Interest

The funding body (AMT) was involved in all aspects of the study (study design, data collection and analysis, and data interpretation in collaboration with the CRO and principal investigator. Five authors (J.dW., J.T., S.vD., N.vdB. and V.S-F) are employees of AMT. The remaining authors declare no conflict of interest. The principal investigator of the study (DG) has no financial interest in AMT and made all final editorial decisions regarding the manuscript.

Figures

Figure 1
Figure 1
Presence of AMT-011 Vector DNA in body fluids: Results are depicted as median values per treatment group, per time point. Dashed lines indicate limit of detection (bottom line) and limit of quantification (top line) of the assay (A: shedding in serum; B; shedding in saliva; C: shedding in urine; D: shedding in semen).
Figure 2
Figure 2
Paraffin-embedded cross-section of the muscle biopsy (injected muscle) from subject 09, stained with H&E; showing a large focal infiltrate and diffuse infiltration throughout the biopsy
Figure 3
Figure 3
Figure 3A: General histology as noted in cryosections of the injected muscle from subject 11 who was one of two patients (09 and 11) to show a more pronounced local response. (A–D) injected muscle (I): (A & B) H&E staining showing (large) perivascular and endomysial infiltration of the injected tissue. Some freezing artifacts are observed. Arrows in (B) indicate small, irregularly shaped degenerating fibers with subsarcolemmal accumulations with neighboring endomysial infiltrates; (C) uneven NADH stain, arrows pointing to increased staining of the subsarcolemmal accumulations noted in (B); (D) accumulation of neutral lipid, visualized by Oil Red O staining. (E&F) non-injected muscle; (E) H&E staining showing largely normal histology except for a single ragged red fiber (indicated by arrow); (F) normal neutral lipid content as visualized using Oil Red O. Figure 3B: Immunohistochemical staining of cryosections of injected muscle from subject 09 who was one of two patients (09 and 11) to show a more pronounced local response. (A) CD3+ T-lymphocytes found within the injected tissue as a large perivascular infiltrate, and more diffuse endomysial infiltration; (B) staining for CD8+ T-lymphocytes in the same area; (C) CD20+ B-lymphocytes in the same area; (D) CD68+ macrophages within the same area; (E and F) staining for MHC class I and –II surface receptors more distal to large infiltrates, showing positivity on the surface of muscle fibers (in addition to positivity of infiltrates).
Figure 3
Figure 3
Figure 3A: General histology as noted in cryosections of the injected muscle from subject 11 who was one of two patients (09 and 11) to show a more pronounced local response. (A–D) injected muscle (I): (A & B) H&E staining showing (large) perivascular and endomysial infiltration of the injected tissue. Some freezing artifacts are observed. Arrows in (B) indicate small, irregularly shaped degenerating fibers with subsarcolemmal accumulations with neighboring endomysial infiltrates; (C) uneven NADH stain, arrows pointing to increased staining of the subsarcolemmal accumulations noted in (B); (D) accumulation of neutral lipid, visualized by Oil Red O staining. (E&F) non-injected muscle; (E) H&E staining showing largely normal histology except for a single ragged red fiber (indicated by arrow); (F) normal neutral lipid content as visualized using Oil Red O. Figure 3B: Immunohistochemical staining of cryosections of injected muscle from subject 09 who was one of two patients (09 and 11) to show a more pronounced local response. (A) CD3+ T-lymphocytes found within the injected tissue as a large perivascular infiltrate, and more diffuse endomysial infiltration; (B) staining for CD8+ T-lymphocytes in the same area; (C) CD20+ B-lymphocytes in the same area; (D) CD68+ macrophages within the same area; (E and F) staining for MHC class I and –II surface receptors more distal to large infiltrates, showing positivity on the surface of muscle fibers (in addition to positivity of infiltrates).
Figure 4
Figure 4
Figure 4A: Distribution of plasma TG and cholesterol in the Sf >400 and Sf 20–400 fractions before therapy and 52 weeks after alipogene tiparvovec administration. Figure 4B: Total APOB in the Sf 20–400 fraction before therapy and 12 and 52 weeks after alipogene tiparvovec administration.
Figure 4
Figure 4
Figure 4A: Distribution of plasma TG and cholesterol in the Sf >400 and Sf 20–400 fractions before therapy and 52 weeks after alipogene tiparvovec administration. Figure 4B: Total APOB in the Sf 20–400 fraction before therapy and 12 and 52 weeks after alipogene tiparvovec administration.

References

    1. Brunzell JD, Deeb SS. Familial lipoprotein lipase deficiency, Apo C-II deficiency and hepatic lipase deficiency. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The Metabolic Basis of Inherited Disease. 8. New York, NY: McGraw-Hill; 2001. pp. 2789–2816.
    1. Black DM, Sprecher DL. Dietary treatment and growth of hyperchylomicronemic children severely restricted in dietary fat. Am J Dis Child. 1993;147:60–62.
    1. Chait A, Robertson HT, Brunzell JD. Chylomicronemia syndrome in diabetes mellitus. Diabetes Care. 1981;4:343–348.
    1. Chait A, Brunzell JD. Chylomicronemia syndrome. Adv Int Med. 1992;37:249–273.
    1. Santamarina-Fojo S. The familial chylomicronemia syndrome. Endocrinol Metab Clin North Am. 1998;27:551–567.
    1. Benlian P, De Gennes JL, Foubert L, Zhang H, Gagné SE, Hayden M. Premature atherosclerosis in patients with familial chylomicronaemia caused by mutations in the lipoprotein lipase gene. N Engl J Med. 1996;335:848–854.
    1. Fortson MR, Freedman SN, Webster PD., III Clinical assessment of hyperlipidemic pancreatitis. Am J Gastroenterol. 1995;90:2134–2139.
    1. Goldberg IJ. Lipoprotein lipase and lipolysis: central roles in lipoprotein metabolism and atherogenesis. J Lipid Res. 1996;37:693–707.
    1. Mahley RW, Ji ZS. Remnant lipoprotein metabolism: key pathways involving cell-surface heparan sulfate proteoglycans and apolipoprotein. E J Lipid Res. 1999;40:1–16.
    1. Mead JR, Irvine SA, Ramji DP. Lipoprotein lipase: structure, function, regulation, and role in disease. J Mol Med. 2002;80:753–769.
    1. Mössner J, Bödeker H, Kimura W, Meyer F, Böhm S, Fischbach W. Isolated rat pancreatic acini as a model to study the potential role of lipase in the pathogenesis of acinar cell destruction. Int J Pancreatol. 1992;12:285–296.
    1. Kimura W, Meyer F, Hess D, Kirchner T, Fischbach W, Mössner J. Comparison of different treatment modalities in experimental pancreatitis in rats. Gastroenterology. 1992;103:1916–1924.
    1. Kimura W, Mössner J. Role of hypertriglyceridemia in the pathogenesis of experimental acute pancreatitis in rats. Int J Pancreatol. 1996;20:177–184.
    1. Saharia P, Margolis S, Zuidema GD, Cameron JL. Acute pancreatitis with hyperlipemia: studies with an isolated perfused canine pancreas. Surgery. 1977;82:60–67.
    1. Wang Y, Sternfeld L, Yang F, Rodriguez JA, Ross C, Hayden MR, et al. Enhanced susceptibility to pancreatitis in severe hypertriglyceridaemic lipoprotein lipase-deficient mice and agonist-like function of pancreatic lipase in pancreatic cells. Gut. 1992;58:422–430.
    1. Tremblay K, Méthot J, Brisson D, Gaudet D. Etiology and risk of lactescent plasma and severe hypertriglyceridemia. J Clin Lipidol. 2011;5:37–44.
    1. Gaudet D, de Wal J, Tremblay K, Déry S, van Deventer S, Freidig A, et al. Review of the clinical development of alipogene tiparvovec gene therapy for lipoprotein lipase deficiency. Atheroscler Suppl. 2010;11:55–60.
    1. Brisson D, Méthot J, Tremblay K, Tremblay M, Perron P, Gaudet D. Comparison of the efficacy of fibrates on hypertriglyceridemic phenotypes with different genetic and clinical characteristics. Pharmacogenet Genomics. 2010;20:742–747.
    1. Schnepp BC, Jensen RL, Clark KR, Johnson PR, et al. Infectious molecular clones of Adeno-associated virus isolated directly from human tissues. J Virol. 2009;83:1456–1464.
    1. Daya S, Berns KI. Gene therapy using adeno-associated virus vectors. Clin Microbiol Rev. 2008;21:583–593.
    1. Rip J, Nierman MC, Sierts JA, Petersen W, Van den Oever K, Van Raalte D, et al. Gene therapy for lipoprotein lipase deficiency: working toward clinical application. Hum Gene Ther. 2005;16:1276–1286.
    1. Ross CJ, Twisk J, Bakker AC, Miao F, Verbart D, Rip J, et al. Correction of feline lipoprotein lipase deficiency with adeno-associated virus serotype 1-mediated gene transfer of the lipoprotein lipase S447X beneficial mutation. Hum Gene Ther. 2006;17:487–499.
    1. Ross CJ, Twisk J, Meulenberg JM, Liu G, van den Oever K, Moraal E, et al. Long-term correction of murine lipoprotein lipase deficiency with AAV1-mediated gene transfer of the naturally occurring LPL(S447X) beneficial mutation. Hum Gene Ther. 2004;15:906–919.
    1. Rip J, Nierman MC, Ross CJ, Jukema JW, Hayden MR, Kastelein JJ, et al. Lipoprotein lipase S447X: a naturally occurring gain-of-function mutation. Arterioscler Thromb Vasc Biol. 2006;26:1236–1245.
    1. Stroes ES, Nierman MC, Meulenberg JJ, Franssen R, Twisk J, Henny CP, et al. Intramuscular administration of AAV1-lipoprotein lipase S447X lowers triglycerides in lipoprotein lipase-deficient patients. Arterioscler Thromb Vasc Biol. 2008;28:2303–2304.
    1. Mingozzi F, Meulenberg JJ, Hui DJ, Basner-Tschakarjan E, Hasbrouck NC, Edmonson SA, et al. AAV-1-mediated gene transfer to skeletal muscle in humans results in dose-dependent activation of capsid-specific T cells. Blood. 2009;114:2077–2086.
    1. Burnett JR, Hooper AJ. Alipogene tiparvovec, an adeno-associated virus encoding the Ser(447)X variant of the human lipoprotein lipase gene for the treatment of patients with lipoprotein lipase deficiency. Curr Opin Mol Ther. 2009;11:681–691.
    1. Clinical . Safety and Efficacy in LPL-Deficient Subjects of AMT-011, an Adeno-Associated Viral Vector Expressing Human Lipoprotein Lipase [S447X] [Last accessed on September 27 2010];Protocol summary. at .
    1. Jiang H, Couto LB, Patarroyo-White S, Liu T, Nagy D, Vargas JA, et al. Effects of transient immunosuppression on adenoassociated, virus-mediated, liver-directed gene transfer in rhesus macaques and implications for human gene therapy. Blood. 2006;108:3321–3328.
    1. Brantly ML, Chulay JD, Wang L, Mueller C, Humphries M, Spencer LT, et al. Sustained transgene expression despite T lymphocyte responses in a clinical trial of rAAV1-AAT gene therapy. Proc Natl Acad Sci U S A. 2009;106:16363–16368.
    1. Jiang H, Pierce GF, Ozelo MC, de Paula EV, Vargas JA, Smith P, et al. Evidence of multiyear factor IX expression by AAV-mediated gene transfer to skeletal muscle in an individual with severe hemophilia B. Mol Ther. 2006;14:452–455.
    1. Flotte TR, Trapnell BC, Humphries M, Carey B, Calcedo R, Rouhani F, et al. Phase 2 Clinical Trial of a Recombinant Adeno-associated Virus Vector Expressing Alpha 1 Antitrypsin: Interim Results. Hum Gene Ther. 2011;22:1239–1247.
    1. Carpentier A, Frisch F, Labbe SM, Methot J, Gagné C, Déry S, et al. Gene Therapy With Alipogene Tiparvovec Results In Enhanced Post-prandial Clearance of Chylomicrons In LPLD Patients. J Clin Endocrinol Metab. 2012 (in press)
    1. Gaudet D, Methot J, Gagné C, Déry S, Tremblay K, De Wal J, et al. Modifications in Triglyceride-Rich Lipoprotein Metabolism Induced by Alipogene Tiparvovec (AAV1-LPLS447X Gene Therapy) Correlate With Clinical Benefit in Patients with Lipoprotein Lipase Deficiency (LPLD). Circulation; Proceedings of the American Heart Association; November 2010; Abstract 21355.
    1. The Society for Surgery of the Alimentary Tract. [last accessed 12 July 2011];Guideline of on the treatment of acute pancreatitis. 2004 ( )
    1. Iverius PH, Brunzell JD. Human adipose tissue lipoprotein lipase: changes with feeding and relation to postheparin plasma enzyme. Am J Physiol. 1985;249:E107–114.
    1. Aronica E, van Kempen AA, van der Heide M, Poll-The BT, van Slooten HJ, Troost D, et al. Congenital disorder of glycosylation type Ia: a clinicopathological report of a newborn infant with cerebellar pathology. Acta Neuropathol. 2005;109:433–442.
    1. Miller FW, Love LA, Barbieri SA, Balow JE, Plotz PH. Lymphocyte activation markers in idiopathic myositis: changes with disease activity and differences among clinical and autoantibody subgroups. Clin Exp Immunol. 1990;81:373–379.
    1. Troost D, Das PK, van den Oord JJ, Louwerse ES. Immunohistological alterations in muscle of patients with amyotrophic lateral sclerosis: mononuclear cell phenotypes and expression of MHC products. 1992;11:115–120.

Source: PubMed

3
구독하다