Recent Developments in Gene Therapy for Homozygous Familial Hypercholesterolemia
详细信息    查看全文
  • 作者:Ezim Ajufo ; Marina Cuchel
  • 关键词:Gene therapy ; Homozygous familial hypercholesterolemia ; LDL receptor ; Adeno ; associated virus (AAV) ; Adenovirus ; iPSC
  • 刊名:Current Atherosclerosis Reports
  • 出版年:2016
  • 出版时间:May 2016
  • 年:2016
  • 卷:18
  • 期:5
  • 全文大小:351 KB
  • 参考文献:1.Sjouke B, Kusters DM, Kindt I, Besseling J, Defesche JC, Sijbrands EJG, et al. Homozygous autosomal dominant hypercholesterolaemia in the Netherlands: prevalence, genotype-phenotype relationship, and clinical outcome. Eur Heart J. 2015;36:560–5.CrossRef PubMed
    2.Cuchel M, Bruckert E, Ginsberg HN, Raal FJ, Santos RD, Hegele RA, et al. Homozygous familial hypercholesterolaemia: new insights and guidance for clinicians to improve detection and clinical management. A position paper from the Consensus Panel on Familial Hypercholesterolaemia of the European Atherosclerosis Society. Eur Heart J England. 2014;35:2146–57.CrossRef
    3.Goldstein JL, Brown MS. The LDL receptor defect in familial hypercholesterolemia. Implications for pathogenesis and therapy. Med Clin North Am UNITED STATES. 1982;66:335–62.
    4.Kolansky DM, Cuchel M, Clark BJ, Paridon S, McCrindle BW, Wiegers SE, et al. Longitudinal evaluation and assessment of cardiovascular disease in patients with homozygous familial hypercholesterolemia. Am J Cardiol. 2008;102:1438–43.CrossRef PubMed
    5.Raal FJ, Honarpour N, Blom DJ, Hovingh GK, Xu F, Scott R, et al. Inhibition of PCSK9 with evolocumab in homozygous familial hypercholesterolaemia (TESLA part B): a randomised, double-blind, placebo-controlled trial. Lancet. 2014/10/06 ed. Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa. Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, USA. Division of Lipidology, Department of Medicine, University of; 2015;385:341–50.
    6.Raal FJ, Pappu AS, Illingworth DR, Pilcher GJ, Marais AD, Firth JC, et al. Inhibition of cholesterol synthesis by atorvastatin in homozygous familial hypercholesterolaemia. Atherosclerosis. 2000;150:421–8.CrossRef PubMed
    7.Gagné C, Gaudet D, Bruckert E. Efficacy and safety of ezetimibe coadministered with atorvastatin or simvastatin in patients with homozygous familial hypercholesterolemia. Circulation. 2002;105:2469–75.CrossRef PubMed
    8.Raal FJ, Pilcher GJ, Panz VR, Van Deventer HE, Brice BC, Blom DJ, et al. Reduction in mortality in subjects with homozygous familial hypercholesterolemia associated with advances in lipid-lowering therapy. Circulation. 2011;124:2202–7.CrossRef PubMed
    9.Thompson GR. LDL apheresis. Atherosclerosis Ireland. 2003;167:1–13.CrossRef
    10.Raal FJ, Santos RD, Blom DJ, Marais AD, Charng M-J, Cromwell WC, et al. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet. 2010;375:998–1006.CrossRef PubMed
    11.Cuchel M, Meagher EA, du Toit Theron H, Blom DJ, Marais AD, Hegele RA, et al. Efficacy and safety of a microsomal triglyceride transfer protein inhibitor in patients with homozygous familial hypercholesterolaemia: a single-arm, open-label, phase 3 study. Lancet. 2012/11/06 ed. Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA. mcuchel@mail.med.upenn.edu; 2013;381:40–6.
    12.Bilheimer DW, Goldstein JL, Grundy SM, Starzl TE, Brown MS. Liver transplantation to provide low-density-lipoprotein receptors and lower plasma cholesterol in a child with homozygous familial hypercholesterolemia. N Engl J Med. 1984;311:1658–64.CrossRef PubMed PubMedCentral
    13.Ibrahim M, El-Hamamsy I, Barbir M, Yacoub MH. Translational lessons from a case of combined heart and liver transplantation for familial hypercholesterolemia 20 years post-operatively. J Cardiovasc Transl Res United States. 2012;5:351–8.CrossRef
    14.Kucukkartallar T, Yankol Y, Kanmaz T, Topaloglu S, Acarli K, Kalayoglu M. Liver transplantation as a treatment option for three siblings with homozygous familial hypercholesterolemia. Pediatr Transplant Denmark. 2011;15:281–4.CrossRef
    15.Grossman M, Rader DJ, Muller DWM, Kolansky DM, Kozarsky K, Clark BJ, et al. A pilot study of ex vivo gene therapy for homozygous familial hypercholesterolaemia. Nat Med. 1995;1:1148–54.CrossRef PubMed
    16.Chowdhury JR, Grossman M, Gupta S, Chowdhury NR, Baker JR, Wilson JM. Long-term improvement of hypercholesterolemia after ex vivo gene therapy in LDLR-deficient rabbits. Science. 1991;254:1802–5.CrossRef PubMed
    17.Van Craeyveld E, Jacobs F, Gordts SC, De Geest B. Gene therapy for familial hypercholesterolemia. Curr Pharm Des. 2011;17:2575–91.CrossRef PubMed
    18.Al-Allaf FA, Coutelle C, Waddington SN, David AL, Harbottle R, Themis M. LDLR-gene therapy for familial hypercholesterolaemia: problems, progress, and perspectives. Int Arch Med. 2010;3:36.CrossRef PubMed PubMedCentral
    19.Buning H, Huber A, Zhang L, Meumann N, Hacker U. Engineering the AAV capsid to optimize vector-host-interactions. Curr Opin Pharmacol. 2015;24:94–104.CrossRef PubMed
    20.Hastie E, Samulski RJ. AAV at 50: a golden anniversary of discovery, research, and gene therapy success, a personal perspective. Hum. Gene Ther. 2015;1–24.
    21.Hermonat PL, Muzyczka N. Use of adeno-associated virus as a mammalian DNA cloning vector: transduction of neomycin resistance into mammalian tissue culture cells. Proc Natl Acad Sci U S A. 1984;81:6466–70.CrossRef PubMed PubMedCentral
    22.Mingozzi F, High KA. Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nat Rev Genet. Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved.; 2011;12:341–55.
    23.Gao G-P, Alvira MR, Wang L, Calcedo R, Johnston J, Wilson JM. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci U S A. 2002;99:11854–9.CrossRef PubMed PubMedCentral
    24.Lisowski L, Tay SS, Alexander IE. Adeno-associated virus serotypes for gene therapeutics. Curr Opin Pharmacol. 2015;24:59–67.CrossRef PubMed
    25.Journal of Gene Medicine. Gene therapy clinical trials worldwide [Internet]. Wiley. 2014 [cited 2015 Dec 12]. p. Charts & Tables, Vectors. Available from: http://​www.​abedia.​com/​wiley/​
    26.•
Kotterman MA, Schaffer DV. Engineering adeno-associated viruses for clinical gene therapy. Nat Rev Genet. 2014;15:445–51. This review provides a good overview of key bioengineering approaches being employed to improve AAV vectors.CrossRef PubMed PubMedCentral
27.Bowles DE, McPhee SWJ, Li C, Gray SJ, Samulski JJ, Camp AS, et al. Phase 1 gene therapy for Duchenne muscular dystrophy using a translational optimized AAV vector. Mol Ther United States. 2012;20:443–55.CrossRef
28.Gaudet D, Méthot J, Déry S, Brisson D, Essiembre C, Tremblay G, et al. Efficacy and long-term safety of alipogene tiparvovec (AAV1-LPLS447X) gene therapy for lipoprotein lipase deficiency: an open-label trial. Gene Ther. 2013;20:361–9.CrossRef PubMed
29.Gaudet D, Méthot J, Kastelein J. Gene therapy for lipoprotein lipase deficiency. Curr Opin Lipidol. 2012;23:310–20.CrossRef PubMed
30.Nathwani AC, Tuddenham EGD, Rangarajan S, Rosales C, McIntosh J, Linch DC, et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med. 2011;365:2357–65.CrossRef PubMed PubMedCentral
31.••
Nathwani AC, Reiss UM, Tuddenham EGD, Rosales C, Chowdary P, McIntosh J, et al. Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N Engl J Med. 2014;371:1994–2004. This paper presents three-year follow-up data from the first liver-directed gene therapy clinical trial with AAV8 in hemophilia B. All of the subjects in this trial achieved steady-state serum FIX levels between 1-7% of normal from baseline levels of <1%. Capsid-specific T cells were associated with a reversible rise in transaminase in 4/6 patients that received the highest dose of the vector which was associated with a decline in serum FIX levels. This trial is of particular significance for the HoFH clinical trial underway due to key parallels between the two diseases being treated and use of the same vector and target.CrossRef PubMed PubMedCentral
32.Boutin S, Monteilhet V, Veron P, Leborgne C, Benveniste O, Montus MF, et al. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors. Hum Gene Ther United States. 2010;21:704–12.CrossRef
33.Lebherz C, Gao G, Louboutin J-P, Millar J, Rader D, Wilson JM. Gene therapy with novel adeno-associated virus vectors substantially diminishes atherosclerosis in a murine model of familial hypercholesterolemia. J Gene Med. 2004;6:663–72.CrossRef PubMed
34.Powell-Braxton L, Véniant M, Latvala RD, Hirano KI, Won WB, Ross J, et al. A mouse model of human familial hypercholesterolemia: markedly elevated low density lipoprotein cholesterol levels and severe atherosclerosis on a low-fat chow diet. Nat Med. 1998;4:934–8.CrossRef PubMed
35.Kassim SH, Li H, Vandenberghe LH, Hinderer C, Bell P, Marchadier D, et al. Gene therapy in a humanized mouse model of familial hypercholesterolemia leads to marked regression of atherosclerosis. PLoS One. 2010;5, e13424.CrossRef PubMed PubMedCentral
36.••
Kassim SH, Li H, Bell P, Somanathan S, Lagor W, Jacobs F, et al. Adeno-associated virus serotype 8 gene therapy leads to significant lowering of plasma cholesterol levels in humanized mouse models of homozygous and heterozygous familial hypercholesterolemia. Hum Gene Ther. 2013;24:19–26. In this follow-up to the 2010 Kassim et al. manuscript referenced above, a more advanced humanised mouse model was used to evaluate the efficacy and safety of a human LDLR transgene in an AAV8 vector. This agent is now in clinicial testing. Key safety analyses reported by this paper include transgene and capsid-specific T cell responses, liver histopathology, liver lipid accumulation and serum transaminases. These were found to be within normal limits.CrossRef PubMed PubMedCentral
37.Lisowski L, Dane AP, Chu K, Zhang Y, Cunningham SC, Wilson EM, et al. Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature England. 2014;506:382–6.CrossRef
38.Nathwani AC, Gray JT, Ng CYC, Zhou J, Spence Y, Waddington SN, et al. Self-complementary adeno-associated virus vectors containing a novel liver-specific human factor IX expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood. 2006;107:2653–61.CrossRef PubMed PubMedCentral
39.Herzog RW. Immune responses to AAV capsid: are mice not humans after all [quest]. Mol Ther. 2007;15:649–50.CrossRef PubMed
40.Martino AT, Basner-Tschakarjan E, Markusic DM, Finn JD, Hinderer C, Zhou S, et al. Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-specific CD8+ T cells. Blood. 2013;121:2224–33.CrossRef PubMed PubMedCentral
41.Calcedo R, Vandenberghe LH, Gao G, Lin J, Wilson JM. Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. J Infect Dis. 2009;199:381–90.CrossRef PubMed
42.Mingozzi F, Maus MV, Hui DJ, Sabatino DE, Murphy SL, Rasko JEJ, et al. CD8(+) T-cell responses to adeno-associated virus capsid in humans. Nat Med. 2007;13:419–22.CrossRef PubMed
43.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 United States. 2006;12:342–7.
44.Liu Q, Huang W, Zhang H, Wang Y, Zhao J, Song A, et al. Neutralizing antibodies against AAV2, AAV5 and AAV8 in healthy and HIV-1-infected subjects in China: implications for gene therapy using AAV vectors. Gene Ther. 2014;21:732–8.CrossRef PubMed
45.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.CrossRef PubMed PubMedCentral
46.Hauck B, Murphy SL, Smith PH, Qu G, Liu X, Zelenaia O, et al. Undetectable transcription of cap in a clinical AAV vector: implications for preformed capsid in immune responses. Mol Ther United States. 2009;17:144–52.CrossRef
47.Li C, Goudy K, Hirsch M, Asokan A, Fan Y, Alexander J, et al. Cellular immune response to cryptic epitopes during therapeutic gene transfer. Proc Natl Acad Sci U S A. 2009;106:10770–4.CrossRef PubMed PubMedCentral
48.Vandenberghe LH, Wang L, Somanathan S, Zhi Y, Figueredo J, Calcedo R, et al. Heparin binding directs activation of T cells against adeno-associated virus serotype 2 capsid. Nat Med. 2006;12:967–71.CrossRef PubMed
49.Hui DJ, Basner-Tschakarjan E, Chen Y, Davidson RJ, Buchlis G, Yazicioglu M, et al. Modulation of CD8+ T cell responses to AAV vectors with IgG-derived MHC class II epitopes. Mol Ther. 2013;21:1727–37.CrossRef PubMed PubMedCentral
50.Maheshri N, Koerber JT, Kaspar BK, Schaffer DV. Directed evolution of adeno-associated virus yields enhanced gene delivery vectors. Nat Biotechnol. 2006;24:198–204.CrossRef PubMed
51.Finn JD, Nichols TC, Svoronos N, Merricks EP, Bellenger DA, Zhou S, et al. The efficacy and the risk of immunogenicity of FIX Padua (R338L) in hemophilia B dogs treated by AAV muscle gene therapy. Blood. 2012;120:4521–3.CrossRef PubMed PubMedCentral
52.Baxalta reports continued progress on phase 1/2 clinical trial of BAX335, investigational gene therapy treatment for hemophilia b [Internet]. [cited 2015 Dec 7]. Available from: http://​www.​businesswire.​com/​news/​home/​20150624005677/​en/​Baxalta-Reports-Continued-Progress-Phase-12-Clinical
53.•
Somanathan S, Jacobs F, Wang Q, Hanlon AL, Wilson JM, Rader DJ. AAV vectors expressing LDLR gain-of-function variants demonstrate increased efficacy in mouse models of familial hypercholesterolemia. Circ Res. 2014;115:591–9. This interesting paper presents in vitro and in vivo efficacy data from three engineered gain-of-function LDLR transgenes which improved LDLR expression and cholesterol reduction compared to the wild-type LDLR transgene.CrossRef PubMed PubMedCentral
54.Hibbitt OC, McNeil E, Lufino MM, Seymour L, Channon K, Wade-Martins R. Long-term physiologically regulated expression of the low-density lipoprotein receptor in vivo using genomic DNA mini-gene constructs. Mol Ther Nature Publishing Group. 2010;18:317–26.
55.•Hibbitt O, Agkatsev S, Owen C, Cioroch M, Seymour L, Channon K, et al. RNAi-mediated knockdown of HMG CoA reductase enhances gene expression from physiologically regulated low-density lipoprotein receptor therapeutic vectors in vivo. Gene Ther. 2012;19:463–7. This study showed that combining RNA-mediated knockdown of HMGCR with LDLR gene transfer increased LDLR gene expression up to 300-fold in vitro with significant metabolic effects in LDLR knockout mice.CrossRef PubMed
56.Rashid ST, Corbineau S, Hannan N, Marciniak SJ, Miranda E, Alexander G, et al. Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J Clin Invest. 2010;120:3127–36.CrossRef PubMed PubMedCentral
57.Cayo MA, Cai J, DeLaForest A, Noto FK, Nagaoka M, Clark BS, et al. JD induced pluripotent stem cell-derived hepatocytes faithfully recapitulate the pathophysiology of familial hypercholesterolemia. Hepatology. 2012;56:2163–71.CrossRef PubMed PubMedCentral
58.Fattahi F, Asgari S, Pournasr B, Seifinejad A, Totonchi M, Taei A, et al. Disease-corrected hepatocyte-like cells from familial hypercholesterolemia-induced pluripotent stem cells. Mol Biotechnol. 2013;54:863–73.CrossRef PubMed
59.•Ramakrishnan VM, Yang J-Y, Tien KT, McKinley TR, Bocard BR, Maijub JG, et al. Restoration of physiologically responsive low-density lipoprotein receptor-mediated endocytosis in genetically deficient induced pluripotent stem cells. Sci Rep England. 2015;5:13231. This in vitro study, building on the Fattahi et al. study referenced above, showed that autologous iPSCs from HoFH patients could be genetically corrected using an LDLR expressing plasmid and differentiated into hepatocyte-like cells with restored LDL internalisation responsive to extracellular sterol and lovostatin.CrossRef
60.Espejel S, Roll GR, Mclaughlin KJ, Lee AY, Zhang JY, Laird DJ, et al. Induced pluripotent stem cell-derived hepatocytes have the functional and proliferative capabilities needed for liver regeneration in mice. J Clin Invest. 2010;120:3120–6.CrossRef PubMed PubMedCentral
61.Cantz T, Sharma AD, Ott M. Concise review: cell therapies for hereditary metabolic liver diseases-concepts, clinical results, and future developments. Stem Cells. 2015;33:1055–62.CrossRef PubMed
62.Ordonez MP, Goldstein LSB. Using human-induced pluripotent stem cells to model monogenic metabolic disorders of the liver. Semin Liver Dis. 2012;32:298–306.PubMed
  • 作者单位:Ezim Ajufo (1)
    Marina Cuchel (1)

    1. Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
  • 刊物主题:Angiology; Cardiology;
  • 出版者:Springer US
  • ISSN:1534-6242
  • 文摘
    Homozygous familial hypercholesterolemia (HoFH) is a life-threatening Mendelian disorder with a mean life expectancy of 33 years despite maximally tolerated standard lipid-lowering therapies. This disease is an ideal candidate for gene therapy, and in the last few years, a number of exciting developments have brought this approach closer to the clinic than ever before. In this review, we discuss in detail the most advanced of these developments, a recombinant adeno-associated virus (AAV) vector carrying a low-density lipoprotein receptor (LDLR) transgene which has recently entered phase 1/2a testing. We also review ongoing development of approaches to enhance transgene expression, improve the efficiency of hepatocyte transduction, and minimize the AAV capsid-specific adaptive immune response. We include a summary of key gene therapy approaches for HoFH in pre-clinical development, including RNA silencing of the gene encoding HMG-CoA reductase (HMGCR) and induced pluripotent stem cell transplant therapy.

    © 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

    地址:北京市海淀区学院路29号 邮编:100083

    电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700