Adeno-Associated Virus Vector–Based Gene Therapy for Monogenetic Metabolic Diseases of the Liver
Norman Junge
Paediatric Gastroenterology and Hepatology, Children's Hospital, Hannover Medical School, Hannover, Germany
Search for more papers by this authorMichael Ott
Twincore, Centre for Experimental and Clinical Infection Research, Hannover, Germany
Search for more papers by this authorCorresponding Author
Ulrich Baumann
Paediatric Gastroenterology and Hepatology, Children's Hospital, Hannover Medical School, Hannover, Germany
Address correspondence and reprint requests to Ulrich Baumann, MD, Hannover Medical School, Paediatric Gastroenterology and Hepatology, Carl-Neuberg-Str 1, 30625 Hannover, Germany (e-mail: [email protected]).Search for more papers by this authorNorman Junge
Paediatric Gastroenterology and Hepatology, Children's Hospital, Hannover Medical School, Hannover, Germany
Search for more papers by this authorMichael Ott
Twincore, Centre for Experimental and Clinical Infection Research, Hannover, Germany
Search for more papers by this authorCorresponding Author
Ulrich Baumann
Paediatric Gastroenterology and Hepatology, Children's Hospital, Hannover Medical School, Hannover, Germany
Address correspondence and reprint requests to Ulrich Baumann, MD, Hannover Medical School, Paediatric Gastroenterology and Hepatology, Carl-Neuberg-Str 1, 30625 Hannover, Germany (e-mail: [email protected]).Search for more papers by this authorF.M. holds patents describing the AAV technology for gene transfer to the liver and has consulted to numerous commercial entities that have developed AAV-based products. The other authors report no conflicts of interest.
ABSTRACT
Liver-based metabolic diseases account for a substantial burden of childhood diseases. In most patients, treatment is often limited to supportive measures and liver transplantation is ultimately required. Even despite the excellent long-term outcome of liver transplantation, the procedure is associated with a significant morbidity and mortality. Gene therapy, in contrast, has great potential to save lives, improve the quality of life, and offer few risks and adverse effects compared with present therapies including liver transplantation. The most promising results to date in liver gene transfer have been achieved with adeno-associated virus. Although safety issues, such as immunogenicity of vector and/or transgene product, remain an important concern, gene therapy is ready for clinical trials in adults and adolescents. Developing and testing safe approaches for efficient and long-term stable applications in newborns and small children, such as targeted integration and gene correction, is one of the remaining future challenges.
REFERENCES
- 1McCandless SE, Brunger JW, Cassidy SB. The burden of genetic disease on inpatient care in a children's hospital. Am J Hum Genet 2004; 74: 121–127.
- 2Rosenberg SA, Aebersold P, Cornetta K, et al. Gene transfer into humans--immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N Engl J Med 1990; 323: 570–578.
- 3Raper SE, Chirmule N, Lee FS, et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab 2003; 80: 148–158.
- 4Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 2000; 288: 669–672.
- 5Hacein-Bey-Abina S, Hauer J, Lim A, et al. Efficacy of gene therapy for X-linked severe combined immunodeficiency. N Engl J Med 2010; 363: 355–364.
- 6Braun CJ, Boztug K, Paruzynski A, et al. Gene therapy for Wiskott-Aldrich syndrome--long-term efficacy and genotoxicity. Sci Transl Med 2014; 6: 227ra33.
- 7Naldini L. Ex vivo gene transfer and correction for cell-based therapies. Nat Rev Genet 2011; 12: 301–315.
- 8Aiuti A, Biasco L, Scaramuzza S, et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science 2013; 341: 1233151.
- 9Cartier N, Hacein-Bey-Abina S, Bartholomae CC, et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 2009; 326: 818–823.
- 10Biffi A, Montini E, Lorioli L, et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science 2013; 341: 1233158.
- 11Cavazzana-Calvo M, Payen E, Negre O, et al. Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature 2010; 467: 318–322.
- 12Bainbridge JW, Smith AJ, Barker SS, et al. Effect of gene therapy on visual function in Leber's congenital amaurosis. N Engl J Med 2008; 358: 2231–2239.
- 13Cideciyan AV, Aleman TS, Boye SL, et al. Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics. Proc Natl Acad Sci U S A 2008; 105: 15112–15117.
- 14Maguire AM, Simonelli F, Pierce EA, et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med 2008; 358: 2240–2248.
- 15Nathwani AC, Tuddenham EG, Rangarajan S, et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med 2011; 365: 2357–2365.
- 16Manno CS, Pierce GF, Arruda VR, et al. Successful transduction of liver in hemophilia by AAV-factor IX and limitations imposed by the host immune response. Nat Med 2006; 12: 342–347.
- 17Kaufmann KB, Buning H, Galy A, et al. Gene therapy on the move. EMBO Mol Med 2013; 5: 1642–1661.
- 18Li H, Malani N, Hamilton SR, et al. Assessing the potential for AAV vector genotoxicity in a murine model. Blood 2011; 117: 3311–3319.
- 19Li H, Haurigot V, Doyon Y, et al. In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature 2011; 475: 217–221.
- 20Paulk NK, Wursthorn K, Wang Z, et al. Adeno-associated virus gene repair corrects a mouse model of hereditary tyrosinemia in vivo. Hepatology 2010; 51: 1200–1208.
- 21Sawyer GJ, Rela M, Davenport M, et al. Hydrodynamic gene delivery to the liver: theoretical and practical issues for clinical application. Curr Gene Ther 2009; 9: 128–135.
- 22Lungwitz U, Breunig M, Blunk T, et al. Polyethylenimine-based non-viral gene delivery systems. Eur J Pharm Biopharm 2005; 60: 247–266.
- 23Mingozzi F, High KA. Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nat Rev Genet 2011; 12: 341–355.
- 24Tebas P, Stein D, Tang WW, et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med 2014; 370: 901–910.
- 25Inagaki K, Piao C, Kotchey NM, et al. Frequency and spectrum of genomic integration of recombinant adeno-associated virus serotype 8 vector in neonatal mouse liver. J Virol 2008; 82: 9513–9524.
- 26Donsante A, Miller DG, Li Y, et al. AAV vector integration sites in mouse hepatocellular carcinoma. Science 2007; 317: 477.
- 27Niemeyer GP, Herzog RW, Mount J, et al. Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy. Blood 2009; 113: 797–806.
- 28Nathwani AC, Rosales C, McIntosh J, et al. Long-term safety and efficacy following systemic administration of a self-complementary AAV vector encoding human FIX pseudotyped with serotype 5 and 8 capsid proteins. Mol Ther 2011; 19: 876–885.
- 29Li H, Malani N, Hamilton SR, et al. Assessing the potential for AAV vector genotoxicity in a murine model. Blood 2011; 117: 3311–3319.
- 30Donsante A, Vogler C, Muzyczka N, et al. Observed incidence of tumorigenesis in long-term rodent studies of rAAV vectors. Gene Ther 2001; 8: 1343–1346.
- 31Wellman JA, Mingozzi F, Ozelo MC, et al. Results from the long-term follow-up of severe hemophilia B subjects previously enrolled in a clinical study of AAV2-FIX gene transfer to the liver. Mol Ther 2012; 20: 28–29.
- 32Jiang H, Couto LB, Patarroyo-White S, 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.
- 33Scallan CD, Jiang H, Liu T, et al. Human immunoglobulin inhibits liver transduction by AAV vectors at low AAV2 neutralizing titers in SCID mice. Blood 2006; 107: 1810–1817.
- 34Calcedo R, Vandenberghe LH, Gao G, et al. Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. J Infect Dis 2009; 199: 381–390.
- 35Calcedo R, Morizono H, Wang L, et al. Adeno-associated virus antibody profiles in newborns, children, and adolescents. Clin Vaccine Immunol 2011; 18: 1586–1588.
- 36Li C, Narkbunnam N, Samulski RJ, et al. Neutralizing antibodies against adeno-associated virus examined prospectively in pediatric patients with hemophilia. Gene Ther 2012; 19: 288–294.
- 37Masat E, Pavani G, Mingozzi F. Humoral immunity to AAV vectors in gene therapy: challenges and potential solutions. Discov Med 2013; 15: 379–389.
- 38Mingozzi F, Anguela XM, Pavani G, et al. Overcoming preexisting humoral immunity to AAV using capsid decoys. Sci Transl Med 2013; 5: 194ra92.
- 39Mingozzi F, Maus MV, Hui DJ, et al. CD8(+) T-cell responses to adeno-associated virus capsid in humans. Nat Med 2007; 13: 419–422.
- 40Vandenberghe LH, Wang L, Somanathan S, et al. Heparin binding directs activation of T cells against adeno-associated virus serotype 2 capsid. Nat Med 2006; 12: 967–971.
- 41Mingozzi F, Meulenberg JJ, Hui DJ, 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.
- 42Montenegro-Miranda PS, ten Bloemendaal L, Kunne C, et al. Mycophenolate mofetil impairs transduction of single-stranded adeno-associated viral vectors. Hum Gene Ther 2011; 22: 605–612.
- 43Unzu C, Hervas-Stubbs S, Sampedro A, et al. Transient and intensive pharmacological immunosuppression fails to improve AAV-based liver gene transfer in non-human primates. J Transl Med 2012; 10: 122.
- 44Finn JD, Hui D, Downey HD, et al. Proteasome inhibitors decrease AAV2 capsid derived peptide epitope presentation on MHC class I following transduction. Mol Ther 2010; 18: 135–142.
- 45Martino AT, Basner-Tschakarjan E, Markusic DM, et al. Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-specific CD8+ T cells. Blood 2013; 121: 2224–2233.
- 46Hui DJ, Basner-Tschakarjan E, Chen Y, et al. Modulation of CD8+ T cell responses to AAV vectors with IgG-derived MHC class II epitopes. Mol Ther 2013; 21: 1727–1737.
- 47Mingozzi F, Liu YL, Dobrzynski E, et al. Induction of immune tolerance to coagulation factor IX antigen by in vivo hepatic gene transfer. J Clin Invest 2003; 111: 1347–1356.
- 48Rogers GL, Martino AT, Aslanidi GV, et al. Innate immune responses to AAV vectors. Front Microbiol 2011; 2: 194.
- 49Wu T, Topfer K, Lin SW, et al. Self-complementary AAVs induce more potent transgene product-specific immune responses compared to a single-stranded genome. Mol Ther 2012; 20: 572–579.
- 50Manno CS, Pierce GF, Arruda VR, et al. Successful transduction of liver in hemophilia by AAV-factor IX and limitations imposed by the host immune response. Nat Med 2006; 12: 342–347.
- 51Favre D, Provost N, Blouin V, et al. Immediate and long-term safety of recombinant adeno-associated virus injection into the nonhuman primate muscle. Mol Ther 2001; 4: 559–566.
- 52Arruda VR, Fields PA, Milner R, et al. Lack of germline transmission of vector sequences following systemic administration of recombinant AAV-2 vector in males. Mol Ther 2001; 4: 586–592.
- 53Schuettrumpf J, Liu JH, Couto LB, et al. Inadvertent germline transmission of AAV2 vector: findings in a rabbit model correlate with those in a human clinical trial. Mol Ther 2006; 13: 1064–1073.
- 54Favaro P, Downey HD, Zhou JS, et al. Host and vector-dependent effects on the risk of germline transmission of AAV vectors. Mol Ther 2009; 17: 1022–1030.
- 55Nathwani AC, Gray JT, McIntosh J, et al. Safe and efficient transduction of the liver after peripheral vein infusion of self-complementary AAV vector results in stable therapeutic expression of human FIX in nonhuman primates. Blood 2007; 109: 1414–1421.
- 56Nathwani AC, Gray JT, Ng CY, 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: 2653–2661.
- 57Finn JD, Nichols TC, Svoronos N, 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: 4521–4523.
- 58Chuah MK, Petrus I, De Bleser P, et al. Liver-specific transcriptional modules identified by genome-wide in silico analysis enable efficient gene therapy in mice and non-human primates. Mol Ther 2014; 22: 1605–1613.
- 59Gao GP, Alvira MR, Wang L, et al. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci U S A 2002; 99: 11854–11859.
- 60Nathwani AC, Reiss UM, Tuddenham EG, et al. Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N Engl J Med 2014; 371: 1994–2004.
- 61Lisowski L, Dane AP, Chu K, et al. Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature 2014; 506: 382–386.
- 62Gao GP, Alvira MR, Wang L, et al. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci U S A 2002; 99: 11854–11859.
- 63Gunn CH. Hereditary acholuric jaundice in a new mutant strain of rats. J Hered 1938; 29: 137–139.
- 64Bortolussi G, Zentilin L, Baj G, et al. Rescue of bilirubin-induced neonatal lethality in a mouse model of Crigler-Najjar syndrome type I by AAV9-mediated gene transfer. FASEB J 2012; 26: 1052–1063.
- 65Fox IJ, Chowdhury JR, Kaufman SS, et al. Treatment of the Crigler-Najjar syndrome type I with hepatocyte transplantation. N Engl J Med 1998; 338: 1422–1426.
- 66Seppen J, Bakker C, de Jong B, et al. Adeno-associated virus vector serotypes mediate sustained correction of bilirubin UDP glucuronosyltransferase deficiency in rats. Mol Ther 2006; 13: 1085–1092.
- 67Collaud F, Ronzitti G, Lacoste F, et al. An optimized AAV vector for liver-directed expression of the UGT1A1 gene in Crigler-Najjar syndrome, towards clinical development. Mol Ther 2014; 22: 110.
- 68Luo X, Hall G, Li S, et al. Hepatorenal correction in murine glycogen storage disease type I with a double-stranded adeno-associated virus vector. Mol Ther 2011; 19: 1961–1970.
- 69Lee YM, Jun HS, Pan CJ, et al. Prevention of hepatocellular adenoma and correction of metabolic abnormalities in murine glycogen storage disease type Ia by gene therapy. Hepatology 2012; 56: 1719–1729.
- 70Koeberl DD. In search of proof-of-concept: gene therapy for glycogen storage disease type Ia. J Inherit Metab Dis 2012; 35: 671–678.
- 71Li H, Haurigot V, Doyon Y, et al. In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature 2011; 475: 217–221.
- 72Kassim SH, Li H, Bell P, 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.
- 73Chandler RJ, Chandrasekaran S, Carrillo-Carrasco N, et al. Adeno-associated virus serotype 8 gene transfer rescues a neonatal lethal murine model of propionic acidemia. Hum Gene Ther 2011; 22: 477–481.
- 74Salido E, Rodriguez-Pena M, Santana A, et al. Phenotypic correction of a mouse model for primary hyperoxaluria with adeno-associated virus gene transfer. Mol Ther 2011; 19: 870–875.
- 75Chandler RJ, Tarasenko TN, Cusmano-Ozog K, et al. Liver-directed adeno-associated virus serotype 8 gene transfer rescues a lethal murine model of citrullinemia type 1. Gene Ther 2013; 20: 1188–1191.
- 76Ye X, Robinson MB, Batshaw ML, et al. Prolonged metabolic correction in adult ornithine transcarbamylase-deficient mice with adenoviral vectors. J Biol Chem 1996; 271: 3639–3646.
- 77Alexander IE, Kok C, Dane AP, et al. Gene therapy for metabolic disorders: an overview with a focus on urea cycle disorders. J Inherit Metab Dis 2012; 35: 641–645.
- 78Cunningham SC, Kok CY, Spinoulas A, et al. AAV-encoded OTC activity persisting to adulthood following delivery to newborn spf(ash) mice is insufficient to prevent shRNA-induced hyperammonaemia. Gene Ther 2013; 20: 1184–1187.
- 79Siew SM, Cunningham SC, Alexander IE. Neonatal treatment using a novel, hybrid recombinant AAVPIGGYBAC transposon vector results in robust, long-term phenotype correction of the progressive familial intrahepatic cholestasis type 3 mouse model in vivo. ESPGHAN. Hepatology 2014 [abstract no. SP-H-0157].
- 80Unzu C, Sampedro A, Mauleon I, et al. Sustained enzymatic correction by rAAV-mediated liver gene therapy protects against induced motor neuropathy in acute porphyria mice. Mol Ther 2011; 19: 243–250.
- 81Paneda A, Lopez-Franco E, Kaeppel C, et al. Safety and liver transduction efficacy of rAAV5-cohPBGD in nonhuman primates: a potential therapy for acute intermittent porphyria. Hum Gene Ther 2013; 24: 1007–1017.
- 82D'Avola D, Lopez-Franco E, Harper P, et al. Phase 1 clinical trial of liver directed gene therapy with rAAV5-PBGD in acute intermittent porphyria: preliminary safety data. Mol Ther 2014; 22 (Suppl 1): S7.
- 83Hofherr SE, Matern D. Systematic ranking of inborn errors of metabolism as targets for gene therapy. Mol Genet Metab 2010; 100: 215–218.