mRNA therapy restores euglycemia and prevents liver tumors in murine model of glycogen storage disease

Jingsong Cao, Minjung Choi, Eleonora Guadagnin, Maud Soty, Marine Silva, Vincent Verzieux, Edward Weisser, Arianna Markel, Jenny Zhuo, Shi Liang, Ling Yin, Andrea Frassetto, Anne-Renee Graham, Kristine Burke, Tatiana Ketova, Cosmin Mihai, Zach Zalinger, Becca Levy, Gilles Besin, Meredith Wolfrom, Barbara Tran, Christopher Tunkey, Erik Owen, Joe Sarkis, Athanasios Dousis, Vladimir Presnyak, Christopher Pepin, Wei Zheng, Lei Ci, Marjie Hard, Edward Miracco, Lisa Rice, Vi Nguyen, Mike Zimmer, Uma Rajarajacholan, Patrick F Finn, Gilles Mithieux, Fabienne Rajas, Paolo G V Martini, Paloma H Giangrande, Jingsong Cao, Minjung Choi, Eleonora Guadagnin, Maud Soty, Marine Silva, Vincent Verzieux, Edward Weisser, Arianna Markel, Jenny Zhuo, Shi Liang, Ling Yin, Andrea Frassetto, Anne-Renee Graham, Kristine Burke, Tatiana Ketova, Cosmin Mihai, Zach Zalinger, Becca Levy, Gilles Besin, Meredith Wolfrom, Barbara Tran, Christopher Tunkey, Erik Owen, Joe Sarkis, Athanasios Dousis, Vladimir Presnyak, Christopher Pepin, Wei Zheng, Lei Ci, Marjie Hard, Edward Miracco, Lisa Rice, Vi Nguyen, Mike Zimmer, Uma Rajarajacholan, Patrick F Finn, Gilles Mithieux, Fabienne Rajas, Paolo G V Martini, Paloma H Giangrande

Abstract

Glycogen Storage Disease 1a (GSD1a) is a rare, inherited metabolic disorder caused by deficiency of glucose 6-phosphatase (G6Pase-α). G6Pase-α is critical for maintaining interprandial euglycemia. GSD1a patients exhibit life-threatening hypoglycemia and long-term liver complications including hepatocellular adenomas (HCAs) and carcinomas (HCCs). There is no treatment for GSD1a and the current standard-of-care for managing hypoglycemia (Glycosade®/modified cornstarch) fails to prevent HCA/HCC risk. Therapeutic modalities such as enzyme replacement therapy and gene therapy are not ideal options for patients due to challenges in drug-delivery, efficacy, and safety. To develop a new treatment for GSD1a capable of addressing both the life-threatening hypoglycemia and HCA/HCC risk, we encapsulated engineered mRNAs encoding human G6Pase-α in lipid nanoparticles. We demonstrate the efficacy and safety of our approach in a preclinical murine model that phenotypically resembles the human condition, thus presenting a potential therapy that could have a significant therapeutic impact on the treatment of GSD1a.

Conflict of interest statement

J.C., M.C., E.G., E.D., J.Z., S.L., L.Y., A.F., A.-R.G., K.B., T.K., C.M., Z.Z., B.L., G.B., M.W., B.T., C.T., E.O., J.S., A.D., V.P., C.P., W.Z., L.C., M.H., E.M., L.R., V.N., M.Z., U.R., P.F.F., P.G.V.M., and P.H.G. are employees of and receive salary and stock options from Moderna Inc. M.Si., M.So., V.V., E.W., A.M., G.M., and F.R. declare no competing interests.

Figures

Fig. 1. In vitro characterization of modified…
Fig. 1. In vitro characterization of modified mRNA encoding hG6Pase-α.
a Hypothetical model of hG6PC mRNA therapy. hG6PC mRNAs are delivered to liver via lipid nanoparticles. Once the mRNA is in the cell (hepatocytes) it is translated by the cellular machinery into a functional protein that is localized to the ER membrane (likely following a co-translational translocation model), resulting in an active G6Pase-α enzyme. b Protein consensus screening by ortholog residue analysis. Top: WebLogo representation of the abundance of each alternative amino acid used at indicated residue positions. Bottom: The degree of conservation of amino acids at each position was quantified as relative entropy (Kullback–Leibler divergence). c Relative hG6Pase-α protein expression (solid circle) and hG6Pase-α enzymatic activity (solid square) in HeLa cells treated with the top ten hG6PC mRNA variants generated using protein consensus analysis. Data were shown as percentage of wild-type (WT) group and presented as mean ± SD of n = 2 (for protein expression), 3 (for enzymatic activity, Q247R), or 4 (for enzymatic activity, all other groups) biologically independent samples. d Subcellular localization of WT hG6Pase-α and S298C variant in HeLa cells. Green: hG6Pase-α, Red: Calnexin, an ER marker (top); TOM20, mitochondrial marker (bottom). Scale bars are 10 µm. The ratio of colocalized signal over total signal was calculated by Mander’s colocalization coefficient analysis (bottom panel). Data were presented as mean ± SD of n = 2 biologically independent samples. Source data are provided as a Source Data File.
Fig. 2. Effect of codon optimization on…
Fig. 2. Effect of codon optimization on expression and activity of hG6PC mRNAs.
a hG6Pase-α protein expression (left panel) and enzymatic activity (right panel) of wild-type hG6PC (WT), codon optimized wild-type hG6PC (WT_CO), hG6PC_S298C (S298C), and codon optimized hG6PC_S298C (S298C_CO) mRNAs evaluated in Hep3B cells. Control cells were treated with eGFP mRNA. Data were presented as mean ± SD of n = 3 biologically independent samples. b hG6Pase-α protein expression (left panel) and enzymatic activity (right panel) of WT and codon optimized hG6PC mRNAs as evaluated in male CD-1 mice. Control animals were treated with eGFP mRNA. Data were presented as mean ± SD of n = 4 mice. For statistical analysis, raw values were Log2 transformed and subjected to one-way ANOVA, followed by the Dunnett’s multiple comparisons test, compared to the non-codon optimized WT hG6PC mRNA. Statistically significant P values (p ≤ 0.05) are shown in the graphs. Source data are provided as a Source Data File.
Fig. 3. Hepatic hG6PC mRNA and hG6Pase-α…
Fig. 3. Hepatic hG6PC mRNA and hG6Pase-α protein and activity half-lives in wild-type mice.
Wild-type (CD-1) male mice were i.v. administered with 1.0 mg/kg of eGFP, hG6PC-wild type (WT), or codon-optimized hG6PC-S298C (hG6PC_S298C_CO) mRNA-LNP and sacrificed at 6, 24, 72, 168, 336 h (n = 4/group/sacrifice time point). a hG6PC mRNA levels (hG6PC-WT and S298C mRNAs). b Hepatic protein levels in mice treated with eGFP mRNA, mRNA encoding hG6Pase-α WT, or codon-optimized mRNA encoding hG6PC-S298C protein variant. c Hepatic enzymatic activity levels in mice treated with eGFP mRNA, mRNA encoding hG6Pase-α WT, or codon-optimized mRNA encoding hG6PC-S298C protein variant. Data were presented as mean ± SD (n = 3–4). Source data are provided as a Source Data File.
Fig. 4. Single i.v. dose of h…
Fig. 4. Single i.v. dose of hG6PC S298C mRNA-LNP restores euglycemia, as well as serum and hepatic biomarkers in L.G6pc−/− mice.
a Blood glucose levels following administration of hG6PC S298C mRNA-LNP in L.G6pc−/− mice. WT, wild-type mice. (WT treated with PBS, n = 8 per group; L.G6pc−/− treated with eGFP, n = 6, 5, 5, and 5 per group for fasting duration of 0, 2.5, 6, and 24 h, respectively; L.G6pc−/− treated with hG6PC S298C at 0.2 mg/kg, n = 7 per group for all time points; L.G6pc−/− treated with hG6PC S298C at 0.5 mg/kg, n = 7, 6, 6, and 6 per group for fasting duration of 0, 2.5, 6, and 24 h, respectively; L.G6pc−/− treated with hG6PC S298C at 1.0 mg/kg, n = 7 per group for all time points). Data were presented as mean ± SD. b Liver morphology (left panel) and liver weight (right panel) following administration of hG6PC S298C mRNA in L.G6pc−/− mice. Representative liver images are shown from n = 8, 5, 7, 6, and 6 mice per group from WT treated with PBS, L.G6pc−/− treated with eGFP, and L.G6pc−/− treated with hG6PC S298C mRNA at 0.2, 0.5, or 1.0 mg/kg, respectively. c hG6Pase-α S298C protein expression and enzymatic activity in livers of L.G6pc−/− mice. d Hepatic biomarker analysis following administration of hG6PC S298C mRNA-LNP in L.G6pc−/− mice. Liver G6P (left panel), liver glycogen (middle panel), liver triglycerides (right panel). e Serum triglycerides following administration of hG6PC S298C mRNA-LNP in L.G6pc−/− mice. hG6PC S298C mRNA-LNP dose range: 0.2, 0.5, and 1.0 mg/kg. For b–e, quantitative data were presented as mean ± SD (n = 8, 5, 7, 6, and 6 mice per group for WT treated with PBS, L.G6pc−/− treated with eGFP, and L.G6pc−/− treated hG6PC S298C mRNA at 0.2, 0.5, or 1.0 mg/kg, respectively). For statistical analysis, raw values were Log2 transformed and subjected to one-way ANOVA, followed by the Dunnett’s multiple comparisons test, compared to the eGFP mRNA treated group. Statistically significant P values (p ≤ 0.05) are shown in the graphs. Source data are provided as a Source Data File.
Fig. 5. Repeat i.v. dose of h…
Fig. 5. Repeat i.v. dose of hG6PC mRNA-LNP results in safe and effective restoration of euglycemia in L.G6pc−/− mice.
a Single-dose duration of action of hG6PC S298C mRNA-LNP (0.5 or 1.0 mg/kg) administered i.v. in L.G6pc−/− mice. Blood glucose levels were measured at fed (0 h) or fasting conditions (2.5- or 6-h post-fasting). Data were presented as mean ± SD (n = 8, 9, 10, 10, and 10 mice per group for wild-type (WT) treated with PBS, L.G6pc−/− treated with eGFP, hG6PC S298C mRNA at 0.5 or 1.0 mg/kg, respectively). For statistical analysis, two-sample t-test (two-sided) was performed and corrected for multiple testing by using a Bonferroni adjusted level of 0.005. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001, comparing hG6PC S298C mRNA 1.0 mg/kg with eGFP (p values are 0.0017 [day 0, 2.5 h], 0.0016 [day 0, 6 h], 0.0006 [day 2, 2.5 h], 0.0005 [day 2, 6 h], 0.001 [day 4, 2.5 h], 0.001 [day 4, 6 h], and 0.048 [day 7, 6 h], respectively). †P ≤ 0.05, ††P ≤ 0.01, †††P ≤ 0.001, ††††P ≤ 0.0001, comparing hG6PC S298C mRNA 0.5 mg/kg with eGFP (p values are 0.033 [day 0, 2.5 h], 0.001 [day 0, 6 h], 0.0004 [day 2, 2.5 h], 0.00003 [day 2, 6 h], 0.002 [day 4, 2.5 h], 0.001 [day 4, 6 h], and 0.009 [day 7, 6 h], respectively) b Blood glucose levels following repeat (five doses) i.v. administrations of hG6PC S298C mRNA-LNP (0.25 mg/kg) in L.G6pc−/− mice. Arrows indicate dose administration. Blood glucose levels were measured at 2.5-h post-fasting. Data were presented as mean ± SD (n = 8, 7, and 9 mice per group for WT treated with PBS, L.G6pc−/− treated with eGFP, and L.G6pc−/− treated with hG6PC S298C mRNA, respectively). For statistical analysis, two-sample t-test (two-sided) was performed and corrected for multiple testing by using a Bonferroni adjusted level of 0.005. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ***P ≤ 0.0001 comparing hG6PC S298C mRNA with eGFP (p values are 0.005 [day 11], 4 × 10−6 [day 25], 0.0007 [day 28], 0.0192 [day38], 1.3 × 10−5 [day 39], 0.001 [day 42], 2 × 10−6 [day 52], and 5 × 10−5 [day 53], respectively). c Serum proinflammatory cytokines (from left to right): IFNɣ, IL-1β, TNFα, and IL6 from the dose-ranging study. d serum ALT (mU/mL) levels from the dose-ranging study. For c and d, data were presented as mean ± SD (n = 6, 5, 8, 7, and 6 mice per group for WT treated with PBS, L.G6pc−/− treated with eGFP, and L.G6pc−/− treated with hG6PC S298C mRNA at 0.2, 0.5, or 1.0 mg/kg, respectively). e Serum proinflammatory cytokines (from left to right): IFNɣ, IL-1β, TNFα, and IL6 from repeat-dose study. Data were presented as mean ± SD (n = 10, 10, and 7 mice per group for WT treated with PBS, L.G6pc−/− treated with eGFP, or hG6PC S298C mRNA). f Antidrug antibody assay measuring anti-G6Pase-α antibodies in sera of mice treated with five doses of hG6PC S298C mRNA-LNP (0.5 mg/kg). Data were presented as mean ± SD (n = 9, 7, 7, 6 mice per group for WT treated with PBS, L.G6pc−/− treated with eGFP, L.G6pc−/− treated with hG6PC S298C mRNA, and positive sera, respectively). g Body weight of L.G6pc−/− mice prior to each repeat i.v. dose treatment of hG6PC mRNA -LNP (0.25 mg/kg) for repeat dose study. Data were presented as mean ± SD (n = 8, 7, and 9 mice per group for WT treated with PBS, L.G6pc−/− treated with eGFP, and L.G6pc−/− treated with hG6PC S298C mRNA, respectively). For statistical analysis of c–f, raw values were Log2 transformed and subjected to one-way ANOVA, followed by the Dunnett’s multiple comparisons test, compared to the eGFP mRNA treated group. P values are shown in the graphs (c–f). Source data are provided as a Source Data File.
Fig. 6. Effect of h G6PC S298C…
Fig. 6. Effect of hG6PC S298C mRNA-LNP on prevention of hepatic adenomas in L.G6pc−/− mice.
a Number of mice with tumors (left), number of tumors per mouse (middle), and tumor burden/area (right). Data were presented as mean ± s.e.m (n = 21, 26, and 34 mice per group for wild-type (WT) treated with PBS, L.G6pc−/− treated with eGFP, and hG6PC S298C mRNA, respectively). b Liver morphology (tumor-circled in yellow) (top panels) and liver histology (bottom panels) of WT and L.G6pc−/− mice treated with either eGFP mRNA or hG6PC S298C mRNAs. c HCA/HCC biomarkers (protein expression). Results are expressed as mean ± SD (n = 15, 27, and 35 mice per group for WT treated with PBS, L.G6pc−/− treated with eGFP, and L.G6pc−/− treated with hG6PC S298C mRNA, respectively). For statistical analysis, raw values were subjected to one-way ANOVA, followed by the Dunnett’s multiple comparisons test, compared to the eGFP mRNA treated group. Statistically significant P values (p ≤ 0.05) are shown in the graphs. Source data are provided as a Source Data File.

References

    1. Burda P, Hochuli M. Hepatic glycogen storage disorders: what have we learned in recent years? Curr. Opin. Clin. Nutr. 2015;18:415–421. doi: 10.1097/MCO.0000000000000181.
    1. Shin YS. Glycogen storage disease: clinical, biochemical, and molecular heterogeneity. Semin. Pediatr. Neurol. 2006;13:115–120. doi: 10.1016/j.spen.2006.06.007.
    1. Lei KJ, et al. Mutations in the glucose-6-phosphatase gene are associated with glycogen storage disease types 1a and 1aSP but not 1b and 1c. J. Clin. Invest. 1995;95:234–240. doi: 10.1172/JCI117645.
    1. Lei K, Shelly L, Pan C, Sidbury J, Chou J. Mutations in the glucose-6-phosphatase gene that cause glycogen storage disease type 1a. Science. 1993;262:580–583. doi: 10.1126/science.8211187.
    1. Pan C, Lei K, Chen H, Ward J, Chou J. Ontogeny of the murine glucose-6-phosphatase system. Arch. Biochem. Biophys. 1998;358:17–24. doi: 10.1006/abbi.1998.0849.
    1. BRUNI N, et al. Enzymatic characterization of four new mutations in the glucose‐6 phosphatase (G6PC) gene which cause glycogen storage disease type 1a. Ann. Hum. Genet. 1999;63:141–146. doi: 10.1046/j.1469-1809.1999.6320141.x.
    1. Chou J, Jun H, Mansfield B. Type I glycogen storage diseases: disorders of the glucose-6-phosphatase/glucose-6-phosphate transporter complexes. J. Inherit. Metab. Dis. 2015;38:511–519. doi: 10.1007/s10545-014-9772-x.
    1. Rajas F, Gautier-Stein A, Mithieux G. Glucose-6 phosphate, a central hub for liver carbohydrate metabolism. Metabolites. 2019;9:282. doi: 10.3390/metabo9120282.
    1. Shah K, O’Dell S. Effect of dietary interventions in the maintenance of normoglycaemia in glycogen storage disease type 1a: a systematic review and meta-analysis. J. Hum. Nutr. Diet. 2013;26:329–339. doi: 10.1111/jhn.12030.
    1. Correia C, et al. Use of modified cornstarch therapy to extend fasting in glycogen storage disease types Ia and Ib. Am. J. Clin. Nutr. 2008;88:1272–1276.
    1. Greene H, Slonim A, O’Neill J, Burr I. Continuous nocturnal intragastric feeding for management of type 1 glycogen-storage disease. N. Engl. J. Med. 1976;294:423–425. doi: 10.1056/NEJM197602192940805.
    1. Labrador E, Kuo C, Weinstein D. Prevention of complications in glycogen storage disease type Ia with optimization of metabolic control. Pediatr. Diabetes. 2017;18:327–331. doi: 10.1111/pedi.12540.
    1. Wang D, Fiske L, Carreras C, Weinstein D. Natural history of hepatocellular adenoma formation in glycogen storage disease type I. J. Pediatr. 2011;159:442–446. doi: 10.1016/j.jpeds.2011.02.031.
    1. Calderaro J, et al. Molecular characterization of hepatocellular adenomas developed in patients with glycogen storage disease type I. J. Hepatol. 2012;58:350–357. doi: 10.1016/j.jhep.2012.09.030.
    1. Franco L, et al. Hepatocellular carcinoma in glycogen storage disease type Ia: a case series. J. Inherit. Metab. Dis. 2005;28:153–162. doi: 10.1007/s10545-005-7500-2.
    1. Labrune P. Glycogen storage disease type I: indications for liver and/or kidney transplantation. Eur. J. Pediatr. 2002;161:S53–S55. doi: 10.1007/s00431-002-1004-y.
    1. Forbes S, Gupta S, Dhawan A. Cell therapy for liver disease: from liver transplantation to cell factory. J. Hepatol. 2015;62:S157–S169. doi: 10.1016/j.jhep.2015.02.040.
    1. Forbes S, Newsome P. New horizons for stem cell therapy in liver disease. J. Hepatol. 2012;56:496–499. doi: 10.1016/j.jhep.2011.06.022.
    1. Clar J, et al. Hepatic lentiviral gene transfer prevents the long-term onset of hepatic tumours of glycogen storage disease type 1a in mice. Hum. Mol. Genet. 2015;24:2287–2296. doi: 10.1093/hmg/ddu746.
    1. Lee Y, Kim G, Pan C, Mansfield B, Chou J. Minimal hepatic glucose-6-phosphatase-α activity required to sustain survival and prevent hepatocellular adenoma formation in murine glycogen storage disease type Ia. Mol. Genet. Metab. Rep. 2015;3:28–32. doi: 10.1016/j.ymgmr.2015.03.001.
    1. Zingone A, et al. Correction of glycogen storage disease type 1a in a mouse model by gene therapy. J. Biol. Chem. 2000;275:828–832. doi: 10.1074/jbc.275.2.828.
    1. Kim G, et al. Glycogen storage disease type Ia mice with less than 2% of normal hepatic glucose-6-phosphatase-α activity restored are at risk of developing hepatic tumors. Mol. Genet. Metab. 2017;120:229–234. doi: 10.1016/j.ymgme.2017.01.003.
    1. Weinstein D. AAV8-mediated liver-directed gene therapy as a potential therapeutic option in adults with glycogen storage disease type Ia (GSDIa): results from a phase 1/2 clinical trial. Mol. Ther. 2020;28:556.
    1. Hareendran S, et al. Adeno‐associated virus (AAV) vectors in gene therapy: immune challenges and strategies to circumvent them. Rev. Med. Virol. 2013;23:399–413. doi: 10.1002/rmv.1762.
    1. Concolino D, Deodato F, Parini R. Enzyme replacement therapy: efficacy and limitations. Ital. J. Pediatr. 2018;44:120. doi: 10.1186/s13052-018-0562-1.
    1. Trepotec Z, Lichtenegger E, Plank C, Aneja M, Rudolph C. Delivery of mRNA therapeutics for treatment of hepatic diseases. Mol. Ther. 2018;27:794–802. doi: 10.1016/j.ymthe.2018.12.012.
    1. Maruggi G, Zhang C, Li J, Ulmer J, Yu D. mRNA as a transformative technology for vaccine development to control infectious diseases. Mol. Ther. 2019;27:757–772. doi: 10.1016/j.ymthe.2019.01.020.
    1. Weissman D. mRNA transcript therapy. Expert Rev. Vaccines. 2014;14:265–281. doi: 10.1586/14760584.2015.973859.
    1. Martini P, Guey LT. A new era for rare genetic diseases: messenger RNA therapy. Hum. Gene Ther. 2019;30:1180–1189. doi: 10.1089/hum.2019.090.
    1. Berraondo P, Martini PGV, Avila MA, Fontanellas A. Messenger RNA therapy for rare genetic metabolic diseases. Gut. 2019;68:1323. doi: 10.1136/gutjnl-2019-318269.
    1. Bernal J. RNA-based tools for nuclear reprogramming and lineage-conversion: towards clinical applications. J. Cardiovasc. Transl. 2013;6:956–968. doi: 10.1007/s12265-013-9494-8.
    1. Kuhn A, et al. mRNA as a versatile tool for exogenous protein expression. Curr. Gene Ther. 2012;12:347–361. doi: 10.2174/156652312802762536.
    1. Kowalski P, Rudra A, Miao L, Anderson D. Delivering the messenger: advances in technologies for therapeutic mRNA delivery. Mol. Ther. 2019;27:710–728. doi: 10.1016/j.ymthe.2019.02.012.
    1. Sabnis S, et al. A novel amino lipid series for mRNA delivery: improved endosomal escape and sustained pharmacology and safety in non-human primates. Mol. Ther. 2018;26:1509–1519. doi: 10.1016/j.ymthe.2018.03.010.
    1. Vaidyanathan S, et al. Uridine depletion and chemical modification increase Cas9 mRNA activity and reduce immunogenicity without HPLC purification. Mol. Ther. Nucleic Acids. 2018;12:530–542. doi: 10.1016/j.omtn.2018.06.010.
    1. Badieyan ZS, Evans T. Concise review: application of chemically modified mRNA in cell fate conversion and tissue engineering. Stem Cell Transl. Med. 2019;8:833–843.
    1. Nelson J, et al. Impact of mRNA chemistry and manufacturing process on innate immune activation. Sci. Adv. 2020;6:eaaz6893. doi: 10.1126/sciadv.aaz6893.
    1. An D, et al. Systemic messenger RNA therapy as a treatment for methylmalonic acidemia. Cell Rep. 2017;21:3548–3558. doi: 10.1016/j.celrep.2017.11.081.
    1. An D, et al. Long-term efficacy and safety of mRNA therapy in two murine models of methylmalonic acidemia. EBioMedicine. 2019;45:519–528. doi: 10.1016/j.ebiom.2019.07.003.
    1. Jiang L, et al. Systemic messenger RNA as an etiological treatment for acute intermittent porphyria. Nat. Med. 2018;24:1899–1909. doi: 10.1038/s41591-018-0199-z.
    1. Parra‐Guillen Z, et al. Disease pharmacokinetic–pharmacodynamic modelling in acute intermittent porphyria to support the development of mRNA‐based therapies. Brit. J. Pharm. 2020;177:3168–3182. doi: 10.1111/bph.15040.
    1. Zhu X, et al. Systemic mRNA therapy for the treatment of Fabry disease: preclinical studies in wild-type mice, Fabry mouse model, and wild-type non-human primates. Am. J. Hum. Genet. 2019;104:625–637. doi: 10.1016/j.ajhg.2019.02.003.
    1. Truong B, et al. Lipid nanoparticle-targeted mRNA therapy as a treatment for the inherited metabolic liver disorder arginase deficiency. Proc. Natl Acad. Sci. USA. 2019;116:21150–21159. doi: 10.1073/pnas.1906182116.
    1. Cao J, et al. mRNA therapy improves metabolic and behavioral abnormalities in a murine model of citrin deficiency. Mol. Ther. 2019;27:1242–1251. doi: 10.1016/j.ymthe.2019.04.017.
    1. Karadagi A, et al. Systemic modified messenger RNA for replacement therapy in alpha 1-antitrypsin deficiency. Sci. Rep. 2020;10:7052. doi: 10.1038/s41598-020-64017-0.
    1. Roseman D, et al. G6PC mRNA therapy positively regulates fasting blood glucose and decreases hepatic abnormalities in a mouse model of glycogen storage disease 1a. Mol. Ther. 2018;26:814–821. doi: 10.1016/j.ymthe.2018.01.006.
    1. Zhang L, Cho J, Arnaoutova I, Mansfield BC, Chou JY. An evolutionary approach to optimizing glucose‐6‐phosphatase‐α enzymatic activity for gene therapy of glycogen storage disease type Ia. J. Inherit. Metab. Dis. 2019;42:470–479. doi: 10.1002/jimd.12069.
    1. Zhang L, et al. Gene therapy using a novel G6PC-S298C variant enhances the long-term efficacy for treating glycogen storage disease type Ia. Biochem. Biophys. Res. Commun. 2020;527:824–830. doi: 10.1016/j.bbrc.2020.04.124.
    1. Pan C, Lei K, Annabi B, Hemrika W, Chou J. Transmembrane topology of glucose-6-phosphatase. J. Biol. Chem. 1998;273:6144–6148. doi: 10.1074/jbc.273.11.6144.
    1. Ghosh A, Shieh J, Pan C, Sun M, Chou J. The catalytic center of glucose-6-phosphatase HIS176 is the nucleophile forming the phosphohistidine-enzyme intermediate during catalysis. J. Biol. Chem. 2002;277:32837–32842. doi: 10.1074/jbc.M201853200.
    1. Cao J, et al. mRNA therapy improves metabolic and behavioral abnormalities in a murine model of citrin deficiency. Mol. Ther. 2019;27:1242–1251. doi: 10.1016/j.ymthe.2019.04.017.
    1. Mutel E, et al. Control of blood glucose in the absence of hepatic glucose production during prolonged fasting in mice. Diabetes. 2011;60:3121–3131. doi: 10.2337/db11-0571.
    1. Mutel E, et al. Targeted deletion of liver glucose-6 phosphatase mimics glycogen storage disease type 1a including development of multiple adenomas. J. Hepatol. 2011;54:529–537. doi: 10.1016/j.jhep.2010.08.014.
    1. Gjorgjieva M, et al. Dietary exacerbation of metabolic stress leads to accelerated hepatic carcinogenesis in glycogen storage disease type Ia. J. Hepatol. 2018;69:1074–1087. doi: 10.1016/j.jhep.2018.07.017.
    1. Callies S, et al. Integrated analysis of preclinical data to support the design of the first in man study of LY2181308, a second generation antisense oligonucleotide: preclinical to clinical PK/PD model and analysis of LY2181308. Brit. J. Clin. Pharm. 2011;71:416–428. doi: 10.1111/j.1365-2125.2010.03836.x.
    1. Nair A, Morsy MA, Jacob S. Dose translation between laboratory animals and human in preclinical and clinical phases of drug development. Drug Dev. Res. 2018;79:373–382. doi: 10.1002/ddr.21461.
    1. Mahmood I. Dosing in children: a critical review of the pharmacokinetic allometric scaling and modelling approaches in paediatric drug development and clinical settings. Clin. Pharmacokinet. 2014;53:327–346. doi: 10.1007/s40262-014-0134-5.
    1. Jensen T, Kiersgaard M, Sørensen D, Mikkelsen L. Fasting of mice: a review. Lab. Anim. 2013;47:225–240. doi: 10.1177/0023677213501659.
    1. Khan S. Genome editing technologies: concept, pros and cons of various genome editing techniques and bioethical concerns for clinical application. Mol. Ther. Nucleic Acids. 2019;16:326–334. doi: 10.1016/j.omtn.2019.02.027.
    1. Rivera-Torres N, Banas K, Bialk P, Bloh K, Kmiec E. Insertional mutagenesis by CRISPR/Cas9 ribonucleoprotein gene editing in cells targeted for point mutation repair directed by short single-stranded DNA oligonucleotides. PloS ONE. 2017;12:e0169350. doi: 10.1371/journal.pone.0169350.
    1. Hacein-Bey-Abina S, et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N. Engl. J. Med. 2003;348:255–256. doi: 10.1056/NEJM200301163480314.
    1. Dunbar C, et al. Gene therapy comes of age. Science. 2018;359:eaan4672. doi: 10.1126/science.aan4672.
    1. Anguela X, High K. Entering the modern era of gene therapy. Annu. Rev. Med. 2017;70:1–16.
    1. Ma C, Wang Z, Xu T, He Z, Wei Y. The approved gene therapy drugs worldwide: from 1998 to 2019. Biotechnol. Adv. 2019;40:107502. doi: 10.1016/j.biotechadv.2019.107502.
    1. Weinstein D, et al. Adeno-associated virus-mediated correction of a canine model of glycogen storage disease type Ia. Hum. Gene Ther. 2010;21:903–910. doi: 10.1089/hum.2009.157.
    1. Jauze L, Monteillet L, Mithieux G, Rajas F, Ronzitti G. Challenges of gene therapy for the treatment of glycogen storage diseases type I and type III. Hum. Gene Ther. 2019;30:1263–1273. doi: 10.1089/hum.2019.102.
    1. Calcedo R, Vandenberghe L, Gao G, Lin J, Wilson J. Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. J. Infect. Dis. 2009;199:381–390. doi: 10.1086/595830.
    1. Lee Y, 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. doi: 10.1002/hep.25717.
    1. Cho J, Lee YM, Starost MF, Mansfield BC, Chou JY. Gene therapy prevents hepatic tumor initiation in murine glycogen storage disease type Ia at the tumor‐developing stage. J. Inherit. Metab. Dis. 2019;42:459–469. doi: 10.1002/jimd.12056.
    1. Sabnis S, et al. A novel amino lipid series for mRNA delivery: improved endosomal escape and sustained pharmacology and safety in non-human primates. Mol. Ther. 2018;26:1509–1519. doi: 10.1016/j.ymthe.2018.03.010.
    1. Taussky H. A microcolorimetric method for the determination of inorganic phosphorus. J. Biol. Chem. 1953;202:675–685. doi: 10.1016/S0021-9258(18)66180-0.

Source: PubMed

3
Tilaa