Skip to main content
Top
Published in: Diabetes Therapy 8/2020

Open Access 01-08-2020 | Insulins | Review

Treatment Options for MODY Patients: A Systematic Review of Literature

Authors: Maurizio Delvecchio, Carmela Pastore, Paola Giordano

Published in: Diabetes Therapy | Issue 8/2020

Login to get access

Abstract

Maturity-onset diabetes of the young (MODY) is an unusual form of diabetes with specific features that distinguish it from type 1 and type 2 diabetes. There are 14 known subtypes of MODY, and mutations in three genes (HNF1A, HNF4A, GCK) account for about 95% of all MODY cases. Diagnosis usually occurs before the age of 25 years, although less frequent forms may occur more often—but not necessarily—later in life. The molecular diagnosis may tailor the choice of the most appropriate treatment, with the aim to optimize blood glucose control, reduce the risk of hypoglycemic events and long-term complications, and enable proper genetic counseling. Treatment is usually unnecessary for patients with mutations in the GCK gene, while oral hypoglycemic agents (generally sulphonylureas) are recommended for patients with mutations in the HNF4A and HNF1A genes. More recent data show that other glucose-lowering agents can be effective in the latter patients, and additional and alternative therapies have been proposed. Proper management guidelines during pregnancy have been developed for carriers of GCK gene mutations, but such guidelines are still a subject of debate in other cases, although some recommendations are available. The other subtypes of MODY are even more rare, and very little data are available in the literature. In this review we summarize the most pertinent findings and recommendations on the treatment of patients with the different subtypes of MODY. Our aim is to provide the reader with an easy-to-read update that can be used to drive the clinician’s therapeutical approach to these patients after the molecular diagnosis.
Literature
1.
go back to reference Hattersley AT, Greeley SAW, Polak M, et al. ISPAD Clinical Practice Consensus Guidelines 2018: the diagnosis and management of monogenic diabetes in children and adolescents. Pediatr Diabetes. 2018;19(Suppl 27):47–63.PubMed Hattersley AT, Greeley SAW, Polak M, et al. ISPAD Clinical Practice Consensus Guidelines 2018: the diagnosis and management of monogenic diabetes in children and adolescents. Pediatr Diabetes. 2018;19(Suppl 27):47–63.PubMed
2.
go back to reference American Diabetes Association. Classification and diagnosis of diabetes: standards of medical care in diabetes—2020. Diabetes Care. 2020;43(Suppl 1):S14–31. American Diabetes Association. Classification and diagnosis of diabetes: standards of medical care in diabetes—2020. Diabetes Care. 2020;43(Suppl 1):S14–31.
3.
go back to reference El Samahy MH, Matter RM, Youssef OI, et al. Relation between carotid intima media thickness and oxidative stress markers in type 1 diabetic children and adolescents. J Diabetes Metab Disord. 2013;12(1):50.PubMedPubMedCentral El Samahy MH, Matter RM, Youssef OI, et al. Relation between carotid intima media thickness and oxidative stress markers in type 1 diabetic children and adolescents. J Diabetes Metab Disord. 2013;12(1):50.PubMedPubMedCentral
4.
go back to reference Faienza MF, Acquafredda A, Tesse R, et al. Risk factors for subclinical atherosclerosis in diabetic and obese children. Int J Med Sci. 2013;10(3):338–43.PubMedPubMedCentral Faienza MF, Acquafredda A, Tesse R, et al. Risk factors for subclinical atherosclerosis in diabetic and obese children. Int J Med Sci. 2013;10(3):338–43.PubMedPubMedCentral
5.
6.
go back to reference Naylor RN, John PM, Winn AN, et al. Cost-effectiveness of MODY genetic testing: translating genomic advances into practical health applications. Diabetes Care. 2014;37(1):202–9.PubMed Naylor RN, John PM, Winn AN, et al. Cost-effectiveness of MODY genetic testing: translating genomic advances into practical health applications. Diabetes Care. 2014;37(1):202–9.PubMed
7.
go back to reference Shields BM, McDonald TJ, Ellard S, Campbell MJ, Hyde C, Hattersley AT. The development and validation of a clinical prediction model to determine the probability of MODY in patients with young-onset diabetes. Diabetologia. 2012;55(5):1265–72.PubMedPubMedCentral Shields BM, McDonald TJ, Ellard S, Campbell MJ, Hyde C, Hattersley AT. The development and validation of a clinical prediction model to determine the probability of MODY in patients with young-onset diabetes. Diabetologia. 2012;55(5):1265–72.PubMedPubMedCentral
8.
go back to reference Bacon S, Kyithar MP, Rizvi SR, et al. Successful maintenance on sulphonylurea therapy and low diabetes complication rates in a HNF1A-MODY cohort. Diabet Med. 2016;33(7):976–84.PubMed Bacon S, Kyithar MP, Rizvi SR, et al. Successful maintenance on sulphonylurea therapy and low diabetes complication rates in a HNF1A-MODY cohort. Diabet Med. 2016;33(7):976–84.PubMed
9.
go back to reference Pearson ER, Boj SF, Steele AM, et al. Macrosomia and hyperinsulinaemic hypoglycaemia in patients with heterozygous mutations in the HNF4A gene. PloS Med. 2007;4:e118.PubMedPubMedCentral Pearson ER, Boj SF, Steele AM, et al. Macrosomia and hyperinsulinaemic hypoglycaemia in patients with heterozygous mutations in the HNF4A gene. PloS Med. 2007;4:e118.PubMedPubMedCentral
10.
go back to reference Pearson ER, Pruhova S, Tack CJ, et al. Molecular genetics and phenotypic characteristics of MODY caused by hepatocyte nuclear factor 4alpha mutations in a large European collection. Diabetologia. 2005;48(5):878–85.PubMed Pearson ER, Pruhova S, Tack CJ, et al. Molecular genetics and phenotypic characteristics of MODY caused by hepatocyte nuclear factor 4alpha mutations in a large European collection. Diabetologia. 2005;48(5):878–85.PubMed
11.
go back to reference Hamilton AJ, Bingham C, McDonald TJ, et al. The HNF4A R76W mutation causes atypical dominant Fanconi syndrome in addition to a beta cell phenotype. J Med Genet. 2014;51(3):165–9.PubMed Hamilton AJ, Bingham C, McDonald TJ, et al. The HNF4A R76W mutation causes atypical dominant Fanconi syndrome in addition to a beta cell phenotype. J Med Genet. 2014;51(3):165–9.PubMed
12.
go back to reference Delvecchio M, Di Paola R, Mangiacotti D, et al. Clinical heterogeneity of abnormal glucose homeostasis associated with the HNF4A R311H mutation. Ital J Pediatr. 2014;40:58.PubMedPubMedCentral Delvecchio M, Di Paola R, Mangiacotti D, et al. Clinical heterogeneity of abnormal glucose homeostasis associated with the HNF4A R311H mutation. Ital J Pediatr. 2014;40:58.PubMedPubMedCentral
13.
go back to reference Stride A, Hattersley AT. Different genes, different diabetes: lessons from maturity-onset diabetes of the young. Ann Med. 2002;34(3):207–16.PubMed Stride A, Hattersley AT. Different genes, different diabetes: lessons from maturity-onset diabetes of the young. Ann Med. 2002;34(3):207–16.PubMed
14.
go back to reference Kyithar MP, Bacon S, Pannu KK, et al. Identification of HNF1A-MODY and HNF4A–MODY in Irish families: phenotypic characteristics and therapeutic implications. Diabetes Metab. 2011;37(6):512–9.PubMed Kyithar MP, Bacon S, Pannu KK, et al. Identification of HNF1A-MODY and HNF4A–MODY in Irish families: phenotypic characteristics and therapeutic implications. Diabetes Metab. 2011;37(6):512–9.PubMed
15.
go back to reference Delvecchio M, Mozzillo E, Salzano G, et al. Monogenic diabetes accounts for 6.3% of cases referred to 15 Italian pediatric diabetes centers during 2007 to 2012. J Clin Endocrinol Metab. 2017;102(6):1826–34.PubMed Delvecchio M, Mozzillo E, Salzano G, et al. Monogenic diabetes accounts for 6.3% of cases referred to 15 Italian pediatric diabetes centers during 2007 to 2012. J Clin Endocrinol Metab. 2017;102(6):1826–34.PubMed
16.
go back to reference Pearson ER, Velho G, Clark P, et al. Beta-cell genes and diabetes: quantitative and qualitative differences in the pathophysiology of hepatic nuclear factor-1alpha and glucokinase mutations. Diabetes. 2001;50(Suppl 1):S101–7.PubMed Pearson ER, Velho G, Clark P, et al. Beta-cell genes and diabetes: quantitative and qualitative differences in the pathophysiology of hepatic nuclear factor-1alpha and glucokinase mutations. Diabetes. 2001;50(Suppl 1):S101–7.PubMed
17.
go back to reference Hansen T, Eiberg H, Rouard M, et al. Novel MODY3 mutations in the hepatocyte nuclear factor-1α gene: evidence for a hyperexcitability of pancreatic β-cells to intravenous secretagogues in a glucose-tolerant carrier of a P447L mutation. Diabetes. 1997;46(4):726–30.PubMed Hansen T, Eiberg H, Rouard M, et al. Novel MODY3 mutations in the hepatocyte nuclear factor-1α gene: evidence for a hyperexcitability of pancreatic β-cells to intravenous secretagogues in a glucose-tolerant carrier of a P447L mutation. Diabetes. 1997;46(4):726–30.PubMed
18.
go back to reference Shepherd M, Brook AJ, Chakera AJ, Hattersley AT. Management of sulfonylurea-treated monogenic diabetes in pregnancy: implications of placental glibenclamide transfer. Diabet Med. 2017;34(10):1332–9.PubMedPubMedCentral Shepherd M, Brook AJ, Chakera AJ, Hattersley AT. Management of sulfonylurea-treated monogenic diabetes in pregnancy: implications of placental glibenclamide transfer. Diabet Med. 2017;34(10):1332–9.PubMedPubMedCentral
19.
go back to reference Steele AM, Wensley KJ, Ellard S, et al. Use of HbA1c in the identification of patients with hyperglycaemia caused by a glucokinase mutation: observational case control studies. PLoS One. 2013;148(6):e65326. Steele AM, Wensley KJ, Ellard S, et al. Use of HbA1c in the identification of patients with hyperglycaemia caused by a glucokinase mutation: observational case control studies. PLoS One. 2013;148(6):e65326.
20.
go back to reference Delvecchio M, Salzano G, Bonura C, et al. Can HbA1c combined with fasting plasma glucose help to assess priority for GCK–MODY vs HNF1A-MODY genetic testing? Acta Diabetol. 2018;55(9):981–3.PubMed Delvecchio M, Salzano G, Bonura C, et al. Can HbA1c combined with fasting plasma glucose help to assess priority for GCK–MODY vs HNF1A-MODY genetic testing? Acta Diabetol. 2018;55(9):981–3.PubMed
21.
go back to reference Pinelli M, Acquaviva F, Barbetti F, et al. Identification of candidate children for maturity-onset diabetes of the young type 2 (MODY2) gene testing: a seven-item clinical flowchart (7-iF). PLoS One. 2013;8(11):e79933.PubMedPubMedCentral Pinelli M, Acquaviva F, Barbetti F, et al. Identification of candidate children for maturity-onset diabetes of the young type 2 (MODY2) gene testing: a seven-item clinical flowchart (7-iF). PLoS One. 2013;8(11):e79933.PubMedPubMedCentral
22.
go back to reference Velho G, Blanché H, Vaxillaire M, et al. Identification of 14 new glucokinase mutations and description of the clinical profile of 42 MODY-2 families. Diabetologia. 1997;40:217–24.PubMed Velho G, Blanché H, Vaxillaire M, et al. Identification of 14 new glucokinase mutations and description of the clinical profile of 42 MODY-2 families. Diabetologia. 1997;40:217–24.PubMed
23.
go back to reference Steele AM, Shields BM, Wensley KJ, Colclough K, Ellard S, Hattersley AT. Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA. 2014;311(3):279–86.PubMed Steele AM, Shields BM, Wensley KJ, Colclough K, Ellard S, Hattersley AT. Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA. 2014;311(3):279–86.PubMed
24.
go back to reference Stride A, Vaxillaire M, Tuomi T, et al. The genetic abnormality in the beta cell determines the response to an oral glucose load. Diabetologia. 2014;45(3):427–35. Stride A, Vaxillaire M, Tuomi T, et al. The genetic abnormality in the beta cell determines the response to an oral glucose load. Diabetologia. 2014;45(3):427–35.
25.
go back to reference Hohendorff J, Szopa M, Skupien J, et al. A single dose of dapagliflozin, an SGLT-2 inhibitor, induces higher glycosuria in GCK- and HNF1A-MODY than in type 2 diabetes mellitus. Endocrine. 2017;57(2):272–9.PubMedPubMedCentral Hohendorff J, Szopa M, Skupien J, et al. A single dose of dapagliflozin, an SGLT-2 inhibitor, induces higher glycosuria in GCK- and HNF1A-MODY than in type 2 diabetes mellitus. Endocrine. 2017;57(2):272–9.PubMedPubMedCentral
26.
go back to reference Chakera AJ, Spyer G, Vincent N, Ellard S, Hattersley AT, Dunne FP. The 0.1% of the population with glucokinase monogenic diabetes can be recognized by clinical characteristics in pregnancy: the Atlantic Diabetes in Pregnancy cohort. Diabetes Care. 2014;37(5):1230–6.PubMed Chakera AJ, Spyer G, Vincent N, Ellard S, Hattersley AT, Dunne FP. The 0.1% of the population with glucokinase monogenic diabetes can be recognized by clinical characteristics in pregnancy: the Atlantic Diabetes in Pregnancy cohort. Diabetes Care. 2014;37(5):1230–6.PubMed
27.
go back to reference Rudland VL, Hinchcliffe M, Pinner J, et al. Identifying glucokinase monogenic diabetes in a multiethnic gestational diabetes mellitus cohort: new pregnancy screening criteria and utility of HbA1c. Diabetes Care. 2016;39(1):50–2.PubMed Rudland VL, Hinchcliffe M, Pinner J, et al. Identifying glucokinase monogenic diabetes in a multiethnic gestational diabetes mellitus cohort: new pregnancy screening criteria and utility of HbA1c. Diabetes Care. 2016;39(1):50–2.PubMed
28.
go back to reference Ellard S, Beards F, Allen LI, et al. A high prevalence of glucokinase mutations in gestational diabetic subjects selected by clinical criteria. Diabetologia. 2000;43(2):250–3.PubMed Ellard S, Beards F, Allen LI, et al. A high prevalence of glucokinase mutations in gestational diabetic subjects selected by clinical criteria. Diabetologia. 2000;43(2):250–3.PubMed
29.
go back to reference Hattersley AT, Beards F, Ballantyne E, et al. Mutations in the glucokinase gene of the fetus result in reduced birth weight. Nat Genet. 1998;19:268–70.PubMed Hattersley AT, Beards F, Ballantyne E, et al. Mutations in the glucokinase gene of the fetus result in reduced birth weight. Nat Genet. 1998;19:268–70.PubMed
30.
go back to reference Spyer G, Macleod KM, Shepherd M, et al. Pregnancy outcome in patients with raised blood glucose due to a heterozygous glucokinase gene mutation. Diabet Med. 2009;26:14–8.PubMed Spyer G, Macleod KM, Shepherd M, et al. Pregnancy outcome in patients with raised blood glucose due to a heterozygous glucokinase gene mutation. Diabet Med. 2009;26:14–8.PubMed
31.
go back to reference Spyer G, Hattersley AT, Sykes JE, Sturley RH, MacLeod KM. Influence of maternal and fetal glucokinase mutations in gestational diabetes. Am J Obstet Gynecol. 2001;185:240–1.PubMed Spyer G, Hattersley AT, Sykes JE, Sturley RH, MacLeod KM. Influence of maternal and fetal glucokinase mutations in gestational diabetes. Am J Obstet Gynecol. 2001;185:240–1.PubMed
32.
go back to reference Hattersley AT, Pearson ER. Minireview: pharmacogenetics and beyond: the interaction of therapeutic response, beta-cell physiology, and genetics in diabetes. Endocrinology. 2006;147:2657–63.PubMed Hattersley AT, Pearson ER. Minireview: pharmacogenetics and beyond: the interaction of therapeutic response, beta-cell physiology, and genetics in diabetes. Endocrinology. 2006;147:2657–63.PubMed
33.
go back to reference Chakera AJ, Steele AM, Gloyn AL, et al. Recognition and management of individuals with hyperglycemia because of a heterozygous glucokinase mutation. Diabetes Care. 2015;38(7):1383–92.PubMed Chakera AJ, Steele AM, Gloyn AL, et al. Recognition and management of individuals with hyperglycemia because of a heterozygous glucokinase mutation. Diabetes Care. 2015;38(7):1383–92.PubMed
34.
go back to reference Murphy R, Ellard S, Hattersley AT. Clinical implications of a molecular genetic classification of monogenic beta-cell diabetes. Nat Clin Pract Endocrinol Metab. 2008;4(4):200–13.PubMed Murphy R, Ellard S, Hattersley AT. Clinical implications of a molecular genetic classification of monogenic beta-cell diabetes. Nat Clin Pract Endocrinol Metab. 2008;4(4):200–13.PubMed
35.
go back to reference Isomaa B, Henricsson M, Lehto M, et al. Chronic diabetic complications in patients with MODY3 diabetes. Diabetologia. 1998;41(4):467–73.PubMed Isomaa B, Henricsson M, Lehto M, et al. Chronic diabetic complications in patients with MODY3 diabetes. Diabetologia. 1998;41(4):467–73.PubMed
36.
go back to reference Stride A, Ellard S, Clark P, et al. Beta-cell dysfunction, insulin sensitivity, and glycosuria precede diabetes in hepatocyte nuclear factor-1alpha mutation carriers. Diabetes Care. 2005;28(7):1751–6.PubMed Stride A, Ellard S, Clark P, et al. Beta-cell dysfunction, insulin sensitivity, and glycosuria precede diabetes in hepatocyte nuclear factor-1alpha mutation carriers. Diabetes Care. 2005;28(7):1751–6.PubMed
37.
go back to reference Valkovicova T, Skopkova M, Stanik J, Gasperikova D. Novel insights into genetics and clinics of the HNF1A-MODY. Endocr Regul. 2019;53(2):110–34.PubMed Valkovicova T, Skopkova M, Stanik J, Gasperikova D. Novel insights into genetics and clinics of the HNF1A-MODY. Endocr Regul. 2019;53(2):110–34.PubMed
38.
go back to reference Wollheim CB. Beta-cell mitochondria in the regulation of insulin secretion: a new culprit in type II diabetes. Diabetologia. 2000;43(3):265–77.PubMed Wollheim CB. Beta-cell mitochondria in the regulation of insulin secretion: a new culprit in type II diabetes. Diabetologia. 2000;43(3):265–77.PubMed
39.
go back to reference Raile K, Schober E, Konrad K, et al. Treatment of young patients with HNF1A mutations (HNF1A-MODY). Diabet Med. 2015;32(4):526–30.PubMed Raile K, Schober E, Konrad K, et al. Treatment of young patients with HNF1A mutations (HNF1A-MODY). Diabet Med. 2015;32(4):526–30.PubMed
40.
go back to reference Khelifa SB, Dendana A, Barboura I, et al. Successful switch from insulin to oral sulfonylurea therapy in HNF1A-MODY Tunisian patient with the P291fsinsC mutation. Diabetes Res Clin Pract. 2016;115:133–6.PubMed Khelifa SB, Dendana A, Barboura I, et al. Successful switch from insulin to oral sulfonylurea therapy in HNF1A-MODY Tunisian patient with the P291fsinsC mutation. Diabetes Res Clin Pract. 2016;115:133–6.PubMed
41.
go back to reference Pearson ER, Starkey BJ, Powell RJ, Gribble FM, Clark PM, Hattersley AT. Genetic cause of hyperglycaemia and response to treatment in diabetes. Lancet. 2003;362(9392):1275–81.PubMed Pearson ER, Starkey BJ, Powell RJ, Gribble FM, Clark PM, Hattersley AT. Genetic cause of hyperglycaemia and response to treatment in diabetes. Lancet. 2003;362(9392):1275–81.PubMed
42.
go back to reference Shepherd M, Pearson ER, Houghton J, Salt G, Ellard S, Hattersley AT. No deterioration in glycemic control in HNF-1alpha maturity-onset diabetes of the young following transfer from long-term insulin to sulphonylureas. Diabetes Care. 2003;26(11):3191–2.PubMed Shepherd M, Pearson ER, Houghton J, Salt G, Ellard S, Hattersley AT. No deterioration in glycemic control in HNF-1alpha maturity-onset diabetes of the young following transfer from long-term insulin to sulphonylureas. Diabetes Care. 2003;26(11):3191–2.PubMed
43.
go back to reference Brunerova L, Rahelić D, Ceriello A, Broz J. Use of oral antidiabetic drugs in the treatment of maturity-onset diabetes of the young: a mini review. Diabetes Metab Res Rev. 2018;34(1):e2940. Brunerova L, Rahelić D, Ceriello A, Broz J. Use of oral antidiabetic drugs in the treatment of maturity-onset diabetes of the young: a mini review. Diabetes Metab Res Rev. 2018;34(1):e2940.
44.
go back to reference Tripathy D, Carlsson Å-L, Lehto M, Isomaa B, Tuomi T, Groop L. Insulin secretion and insulin sensitivity in diabetic subgroups: studies in the prediabetic and diabetic state. Diabetologia. 2000;43(12):1476–83.PubMed Tripathy D, Carlsson Å-L, Lehto M, Isomaa B, Tuomi T, Groop L. Insulin secretion and insulin sensitivity in diabetic subgroups: studies in the prediabetic and diabetic state. Diabetologia. 2000;43(12):1476–83.PubMed
45.
go back to reference Pearson E, Liddell W, Shepherd M, Corrall R, Hattersley A. Sensitivity to sulphonylureas in patients with HNF-1α gene mutations: evidence for pharmacogenetics in diabetes. Diabet Med. 2000;17:543–5.PubMed Pearson E, Liddell W, Shepherd M, Corrall R, Hattersley A. Sensitivity to sulphonylureas in patients with HNF-1α gene mutations: evidence for pharmacogenetics in diabetes. Diabet Med. 2000;17:543–5.PubMed
46.
go back to reference Urbanova J, Andel M, Potockova J, et al. Half-life of sulfonylureas in HNF1A and HNF4A human MODY patients is not prolonged as suggested by the mouse Hnf1a(−/−) model. Curr Pharm Des. 2015;21(39):5736–48.PubMed Urbanova J, Andel M, Potockova J, et al. Half-life of sulfonylureas in HNF1A and HNF4A human MODY patients is not prolonged as suggested by the mouse Hnf1a(−/−) model. Curr Pharm Des. 2015;21(39):5736–48.PubMed
47.
go back to reference Boileau P, Wolfrum C, Shih D, Stoffel M. Decreased glibenclamide uptake in hepatocytes of HNF-1α defficient mice. Diabetes. 2002;51(3):S343–8.PubMed Boileau P, Wolfrum C, Shih D, Stoffel M. Decreased glibenclamide uptake in hepatocytes of HNF-1α defficient mice. Diabetes. 2002;51(3):S343–8.PubMed
48.
go back to reference Demol S, Lebenthal Y, Bar-Meisels M, Phillip M, Gat-Yablonski G, Gozlan Y. A family with a novel termination mutation in hepatic nuclear factor 1α in maturity-onset diabetes of the young type 3 which is unresponsive to sulphonylurea therapy. Horm Res Paediatr. 2014;81(4):280–4.PubMed Demol S, Lebenthal Y, Bar-Meisels M, Phillip M, Gat-Yablonski G, Gozlan Y. A family with a novel termination mutation in hepatic nuclear factor 1α in maturity-onset diabetes of the young type 3 which is unresponsive to sulphonylurea therapy. Horm Res Paediatr. 2014;81(4):280–4.PubMed
49.
go back to reference Shepherd M, Shields B, Ellard S, Rubio-Cabezas O, Hattersley AT. A genetic diagnosis of HNF1A diabetes alters treatment and improves glycaemic control in the majority of insulin-treated patients. Diabet Med. 2009;26:437–41.PubMed Shepherd M, Shields B, Ellard S, Rubio-Cabezas O, Hattersley AT. A genetic diagnosis of HNF1A diabetes alters treatment and improves glycaemic control in the majority of insulin-treated patients. Diabet Med. 2009;26:437–41.PubMed
50.
go back to reference Tuomi T, Honkanen EH, Isomaa B, Sarelin L, Groop LC. Improved prandial glucose control with lower risk of hypoglycemia with nateglinide than with glibenclamide in patients with maturity-onset diabetes of the young type 3. Diabetes Care. 2006;29(2):189–94.PubMed Tuomi T, Honkanen EH, Isomaa B, Sarelin L, Groop LC. Improved prandial glucose control with lower risk of hypoglycemia with nateglinide than with glibenclamide in patients with maturity-onset diabetes of the young type 3. Diabetes Care. 2006;29(2):189–94.PubMed
51.
go back to reference Østoft SH, Bagger JI, Hansen T, et al. Incretin effect and glucagon responses to oral and intravenous glucose in patients with maturity-onset diabetes of the young—type 2 and type 3. Diabetes. 2014;63(8):2838–44.PubMed Østoft SH, Bagger JI, Hansen T, et al. Incretin effect and glucagon responses to oral and intravenous glucose in patients with maturity-onset diabetes of the young—type 2 and type 3. Diabetes. 2014;63(8):2838–44.PubMed
52.
go back to reference Becker M, Galler A, Raile K. Meglitinide analogues in adolescent patients with HNF1A-MODY (MODY 3). Pediatrics. 2014;133(3):e775–9.PubMed Becker M, Galler A, Raile K. Meglitinide analogues in adolescent patients with HNF1A-MODY (MODY 3). Pediatrics. 2014;133(3):e775–9.PubMed
53.
go back to reference Docena MK, Faiman C, Stanley CM, Pantalone KM. Mody-3: novel HNF1A mutation and the utility of glucagon-like peptide (GLP)-1 receptor agonist therapy. Endocr Pract. 2014;20(2):107–11.PubMed Docena MK, Faiman C, Stanley CM, Pantalone KM. Mody-3: novel HNF1A mutation and the utility of glucagon-like peptide (GLP)-1 receptor agonist therapy. Endocr Pract. 2014;20(2):107–11.PubMed
54.
go back to reference Fantasia KL, Steenkamp DW. Optimal glycemic control in a patient with HNF1A MODY with GLP-1 RA monotherapy: implications for future therapy. J Endocr Soc. 2019;3(12):2286–9.PubMedPubMedCentral Fantasia KL, Steenkamp DW. Optimal glycemic control in a patient with HNF1A MODY with GLP-1 RA monotherapy: implications for future therapy. J Endocr Soc. 2019;3(12):2286–9.PubMedPubMedCentral
55.
go back to reference Østoft SH, Bagger JI, Hansen T, et al. Glucose-lowering effects and low risk of hypoglycemia in patients with maturity-onset diabetes of the young when treated with a GLP-1 receptor agonist: a double-blind, randomized, crossover trial. Diabetes Care. 2014;37(7):1797–805.PubMed Østoft SH, Bagger JI, Hansen T, et al. Glucose-lowering effects and low risk of hypoglycemia in patients with maturity-onset diabetes of the young when treated with a GLP-1 receptor agonist: a double-blind, randomized, crossover trial. Diabetes Care. 2014;37(7):1797–805.PubMed
56.
go back to reference Christensen AS, Haedersdal S, Storgaard H, et al. 981-P:GIP and GLP-1 potentiate sulfonylurea-induced insulin secretion in HNF1A diabetes. Diabetes. 2019;68(Supp 1):981-P. Christensen AS, Haedersdal S, Storgaard H, et al. 981-P:GIP and GLP-1 potentiate sulfonylurea-induced insulin secretion in HNF1A diabetes. Diabetes. 2019;68(Supp 1):981-P.
57.
go back to reference Christensen AS, Storgaard H, Hædersdal S, Hansen T, Knop FK, Vilsbøll T. Glimepiride monotherapy versus combination of glimepiride and linagliptin therapy in patients with HNF1A-diabetes: a protocol for a randomised, double-blinded, placebo-controlled trial. BMJ Open. 2018;8(10):e022517. Christensen AS, Storgaard H, Hædersdal S, Hansen T, Knop FK, Vilsbøll T. Glimepiride monotherapy versus combination of glimepiride and linagliptin therapy in patients with HNF1A-diabetes: a protocol for a randomised, double-blinded, placebo-controlled trial. BMJ Open. 2018;8(10):e022517.
58.
go back to reference Delvecchio M, Ludovico O, Menzaghi C, et al. Low prevalence of HNF1A mutations after molecular screening of multiple MODY genes in 58 Italian families recruited in the pediatric or adult diabetes clinic from a single Italian hospital. Diabetes Care. 2014;37(12):e258–60.PubMed Delvecchio M, Ludovico O, Menzaghi C, et al. Low prevalence of HNF1A mutations after molecular screening of multiple MODY genes in 58 Italian families recruited in the pediatric or adult diabetes clinic from a single Italian hospital. Diabetes Care. 2014;37(12):e258–60.PubMed
59.
go back to reference Ahlgren U, Jonsson J, Jonsson L, Simu K, Edlund H. Beta-cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the betacell phenotype and maturity onset diabetes. Genes Dev. 1998;12:1763–8.PubMedPubMedCentral Ahlgren U, Jonsson J, Jonsson L, Simu K, Edlund H. Beta-cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the betacell phenotype and maturity onset diabetes. Genes Dev. 1998;12:1763–8.PubMedPubMedCentral
60.
go back to reference Edfalk S, Steneberg P, Edlund H. Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion. Diabetes. 2008;57(9):2280–7.PubMedPubMedCentral Edfalk S, Steneberg P, Edlund H. Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion. Diabetes. 2008;57(9):2280–7.PubMedPubMedCentral
61.
go back to reference Fajans SS, Bell GI, Polonsky KS. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med. 2001;345(13):971–80.PubMed Fajans SS, Bell GI, Polonsky KS. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med. 2001;345(13):971–80.PubMed
62.
go back to reference Deng M, Xiao X, Zhou L, Wang T. First case report of maturity-onset diabetes of the young type 4 pedigree in a Chinese family. Front Endocrinol (Lausanne). 2019;10:406.PubMedPubMedCentral Deng M, Xiao X, Zhou L, Wang T. First case report of maturity-onset diabetes of the young type 4 pedigree in a Chinese family. Front Endocrinol (Lausanne). 2019;10:406.PubMedPubMedCentral
63.
go back to reference Mangrum C, Rush E, Shivaswamy V. Genetically targeted dipeptidyl peptidase-4 inhibitor use in a patient with a novel mutation of MODY type 4. Clin Med Insights Endocrinol Diabetes. 2015;8:83–6.PubMedPubMedCentral Mangrum C, Rush E, Shivaswamy V. Genetically targeted dipeptidyl peptidase-4 inhibitor use in a patient with a novel mutation of MODY type 4. Clin Med Insights Endocrinol Diabetes. 2015;8:83–6.PubMedPubMedCentral
64.
go back to reference Ulinski T, Lescure S, Beaufils S, et al. Renal phenotypes related to hepatocyte nuclear factor-1beta (TCF2) mutations in a pediatric cohort. J Am Soc Nephrol. 2006;17(2):497–503.PubMed Ulinski T, Lescure S, Beaufils S, et al. Renal phenotypes related to hepatocyte nuclear factor-1beta (TCF2) mutations in a pediatric cohort. J Am Soc Nephrol. 2006;17(2):497–503.PubMed
65.
go back to reference Bellanné-Chantelot C, Clauin S, Chauveau D, et al. Large genomic rearrangements in the hepatocyte nuclear factor-1beta (TCF2) gene are the most frequent cause of maturity-onset diabetes of the young type 5. Diabetes. 2005;54(11):3126–32.PubMed Bellanné-Chantelot C, Clauin S, Chauveau D, et al. Large genomic rearrangements in the hepatocyte nuclear factor-1beta (TCF2) gene are the most frequent cause of maturity-onset diabetes of the young type 5. Diabetes. 2005;54(11):3126–32.PubMed
66.
go back to reference Dubois-Laforgue D, Cornu E, Saint-Martin C, et al. Diabetes, associated clinical spectrum, long-term prognosis, and genotype/phenotype correlations in 201 adult patients with hepatocyte nuclear factor 1B (HNF1B) molecular defects. Diabetes Care. 2017;40(11):1436–43.PubMed Dubois-Laforgue D, Cornu E, Saint-Martin C, et al. Diabetes, associated clinical spectrum, long-term prognosis, and genotype/phenotype correlations in 201 adult patients with hepatocyte nuclear factor 1B (HNF1B) molecular defects. Diabetes Care. 2017;40(11):1436–43.PubMed
67.
go back to reference Pearson ER, Badman MK, Lockwood CR, et al. Contrasting diabetes phenotypes associated with hepatocyte nuclear factor-1alpha and-1beta mutations. Diabetes Care. 2004;27(5):1102–7.PubMed Pearson ER, Badman MK, Lockwood CR, et al. Contrasting diabetes phenotypes associated with hepatocyte nuclear factor-1alpha and-1beta mutations. Diabetes Care. 2004;27(5):1102–7.PubMed
68.
69.
go back to reference Bingham C, Hattersley AT. Renal cysts and diabetes syndrome resulting from mutations in hepatocyte nuclear factor-1beta. Nephrol Dial Transplant. 2004;19(11):2703–8.PubMed Bingham C, Hattersley AT. Renal cysts and diabetes syndrome resulting from mutations in hepatocyte nuclear factor-1beta. Nephrol Dial Transplant. 2004;19(11):2703–8.PubMed
70.
go back to reference Horikawa Y, Enya M, Mabe H, et al. NEUROD1-deficient diabetes (MODY6): identification of the first cases in Japanese and the clinical features. Pediatr Diabetes. 2018;19:236–42.PubMed Horikawa Y, Enya M, Mabe H, et al. NEUROD1-deficient diabetes (MODY6): identification of the first cases in Japanese and the clinical features. Pediatr Diabetes. 2018;19:236–42.PubMed
71.
go back to reference Horikawa Y, Enya M. Genetic dissection and clinical features of MODY6 (NEUROD1-MODY). Curr Diab Rep. 2019;19(3):12.PubMed Horikawa Y, Enya M. Genetic dissection and clinical features of MODY6 (NEUROD1-MODY). Curr Diab Rep. 2019;19(3):12.PubMed
72.
go back to reference Abreu GM, Tarantino RM, Cabello PH, et al. The first case of NEUROD1-MODY reported in Latin America. Mol Genet Genom Med. 2019;7(12):e989. Abreu GM, Tarantino RM, Cabello PH, et al. The first case of NEUROD1-MODY reported in Latin America. Mol Genet Genom Med. 2019;7(12):e989.
73.
go back to reference Neve B, Fernandez-Zapico ME, Ashkenazi-Katalan V, et al. Role of transcription factor KLF11 and its diabetes-associated gene variants in pancreatic beta cell function. Proc Natl Acad Sci USA. 2005;102(13):4807–12.PubMed Neve B, Fernandez-Zapico ME, Ashkenazi-Katalan V, et al. Role of transcription factor KLF11 and its diabetes-associated gene variants in pancreatic beta cell function. Proc Natl Acad Sci USA. 2005;102(13):4807–12.PubMed
74.
go back to reference Ushijima K, Narumi S, Ogata T, et al. KLF11 variant in a family clinically diagnosed with early childhood-onset type 1B diabetes. Pediatr Diabetes. 2019;20:712–9.PubMed Ushijima K, Narumi S, Ogata T, et al. KLF11 variant in a family clinically diagnosed with early childhood-onset type 1B diabetes. Pediatr Diabetes. 2019;20:712–9.PubMed
75.
go back to reference Raeder H, Johansson S, Holm PI, et al. Mutations in the CEL VNTR cause a syndrome of diabetes and pancreatic exocrine dysfunction. Nat Genet. 2006;38:54–62.PubMed Raeder H, Johansson S, Holm PI, et al. Mutations in the CEL VNTR cause a syndrome of diabetes and pancreatic exocrine dysfunction. Nat Genet. 2006;38:54–62.PubMed
76.
go back to reference Plengvidhya N, Kooptiwut S, Songtawee N, et al. PAX4 mutations in Thais with maturity-onset diabetes of the young. J Clin Endocrinol Metab. 2007;92(7):2821–6.PubMed Plengvidhya N, Kooptiwut S, Songtawee N, et al. PAX4 mutations in Thais with maturity-onset diabetes of the young. J Clin Endocrinol Metab. 2007;92(7):2821–6.PubMed
77.
go back to reference Sujjitjoon J, Kooptiwut S, Chongjaroen N, Tangjittipokin W, Plengvidhya N, Yenchitsomanus PT. Aberrant mRNA splicing of paired box 4 (PAX4) IVS7-1G>A mutation causing maturity-onset diabetes of the young, type 9. Acta Diabetol. 2016;53(2):205–16.PubMed Sujjitjoon J, Kooptiwut S, Chongjaroen N, Tangjittipokin W, Plengvidhya N, Yenchitsomanus PT. Aberrant mRNA splicing of paired box 4 (PAX4) IVS7-1G>A mutation causing maturity-onset diabetes of the young, type 9. Acta Diabetol. 2016;53(2):205–16.PubMed
78.
go back to reference Molven A, Ringdal M, Nordbø AM, et al. Mutations in the insulin gene can cause MODY and autoantibody-negative type 1 diabetes. Diabetes. 2008;57(4):1131–5.PubMed Molven A, Ringdal M, Nordbø AM, et al. Mutations in the insulin gene can cause MODY and autoantibody-negative type 1 diabetes. Diabetes. 2008;57(4):1131–5.PubMed
79.
go back to reference Edghill EL, Flanagan SE, Patch AM, et al. Insulin mutation screening in 1,044 patients with diabetes: mutations in the INS gene are a common cause of neonatal diabetes but a rare cause of diabetes diagnosed in childhood or adulthood. Diabetes. 2008;57(4):1034–42.PubMed Edghill EL, Flanagan SE, Patch AM, et al. Insulin mutation screening in 1,044 patients with diabetes: mutations in the INS gene are a common cause of neonatal diabetes but a rare cause of diabetes diagnosed in childhood or adulthood. Diabetes. 2008;57(4):1034–42.PubMed
80.
go back to reference Bonnefond A, Yengo L, Philippe J, et al. Reassessment of the putative role of BLK-p.A71T loss-of-function mutation in MODY and type 2 diabetes. Diabetologia. 2013;56(3):492–6.PubMed Bonnefond A, Yengo L, Philippe J, et al. Reassessment of the putative role of BLK-p.A71T loss-of-function mutation in MODY and type 2 diabetes. Diabetologia. 2013;56(3):492–6.PubMed
81.
go back to reference Mruthyunjaya MD, Chapla A, Shyamasunder AH, et al. Comprehensive maturity onset diabetes of the young (MODY) gene screening in pregnant women with diabetes in India. PLoS One. 2017;12(1):e0168656. Mruthyunjaya MD, Chapla A, Shyamasunder AH, et al. Comprehensive maturity onset diabetes of the young (MODY) gene screening in pregnant women with diabetes in India. PLoS One. 2017;12(1):e0168656.
82.
go back to reference Borowiec M, Liew CW, Thompson R, et al. Mutations at the BLK locus linked to maturity onset diabetes of the young and beta-cell dysfunction. Proc Natl Acad Sci USA. 2009;106(34):14460–5.PubMed Borowiec M, Liew CW, Thompson R, et al. Mutations at the BLK locus linked to maturity onset diabetes of the young and beta-cell dysfunction. Proc Natl Acad Sci USA. 2009;106(34):14460–5.PubMed
83.
go back to reference Kapoor RR, Flanagan SE, James CT, et al. Hyperinsulinaemic hypoglycaemia and diabetes mellitus due to dominant ABCC8/KCNJ11 mutations. Diabetologia. 2011;54:2575–83.PubMedPubMedCentral Kapoor RR, Flanagan SE, James CT, et al. Hyperinsulinaemic hypoglycaemia and diabetes mellitus due to dominant ABCC8/KCNJ11 mutations. Diabetologia. 2011;54:2575–83.PubMedPubMedCentral
84.
go back to reference Occal G, Flanagan SE, Hacihamdioglu B, et al. Clinical characteristics of recessive and dominant congenital hyperinsulinism due to mutation(s) in the ABCC8/KCNJ11 genes encoding the ATP-sensitive potassium channel in the pancreatic beta cell. J Pediatr Endocrinol Metab. 2011;24:1019–23. Occal G, Flanagan SE, Hacihamdioglu B, et al. Clinical characteristics of recessive and dominant congenital hyperinsulinism due to mutation(s) in the ABCC8/KCNJ11 genes encoding the ATP-sensitive potassium channel in the pancreatic beta cell. J Pediatr Endocrinol Metab. 2011;24:1019–23.
85.
go back to reference Rafiq M, Flanagan SE, Patch AM, Shields BM, Ellard S, Hattersley AT. Neonatal Diabetes International Collaborative Group. Effective treatment with oral sulfonylureas in peoples with diabetes due to sulfonylurea receptor 1 (SUR1) mutations. Diabetes Care. 2008;31:204–9.PubMed Rafiq M, Flanagan SE, Patch AM, Shields BM, Ellard S, Hattersley AT. Neonatal Diabetes International Collaborative Group. Effective treatment with oral sulfonylureas in peoples with diabetes due to sulfonylurea receptor 1 (SUR1) mutations. Diabetes Care. 2008;31:204–9.PubMed
86.
go back to reference Reilly F, Sanchez-Lechuga B, Clinton S, et al. Phenotype, genotype and glycaemic variability in people with activating mutations in the ABCC8 gene: response to appropriate therapy. Diabet Med. 2020;37(5):876–84.PubMed Reilly F, Sanchez-Lechuga B, Clinton S, et al. Phenotype, genotype and glycaemic variability in people with activating mutations in the ABCC8 gene: response to appropriate therapy. Diabet Med. 2020;37(5):876–84.PubMed
87.
go back to reference Bowman P, Sulen Å, Barbetti F, et al. Effectiveness and safety of long-term treatment with sulfonylureas in patients with neonatal diabetes due to KCNJ11 mutations: an international cohort study. Lancet Diabetes Endocrinol. 2018;6(8):637–46.PubMedPubMedCentral Bowman P, Sulen Å, Barbetti F, et al. Effectiveness and safety of long-term treatment with sulfonylureas in patients with neonatal diabetes due to KCNJ11 mutations: an international cohort study. Lancet Diabetes Endocrinol. 2018;6(8):637–46.PubMedPubMedCentral
88.
go back to reference Bonnefond A, Philippe J, Durand E, et al. Whole-exome sequencing and high throughput genotyping identified KCNJ11 as the thirteenth MODY gene. PLoS One. 2012;7(6):e37423.PubMedPubMedCentral Bonnefond A, Philippe J, Durand E, et al. Whole-exome sequencing and high throughput genotyping identified KCNJ11 as the thirteenth MODY gene. PLoS One. 2012;7(6):e37423.PubMedPubMedCentral
89.
go back to reference Prudente S, Jungtrakoon P, Marucci A, et al. Loss-of-function mutations in APPL1 in familial diabetes mellitus. Am J Hum Genet. 2015;97:177–85.PubMedPubMedCentral Prudente S, Jungtrakoon P, Marucci A, et al. Loss-of-function mutations in APPL1 in familial diabetes mellitus. Am J Hum Genet. 2015;97:177–85.PubMedPubMedCentral
Metadata
Title
Treatment Options for MODY Patients: A Systematic Review of Literature
Authors
Maurizio Delvecchio
Carmela Pastore
Paola Giordano
Publication date
01-08-2020
Publisher
Springer Healthcare
Published in
Diabetes Therapy / Issue 8/2020
Print ISSN: 1869-6953
Electronic ISSN: 1869-6961
DOI
https://doi.org/10.1007/s13300-020-00864-4

Other articles of this Issue 8/2020

Diabetes Therapy 8/2020 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.