Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-28T14:55:21.965Z Has data issue: false hasContentIssue false

GLP-1-mediated gene therapy approaches for diabetes treatment

Published online by Cambridge University Press:  26 March 2014

Mukerrem Hale Tasyurek
Affiliation:
Human Gene and Cell Therapy Center, Akdeniz University Hospitals, Antalya 07058, Turkey Department of Medical Biology and Genetics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
Hasan Ali Altunbas
Affiliation:
Human Gene and Cell Therapy Center, Akdeniz University Hospitals, Antalya 07058, Turkey Department of Internal Medicine, Division of Endocrinology and Metabolism, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
Halit Canatan
Affiliation:
Genome and Stem Cell Research Center, Department of Medical Biology, Erciyes University Faculty of Medicine, Melikgazi, Kayseri 38039, Turkey
Thomas S. Griffith
Affiliation:
Department of Urology, University of Minnesota, Minneapolis, MN 55455, USA
Salih Sanlioglu*
Affiliation:
Human Gene and Cell Therapy Center, Akdeniz University Hospitals, Antalya 07058, Turkey Department of Medical Biology and Genetics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
*
*Corresponding author: Professor Dr. Salih Sanlioglu VMD, PhD, Human Gene and Cell Therapy Center, Akdeniz University Hospitals and Clinics, B Block, 1st floor, Campus, Antalya 07058, Turkey. E-mail: sanlioglu@akdeniz.edu.tr

Abstract

Glucagon-like peptide (GLP)-1 is an incretin hormone with several antidiabetic functions including stimulation of glucose-dependent insulin secretion, increase in insulin gene expression and beta-cell survival. Despite the initial technical difficulties and profound inefficiency of direct gene transfer into the pancreas that seriously restricted in vivo gene transfer experiments with GLP-1, recent exploitation of various routes of gene delivery and alternative means of gene transfer has permitted the detailed assessment of the therapeutic efficacy of GLP-1 in animal models of type 2 diabetes (T2DM). As a result, many clinical benefits of GLP-1 peptide/analogues observed in clinical trials involving induction of glucose tolerance, reduction of hyperglycaemia, suppression of appetite and food intake linked to weight loss have been replicated in animal models using gene therapy. Furthermore, GLP-1-centered gene therapy not only improved insulin sensitivity, but also reduced abdominal and/or hepatic fat associated with obesity-induced T2DM with drastic alterations in adipokine profiles in treated subjects. Thus, a comprehensive assessment of recent GLP-1-mediated gene therapy approaches with detailed analysis of current hurdles and resolutions, is discussed.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2014 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Vilsboll, T. and Holst, J.J. (2004) Incretins, insulin secretion and type 2 diabetes mellitus. Diabetologia 47, 357-366CrossRefGoogle ScholarPubMed
2Russell, S. (2013) Incretin-based therapies for type 2 diabetes mellitus: a review of direct comparisons of efficacy, safety and patient satisfaction. International Journal of Clinical Pharmacy 35, 159-172CrossRefGoogle ScholarPubMed
3Kieffer, T.J. and Habener, J.F. (1999) The glucagon-like peptides. Endocrine Reviews 20, 876-913CrossRefGoogle ScholarPubMed
4Roberge, J.N. and Brubaker, P.L. (1993) Regulation of intestinal proglucagon-derived peptide secretion by glucose-dependent insulinotropic peptide in a novel enteroendocrine loop. Endocrinology 133, 233-240CrossRefGoogle Scholar
5Sanlioglu, A.D. et al. (2012) Therapeutic potential of VIP vs PACAP in diabetes. Journal of Molecular Endocrinology 49, R157-R167CrossRefGoogle ScholarPubMed
6Drucker, D.J. et al. (1987) Glucagon-like peptide I stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell line. Proceedings of the National Academy of Sciences of the United States of America 84, 3434-3438CrossRefGoogle Scholar
7Kreymann, B. et al. (1987) Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet 2, 1300-1304CrossRefGoogle ScholarPubMed
8Fehmann, H.C. and Habener, J.F. (1992) Insulinotropic hormone glucagon-like peptide-I(7-37) stimulation of proinsulin gene expression and proinsulin biosynthesis in insulinoma beta TC-1 cells. Endocrinology 130, 159-166CrossRefGoogle ScholarPubMed
9Thorens, B. et al. (1993) Cloning and functional expression of the human islet GLP-1 receptor. Demonstration that exendin-4 is an agonist and exendin-(9-39) an antagonist of the receptor. Diabetes 42, 1678-1682CrossRefGoogle ScholarPubMed
10Gribble, F.M. (2012) The gut endocrine system as a coordinator of postprandial nutrient homoeostasis. Proceedings of the Nutrition Society 71, 456-462CrossRefGoogle ScholarPubMed
11Tolhurst, G., Reimann, F. and Gribble, F.M. (2012) Intestinal sensing of nutrients. Handbook of Experimental Pharmacology 209, 309-335.CrossRefGoogle Scholar
12Ritzel, R. et al. (1995) Pharmacokinetic, insulinotropic, and glucagonostatic properties of GLP-1 [7-36 amide] after subcutaneous injection in healthy volunteers. Dose-response-relationships. Diabetologia 38, 720-725CrossRefGoogle ScholarPubMed
13Wang, Y. et al. (1997) Glucagon-like peptide-1 can reverse the age-related decline in glucose tolerance in rats. Journal of Clinical Investigation 99, 2883-2889CrossRefGoogle ScholarPubMed
14Abraham, E.J. et al. (2002) Insulinotropic hormone glucagon-like peptide-1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells. Endocrinology 143, 3152-3161CrossRefGoogle ScholarPubMed
15Tourrel, C. et al. (2002) Persistent improvement of type 2 diabetes in the Goto-Kakizaki rat model by expansion of the beta-cell mass during the prediabetic period with glucagon-like peptide-1 or exendin-4. Diabetes 51, 1443-1452CrossRefGoogle ScholarPubMed
16Turton, M.D. et al. (1996) A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379, 69-72CrossRefGoogle ScholarPubMed
17Wettergren, A. et al. (1993) Truncated GLP-1 (proglucagon 78-107-amide) inhibits gastric and pancreatic functions in man. Digestive Diseases and Sciences 38, 665-673CrossRefGoogle ScholarPubMed
18Zhao, T.C. (2013) Glucagon-like peptide-1 (GLP-1) and protective effects in cardiovascular disease: a new therapeutic approach for myocardial protection. Cardiovascular Diabetology 12, 90CrossRefGoogle ScholarPubMed
19Knop, F.K. et al. (2007) Reduced incretin effect in type 2 diabetes: cause or consequence of the diabetic state? Diabetes 56, 1951-1959CrossRefGoogle ScholarPubMed
20Toft-Nielsen, M.B. et al. (2001) Determinants of the impaired secretion of glucagon-like peptide-1 in type 2 diabetic patients. Journal of Clinical Endocrinology and Metabolism 86, 3717-3723CrossRefGoogle ScholarPubMed
21Nauck, M.A. et al. (1993) Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. Journal of Clinical Investment 91, 301-307CrossRefGoogle Scholar
22Kjems, L.L. et al. (2003) The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes 52, 380-386CrossRefGoogle ScholarPubMed
23Bell, G.I., Santerre, R.F. and Mullenbach, G.T. (1983) Hamster preproglucagon contains the sequence of glucagon and two related peptides. Nature 302, 716-718CrossRefGoogle ScholarPubMed
24Novak, U. et al. (1987) Identical mRNA for preproglucagon in pancreas and gut. European Journal of Biochemistry 164, 553-558CrossRefGoogle ScholarPubMed
25Holst, J.J. et al. (1994) Proglucagon processing in porcine and human pancreas. Journal of Biological Chemistry 269, 18827-18833CrossRefGoogle ScholarPubMed
26Ugleholdt, R. et al. (2004) Impaired intestinal proglucagon processing in mice lacking prohormone convertase 1. Endocrinology 145, 1349-1355CrossRefGoogle ScholarPubMed
27Alhadeff, A.L., Rupprecht, L.E. and Hayes, M.R. (2012) GLP-1 neurons in the nucleus of the solitary tract project directly to the ventral tegmental area and nucleus accumbens to control for food intake. Endocrinology 153, 647-658CrossRefGoogle Scholar
28Skibicka, K.P. (2013) The central GLP-1: implications for food and drug reward. Frontiers in Neuroscience 7, 181CrossRefGoogle ScholarPubMed
29Zhao, S. et al. (2012) Hindbrain leptin and glucagon-like-peptide-1 receptor signaling interact to suppress food intake in an additive manner. International Journal of Obesity (London) 36, 1522-1528CrossRefGoogle Scholar
30Hayes, M.R., Bradley, L. and Grill, H.J. (2009) Endogenous hindbrain glucagon-like peptide-1 receptor activation contributes to the control of food intake by mediating gastric satiation signaling. Endocrinology 150, 2654-2659CrossRefGoogle Scholar
31Huo, L. et al. (2007) Leptin and the control of food intake: neurons in the nucleus of the solitary tract are activated by both gastric distension and leptin. Endocrinology 148, 2189-2197CrossRefGoogle ScholarPubMed
32Kim, W. and Egan, J.M. (2008) The role of incretins in glucose homeostasis and diabetes treatment. Pharmacological Reviews 60, 470-512CrossRefGoogle ScholarPubMed
33Vilsboll, T. et al. (2001) No reactive hypoglycaemia in Type 2 diabetic patients after subcutaneous administration of GLP-1 and intravenous glucose. Diabetic Medicine 18, 144-149CrossRefGoogle ScholarPubMed
34Rowzee, A.M. et al. (2011) Glucagon-like peptide-1 gene therapy. Experimental Diabetes Research 2011, 601047CrossRefGoogle ScholarPubMed
35Oh, S. et al. (2003) GLP-1 gene delivery for the treatment of type 2 diabetes. Molecular Therapy 7, 478-483CrossRefGoogle ScholarPubMed
36Choi, S. et al. (2005) Glucagon-like peptide-1 plasmid construction and delivery for the treatment of type 2 diabetes. Molecular Therapy 12, 885-891CrossRefGoogle ScholarPubMed
37Kumar, M. et al. (2007) Gene therapy of diabetes using a novel GLP-1/IgG1-Fc fusion construct normalizes glucose levels in db/db mice. Gene Therapy 14, 162-172CrossRefGoogle ScholarPubMed
38Dash, P.R. et al. (1999) Factors affecting blood clearance and in vivo distribution of polyelectrolyte complexes for gene delivery. Gene Therapy 6, 643-650CrossRefGoogle ScholarPubMed
39Thibault, M. et al. (2010) Intracellular trafficking and decondensation kinetics of chitosan-pDNA polyplexes. Molucular Therapy 18, 1787-1795CrossRefGoogle ScholarPubMed
40Jean, M. et al. (2011) Effective and safe gene-based delivery of GLP-1 using chitosan/plasmid-DNA therapeutic nanocomplexes in an animal model of type 2 diabetes. Gene Therapy 18, 807-816CrossRefGoogle Scholar
41Jean, M. et al. (2012) Chitosan-based therapeutic nanoparticles for combination gene therapy and gene silencing of in vitro cell lines relevant to type 2 diabetes. European Journal of Pharmaceutical Sciences 45, 138-149CrossRefGoogle ScholarPubMed
42Kim, P.H., Lee, M. and Kim, S.W. (2012) Delivery of two-step transcription amplification exendin-4 plasmid system with arginine-grafted bioreducible polymer in type 2 diabetes animal model. Journal of Controlled Release 162, 9-18CrossRefGoogle ScholarPubMed
43Parsons, G.B. et al. (2007) Ectopic expression of glucagon-like peptide 1 for gene therapy of type II diabetes. Gene Therapy 14, 38-48CrossRefGoogle ScholarPubMed
44Voutetakis, A. et al. (2010) Systemic delivery of bioactive glucagon-like peptide 1 after adenoviral-mediated gene transfer in the murine salivary gland. Endocrinology 151, 4566-4572CrossRefGoogle ScholarPubMed
45Liu, M.J. et al. (2007) Prolonged remission of diabetes by regeneration of beta cells in diabetic mice treated with recombinant adenoviral vector expressing glucagon-like peptide-1. Molecular Therapy 15, 86-93CrossRefGoogle ScholarPubMed
46Lee, Y. et al. (2008) Adenoviral vector-mediated glucagon-like peptide 1 gene therapy improves glucose homeostasis in Zucker diabetic fatty rats. Journal of Gene Medicine 10, 260-268CrossRefGoogle ScholarPubMed
47Bastard, J.P. et al. (2006) Recent advances in the relationship between obesity, inflammation, and insulin resistance. European Cytokine Network 17, 4-12Google ScholarPubMed
48Lee, Y.S. et al. (2012) Glucagon-like peptide-1 inhibits adipose tissue macrophage infiltration and inflammation in an obese mouse model of diabetes. Diabetologia 55, 2456-2468CrossRefGoogle Scholar
49Usui, C. et al. (2010) Visceral fat is a strong predictor of insulin resistance regardless of cardiorespiratory fitness in non-diabetic people. Journal of Nutritional Science and Vitaminology (Tokyo) 56, 109-116CrossRefGoogle Scholar
50Samson, S.L. et al. (2008) Gene therapy for diabetes: metabolic effects of helper-dependent adenoviral exendin 4 expression in a diet-induced obesity mouse model. Molecular Therapy 16, 1805-1812CrossRefGoogle Scholar
51Wang, Z. et al. (2006) Widespread and stable pancreatic gene transfer by adeno-associated virus vectors via different routes. Diabetes 55, 875-884CrossRefGoogle ScholarPubMed
52Riedel, M.J. et al. (2010) DsAAV8-mediated expression of glucagon-like peptide-1 in pancreatic beta-cells ameliorates streptozotocin-induced diabetes. Gene Therapy 17, 171-180CrossRefGoogle ScholarPubMed
53Gaddy, D.F. et al. (2012) dsAAV8-mediated gene transfer and beta-cell expression of IL-4 and beta-cell growth factors are capable of reversing early-onset diabetes in NOD mice. Gene Therapy 19, 791-799CrossRefGoogle ScholarPubMed
54Lopez-Talavera, J.C. et al. (2004) Hepatocyte growth factor gene therapy for pancreatic islets in diabetes: reducing the minimal islet transplant mass required in a glucocorticoid-free rat model of allogeneic portal vein islet transplantation. Endocrinology 145, 467-474CrossRefGoogle Scholar
55Gaddy, D.F. et al. (2010) In vivo expression of HGF/NK1 and GLP-1 From dsAAV vectors enhances pancreatic ss-cell proliferation and improves pathology in the db/db mouse model of diabetes. Diabetes 59, 3108-3116CrossRefGoogle ScholarPubMed
56Choi, S.H. and Lee, H.C. (2011) Long-term, antidiabetogenic effects of GLP-1 gene therapy using a double-stranded, adeno-associated viral vector. Gene Therapy 18, 155-163CrossRefGoogle ScholarPubMed
57Voutetakis, A. et al. (2005) Salivary glands as a potential gene transfer target for gene therapeutics of some monogenetic endocrine disorders. Journal of Endocrinology 185, 363-372CrossRefGoogle ScholarPubMed
58Katano, H. et al. (2006) Enhanced transduction of mouse salivary glands with AAV5-based vectors. Gene Therapy 13, 594-601CrossRefGoogle ScholarPubMed
59Di Pasquale, G. et al. (2012) Sustained Exendin-4 Secretion through Gene Therapy Targeting Salivary Glands in Two Different Rodent Models of Obesity/Type 2 Diabetes. PLoS ONE 7, e40074CrossRefGoogle ScholarPubMed
60Sanlioglu, A.D. et al. (2012) Insulin gene therapy from design to beta cell generation. Expert Reviews in Molecular Medicine 14, e18CrossRefGoogle ScholarPubMed
61Doerschug, K. et al. (2002) First-generation adenovirus vectors shorten survival time in a murine model of sepsis. Journal of Immunology 169, 6539-6545CrossRefGoogle Scholar
62Raper, S.E. et al. (2003) Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Molecular Genetics and Metabolism 80, 148-158CrossRefGoogle Scholar
63Lowenstein, P.R. et al. (2002) High-capacity, helper-dependent, “gutless” adenoviral vectors for gene transfer into brain. Methods Enzymol 346, 292-311CrossRefGoogle ScholarPubMed
64Vetrini, F. and Ng, P. (2010) Gene therapy with helper-dependent adenoviral vectors: current advances and future perspectives. Viruses 2, 1886-1917CrossRefGoogle ScholarPubMed
65Muhammad, A.K. et al. (2012) Safety profile of gutless adenovirus vectors delivered into the normal brain parenchyma: implications for a glioma phase 1 clinical trial. Human Gene Therapy Methods 23, 271-284CrossRefGoogle ScholarPubMed
66Vetrini, F. and Ng, P. (2011) Liver-directed gene therapy with helper-dependent adenoviral vectors: current state of the art and future challenges. Current Pharmaceutical Design 17, 2488-2499CrossRefGoogle ScholarPubMed
67Sanlioglu, S. et al. (2001) Rate limiting steps of AAV transduction and implications for human gene therapy. Current Gene Therapy 1, 137-147CrossRefGoogle ScholarPubMed
68Sanlioglu, S., Duan, D. and Engelhardt, J.F. (1999) Two independent molecular pathways for recombinant adeno-associated virus genome conversion occur after UV-C and E4orf6 augmentation of transduction. Human Gene Therapy 10, 591-602CrossRefGoogle ScholarPubMed
69Wang, Z. et al. (2003) Rapid and highly efficient transduction by double-stranded adeno-associated virus vectors in vitro and in vivo. Gene Therapy 10, 2105-2111CrossRefGoogle ScholarPubMed
70Wang, A.Y. et al. (2004) Comparison of adenoviral and adeno-associated viral vectors for pancreatic gene delivery in vivo. Human Gene Therapy 15, 405-413CrossRefGoogle ScholarPubMed
71Sanlioglu, S. and Engelhardt, J.F. (1999) Cellular redox state alters recombinant adeno-associated virus transduction through tyrosine phosphatase pathways. Gene Therapy 6, 1427-1437CrossRefGoogle ScholarPubMed
72Sanlioglu, A.D. et al. (2004) Novel approaches to augment adeno-associated virus type-2 endocytosis and transduction. Virus Research 104, 51-59CrossRefGoogle ScholarPubMed
73Sanlioglu, S. et al. (2000) Endocytosis and nuclear trafficking of adeno-associated virus type 2 are controlled by rac1 and phosphatidylinositol-3 kinase activation. Journal of Virology 74, 9184-9196CrossRefGoogle ScholarPubMed
74Sanlioglu, S., Benson, P. and Engelhardt, J.F. (2000) Loss of ATM function enhances recombinant adeno-associated virus transduction and integration through pathways similar to UV irradiation. Virology 268, 68-78CrossRefGoogle ScholarPubMed
75Mukai, E. et al. (2007) Efficient gene transfer into murine pancreatic islets using adenovirus vectors. Journal of Controlled Release 119, 136-141CrossRefGoogle ScholarPubMed
76Rosas, L.E. et al. (2012) Patterns of scAAV vector insertion associated with oncogenic events in a mouse model for genotoxicity. Molecular Therapy 20, 2098-2110CrossRefGoogle Scholar
77Russell, D.W. (2007) AAV vectors, insertional mutagenesis, and cancer. Molecular Therapy 15, 1740-1743CrossRefGoogle ScholarPubMed
78Yap, I.S. et al. (2003) Lack of islet neogenesis plays a key role in beta-cell depletion in mice infected with a diabetogenic variant of coxsackievirus B4. Journal of General Virology 84, 3051-3068CrossRefGoogle Scholar
79Dan, M. and Chantler, J.K. (2005) A genetically engineered attenuated coxsackievirus B3 strain protects mice against lethal infection. Journal of Virology 79, 9285-9295CrossRefGoogle ScholarPubMed
80Dan, M. and Chantler, J.K. (2011) A novel pancreatropic coxsackievirus vector expressing glucagon-like peptide 1 reduces hyperglycemia in streptozotocin-treated mice. Journal of Virology 85, 12759-12768CrossRefGoogle ScholarPubMed
81Di Nunzio, F. et al. (2012) HIV-derived vectors for therapy and vaccination against HIV. Vaccine 30, 2499-2509CrossRefGoogle Scholar
82Durand, S. and Cimarelli, A. (2011) The inside out of lentiviral vectors. Viruses 3, 132-159CrossRefGoogle ScholarPubMed
83Naldini, L. and Verma, I.M. (2000) Lentiviral vectors. Advances in Virus Research 55, 599-609CrossRefGoogle ScholarPubMed
84Copreni, E. et al. (2010) A VSV-G pseudotyped last generation lentiviral vector mediates high level and persistent gene transfer in models of airway epithelium in vitro and in vivo. Viruses 2, 1577-1588CrossRefGoogle ScholarPubMed
85Riedel, M.J., Lee, C.W. and Kieffer, T.J. (2009) Engineered glucagon-like peptide-1-producing hepatocytes lower plasma glucose levels in mice. American Journal of Physiology, Endocrinology and Metabolism 296, E936-E944CrossRefGoogle ScholarPubMed
86McCormack, M.P. and Rabbitts, T.H. (2004) Activation of the T-cell oncogene LMO2 after gene therapy for X-linked severe combined immunodeficiency. New England Journal of Medicine 350, 913-922CrossRefGoogle ScholarPubMed
87Kotin, R.M., Linden, R.M. and Berns, K.I. (1992) Characterization of a preferred site on human chromosome 19q for integration of adeno-associated virus DNA by non-homologous recombination. EMBO Journal 11, 5071-5078CrossRefGoogle ScholarPubMed
88Xu, Z.X. et al. (2009) A 16-bp RBE element mediated Rep-dependent site-specific integration in AAVS1 transgenic mice for expression of hFIX. Gene Therapy 16, 589-595CrossRefGoogle ScholarPubMed
89Liu, R. et al. (2010) A site-specific genomic integration strategy for sustained expression of glucagon-like peptide-1 in mouse muscle for controlling energy homeostasis. Biochemical and Biophysical Research Communication 403, 172-177CrossRefGoogle ScholarPubMed
90Sanlioglu, A.D. et al. (2008) Molecular mechanisms of death ligand-mediated immune modulation: a gene therapy model to prolong islet survival in type 1 diabetes. Journal of Cellular Biochemistry 104, 710-720CrossRefGoogle ScholarPubMed
91Dirice, E. et al. (2009) Adenovirus-mediated TRAIL gene (Ad5hTRAIL) delivery into pancreatic islets prolongs normoglycemia in streptozotocin-induced diabetic rats. Human Gene Therapy 20, 1177-1189CrossRefGoogle ScholarPubMed
92Mahato, R.I. et al. (2003) Cationic lipid and polymer-based gene delivery to human pancreatic islets. Molecular Therapy 7, 89-100CrossRefGoogle ScholarPubMed
93Leibowitz, G. et al. (1999) Gene transfer to human pancreatic endocrine cells using viral vectors. Diabetes 48, 745-753CrossRefGoogle ScholarPubMed
94Raper, S.E. and DeMatteo, R.P. (1996) Adenovirus-mediated in vivo gene transfer and expression in normal rat pancreas. Pancreas 12, 401-410CrossRefGoogle ScholarPubMed
95Taniguchi, H. et al. (2003) beta-cell neogenesis induced by adenovirus-mediated gene delivery of transcription factor pdx-1 into mouse pancreas. Gene Therapy 10, 15-23CrossRefGoogle ScholarPubMed
96Ayuso, E. et al. (2004) In vivo gene transfer to pancreatic beta cells by systemic delivery of adenoviral vectors. Human Gene Therapy 15, 805-812CrossRefGoogle ScholarPubMed
97Gould, G.W. and Holman, G.D. (1993) The glucose transporter family: structure, function and tissue-specific expression. Biochemical Journal 295, 329-341CrossRefGoogle ScholarPubMed
98Shapiro, A.M. et al. (2005) Strategic opportunities in clinical islet transplantation. Transplantation 79, 1304-1307CrossRefGoogle ScholarPubMed
99Auricchio, A. et al. (2002) Constitutive and regulated expression of processed insulin following in vivo hepatic gene transfer. Gene Therapy 9, 963-971CrossRefGoogle ScholarPubMed
100Barash, S., Wang, W. and Shi, Y. (2002) Human secretory signal peptide description by hidden Markov model and generation of a strong artificial signal peptide for secreted protein expression. Biochemical and Biophysical Research Communication 294, 835-842CrossRefGoogle Scholar
101Burgess, T.L. and Kelly, R.B. (1987) Constitutive and regulated secretion of proteins. Annual Review of Cell Biology 3, 243-293CrossRefGoogle ScholarPubMed
102Sanlioglu, A.D. et al. (2013) Clinical utility of insulin and insulin analogs. Islets 5, 67-78CrossRefGoogle ScholarPubMed
103Pollock, R. and Clackson, T. (2002) Dimerizer-regulated gene expression. Current Opinion in Biotechnology 13, 459-467CrossRefGoogle ScholarPubMed
104Tonne, J.M. et al. (2013) Global gene expression profiling of pancreatic islets in mice during streptozotocin-induced beta-cell damage and pancreatic Glp-1 gene therapy. Disease Models and Mechanisms 6, 1236-1245Google ScholarPubMed
105Rosenberg, L. et al. (2004) A pentadecapeptide fragment of islet neogenesis-associated protein increases beta-cell mass and reverses diabetes in C57BL/6J mice. Annals of Surgery 240, 875-884CrossRefGoogle ScholarPubMed
106Lee, Y.S. et al. (2007) Glucagon-like peptide-1 gene therapy in obese diabetic mice results in long-term cure of diabetes by improving insulin sensitivity and reducing hepatic gluconeogenesis. Diabetes 56, 1671-1679CrossRefGoogle ScholarPubMed
107Williams, D.L., Baskin, D.G. and Schwartz, M.W. (2006) Leptin regulation of the anorexic response to glucagon-like peptide-1 receptor stimulation. Diabetes 55, 3387-3393CrossRefGoogle ScholarPubMed
108Wirth, T., Parker, N. and Yla-Herttuala, S. (2013) History of gene therapy. Gene 525, 162-169CrossRefGoogle ScholarPubMed
109Salmon, F., Grosios, K. and Petry, H. (2014) Safety profile of recombinant adeno-associated viral vectors: focus on alipogene tiparvovec (Glybera(R)). Expert Review of Clinical Pharmacology 7, 53-65CrossRefGoogle Scholar
110Yla-Herttuala, S. (2012) Endgame: glybera finally recommended for approval as the first gene therapy drug in the European Union. Molecular Therapy 20, 1831-1832CrossRefGoogle ScholarPubMed
111Garber, A.J. (2011) Novel incretin-based agents and practical regimens to meet needs and treatment goals of patients with type 2 diabetes mellitus. Journal of American Osteopathic Association 111, S20-S30Google ScholarPubMed
112Derosa, G. and Maffioli, P. (2012) GLP-1 agonists exenatide and liraglutide: a review about their safety and efficacy. Current Clinical Pharmacology 7, 214-228CrossRefGoogle ScholarPubMed
113Edwards, K.L. et al. (2012) An update in incretin-based therapy: a focus on glucagon-like peptide-1 receptor agonists. Diabetes Technology and Therapeutics 14, 951-967CrossRefGoogle ScholarPubMed
114Ryan, G.J., Moniri, N.H. and Smiley, D.D. (2013) Clinical effects of once-weekly exenatide for the treatment of type 2 diabetes mellitus. American Journal of Health-System Pharmacy 70, 1123-1131CrossRefGoogle ScholarPubMed
115Macconell, L. et al. (2012) Safety and tolerability of exenatide twice daily in patients with type 2 diabetes: integrated analysis of 5594 patients from 19 placebo-controlled and comparator-controlled clinical trials. Diabetes, Metabolic Syndrome and Obesity 5, 29-41Google Scholar
116Tasyurek, H.M. et al. (2014) Incretins: their physiology and application in the treatment of diabetes mellitus. Diabetes/Metabolism Research and Reviews doi:i10.1002/dmrr.2501CrossRefGoogle ScholarPubMed

Further reading, resources and contacts

Bryant, L.M. et al. (2013) Lessons learned from the clinical development and market authorization of Glybera. Human Gene Therapy Clinical Development 24, 55-64CrossRefGoogle ScholarPubMed
Tibaldi, J.M. (2014) Incorporating incretin-based therapies into clinical practice for patients with type 2 diabetes. Advances in Therapy doi:10.1007/s12325-014-0100-5CrossRefGoogle ScholarPubMed