Hostname: page-component-76dd75c94c-x59qb Total loading time: 0 Render date: 2024-04-30T08:15:18.033Z Has data issue: false hasContentIssue false

Updates of precision medicine in type 2 diabetes

Published online by Cambridge University Press:  14 April 2023

Mingfeng Xia
Affiliation:
Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
Xiaoying Li*
Affiliation:
Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
*
Corresponding author: Xiaoying Li; Email: li.xiaoying@zs-hospital.sh.cn
Rights & Permissions [Opens in a new window]

Abstract

Diabetes mellitus is prevalent worldwide and affects 1 in 10 adults. Despite the successful development of glucose-lowering drugs, such as glucagon-like peptide-1 (GLP-1) receptor agonists and sodium-glucose cotransporter-2 inhibitors recently, the proportion of patients achieving satisfactory glucose control has not risen as expected. The heterogeneity of diabetes determines that a one-size-fits-all strategy is not suitable for people with diabetes. Diabetes is undoubtedly more heterogeneous than the conventional subclassification, such as type 1, type 2, monogenic and gestational diabetes. The recent progress in genetics and epigenetics of diabetes has gradually unveiled the mechanisms underlying the heterogeneity of diabetes, and cluster analysis has shown promising results in the substratification of type 2 diabetes, which accounts for 95% of diabetic patients. More recently, the rapid development of sophisticated glucose monitoring and artificial intelligence technologies further enabled comprehensive consideration of the complex individual genetic and clinical information and might ultimately realize a precision diagnosis and treatment in diabetics.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2023. Published by Cambridge University Press

Impact statement

Diabetes mellitus has become a global public health crisis that affects 537 million people worldwide. Despite the great success in the development of novel glucose-lowering drugs, the proportion of patients achieving satisfactory glucose control has not risen as expected during the last decade. The heterogeneity of diabetes determines that a one-size-fits-all strategy is not suitable for people with diabetes. Our review article summarized the current progress in heterogeneity of diabetes from the perspective of genetics and epigenetics, and introduced a promising clinical substratification of type 2 diabetes. Thanks to the recent rapid development of sophisticated glucose monitoring and artificial intelligence technologies in the management of diabetes, we are able to process a large number of individual multi-dimensional genetic, anthropometric, clinical, biochemical and imaging information and make objective and correct judgment to improve the long-term outcome of diabetic patients. The emergence of new technologies might provide solutions for precision diagnosis and treatment of diabetes.

Introduction

Diabetes mellitus is diagnosed if blood glucose concentration exceeds a threshold, which predisposes to microvascular and microvascular end-organ complications. Diabetes continues to increase in prevalence worldwide (Ingelfinger and Jarcho, Reference Ingelfinger and Jarcho2017). Currently, diabetes affects 537 million people worldwide (IDF, 2021). Diabetes is also the leading cause of disability globally (GBD 2017 Disease and Injury Incidence and Prevalence Collaborators, 2018), as well as causing an increase in the risk of death from cardiovascular disease, renal disease, and cancer, and reducing life expectancy by 4–10 years on average (Rao Kondapally Seshasai et al., Reference Rao Kondapally Seshasai, Kaptoge, Thompson, di Angelantonio, Gao, Sarwar, Whincup, Mukamal, Gillum, Holme, Njølstad, Fletcher, Nilsson, Lewington, Collins, Gudnason, Thompson, Sattar, Selvin, Hu and Danesh2011). Early and intensive management of diabetes to achieve the recommended glycemic and metabolic targets can reduce long-term diabetes complications (Khunti and Millar-Jones, Reference Khunti and Millar-Jones2017).

In recent decades, great success has been achieved in the development of novel glucose-lowering drugs, such as glucagon-like peptide-1 (GLP-1) receptor agonists and sodium-glucose cotransporter-2 inhibitors. At present, pharmacological therapies, comprising 10 classes of medicines approved by the FDA, could be utilized to control blood glucose. However, the proportion of patients achieving satisfactory glucose control has not risen as expected (Bhat et al., Reference Bhat, Chowta, Chowta, Shastry and Kamath2021). The inadequate understanding of the diverse pathophysiological mechanisms and personalized treatment of diabetes partly limits our ability to treat diabetes.

The aim of our current review is to provide an overview of precision medicine in diabetes, focusing on the genetics and epigenetics, clinical stratification and personalized prevention, treatment of this disease and its related complications (Figure 1).

Figure 1. The precision medicine in diabetes management.

Heterogeneity of diabetes

The heterogeneity of diabetes was recognized several decades ago, when diabetic patients were divided into insulin-sensitive and insulin-insensitive subgroups based on the oral glucose tolerance test (OGTT) (Himsworth and Kerr, Reference Himsworth and Kerr1939). In 1979, the classification of type 1 and type 2 diabetes was first proposed by the American Diabetes Association (National Diabetes Data Group, 1979). Meanwhile, Fajans and Tattersall described a subgroup of diabetes with inheritance across many generations called “maturity onset diabetes in the young” (MODY) (Tattersall and Fajans, Reference Tattersall and Fajans1975). However, it was not until the introduction of genomic medicine in the recent 20 years, that the molecular mechanisms of the monogenic diabetes were uncovered.

Patients with diabetes do not equally respond to glucose-lowering therapies. Thus, pharmacologic intervention should be individualized based on factors such as duration of diabetes, presence of existing comorbidities, expected duration of life, weight, age, family history of diabetes-related complications and funding for prescribed medications and technology. Previous studies indicated that approximately 30% of individuals with type 2 diabetes do not respond well to metformin, and 5% have intolerable side effects (Kahn et al., Reference Kahn, Haffner, Heise, Herman, Holman, Jones, Kravitz, Lachin, O’Neill, Zinman and Viberti2006; Cook et al., Reference Cook, Girman, Stein and Alexander2007). Clinical diabetic patients with different etiological processes, such as obesity, metabolic syndrome, beta cell dysfunction or lipodystrophy, respond differently to glucose-lowering drugs with different mechanisms (Udler and Kim, Reference Udler, Kim, von Grotthuss, Bonàs-Guarch, Cole, Chiou, Boehnke, Laakso, Atzmon, Glaser, Mercader, Gaulton, Flannick, Getz and Florez2018). Patients from different ethnicities respond differently to glucose-lowering drugs, including dipeptidyl peptidase-4 inhibitors (Kim et al., Reference Kim, Hahn, Oh, Kwak, Park and Cho2013), metformin (Williams et al., Reference Williams, Padhukasahasram, Ahmedani, Peterson, Wells, González Burchard and Lanfear2014) and glucagon-like peptide 1 agonist (Velásquez-Mieyer et al., Reference Velásquez-Mieyer, Cowan, Pérez-Faustinelli, Nieto-Martínez, Villegas-Barreto, Tolley, Lustig and Alpert2008). Thus, stratification of diabetic patients based on their genetic backgrounds and pathophysiological mechanisms could be considered for implementation of precision medicine in diabetes.

Genetics of diabetes

Precision medication in monogenic diabetes has successfully guided clinical treatment. For example, individuals with rare mutation in HNF1A (MODY3), HNF4A (MODY1) and ABCC8 (MODY12) are incredibly sensitive to the effects of sulfonylureas (Pearson et al., Reference Pearson, Starkey, Powell, Gribble, Clark and Hattersley2003). While individuals with loss-of-function mutations in the GCK gene are unlikely to develop diabetic complications, and have no need for unnecessary treatment (Steele et al., Reference Steele, Shields, Wensley, Colclough, Ellard and Hattersley2014). However, identification of monogenic diabetes still has not solved discrepancy in the individual response to glucose-lowering medications in a large number of patients with type 2 diabetes.

In 2007, the first genome-wide association study (GWAS) in type 2 diabetes was reported (Sladek et al., Reference Sladek, Rocheleau, Rung, Dina, Shen, Serre, Boutin, Vincent, Belisle, Hadjadj, Balkau, Heude, Charpentier, Hudson, Montpetit, Pshezhetsky, Prentki, Posner, Balding, Meyre, Polychronakos and Froguel2007). There was much hope that genetics would also represent a breakthrough in understanding of the heterogeneity of type 2 diabetes at that time, however, it turned out that more than 400 genetic variants associated with type 2 diabetes to date could only explain 18% of the risk of diabetes (Mahajan et al., Reference Mahajan, Wessel and Willems2018). Moreover, every individual variant is very modestly associated with the risk of type 2 diabetes, except for the variant in the TCF7L2 gene (Lyssenko et al., Reference Lyssenko, Lupi, Marchetti, del Guerra, Orho-Melander, Almgren, Sjögren, Ling, Eriksson, Lethagen, Mancarella, Berglund, Tuomi, Nilsson, del Prato and Groop2007). Therefore, the genomic information alone has limited value in guiding precision medicine in type 2 diabetes.

Recently, GWAS has also been performed to investigate the genetic risk alleles of type 1 diabetes (Sharp et al., Reference Sharp, Rich, Wood, Jones, Beaumont, Harrison, Schneider, Locke, Tyrrell, Weedon, Hagopian and Oram2019). Fortunately, recent studies have shown that the identified genetic variants could account for the majority of the risk of type 1 diabetes in certain populations, and type 1 diabetes genetic risk scores could well predict the risk of type 1 diabetes with both sensitivity and specificity exceeding 80% in neonatal and African-ancestry populations (Onengut-Gumuscu et al., Reference Onengut-Gumuscu, Chen, Robertson, Bonnie, Farber, Zhu, Oksenberg, Brant, Bridges, Edberg, Kimberly, Gregersen, Rewers, Steck, Black, Dabelea, Pihoker, Atkinson, Wagenknecht, Divers, Bell, Erlich, Concannon and Rich2019; Sharp et al., Reference Sharp, Rich, Wood, Jones, Beaumont, Harrison, Schneider, Locke, Tyrrell, Weedon, Hagopian and Oram2019). The genetics of diabetes mellitus and diabetes complications is well summarized in the literature (Cole and Florez, Reference Cole and Florez2020; Riddle et al., Reference Riddle, Philipson, Rich, Carlsson, Franks, Greeley, Nolan, Pearson, Zeitler and Hattersley2020; Deutsch et al., Reference Deutsch, Ahlqvist and Udler2022).

Epigenetics of diabetes

Obesity and metabolic syndrome is the main risk factor for type 2 diabetes which is caused by a complex inheritance-environment interaction (Wu et al., Reference Wu, Ding, Tanaka and Zhang2014). Epigenetics represents the heritable reversible modifications to the genome associated with environmental factors and clinical phenotypes (BLUEPRINT Consortium, 2016). Epigenetics explores the mechanism in which phenotypes are changed by non-DNA sequence variation, including DNA methylation, histone modifications, non-coding RNAs regulations. Previous studies have shown that blood methylation markers in the TXNIP, ABCG1, PHOSPHO1, SOCS3 and SREBF1 genes were associated with the risk of incident type 2 diabetes (Chambers et al., Reference Chambers, Loh, Lehne, Drong, Kriebel, Motta, Wahl, Elliott, Rota, Scott, Zhang, Tan, Campanella, Chadeau-Hyam, Yengo, Richmond, Adamowicz-Brice, Afzal, Bozaoglu, Mok, Ng, Pattou, Prokisch, Rozario, Tarantini, Abbott, Ala-Korpela, Albetti, Ammerpohl, Bertazzi, Blancher, Caiazzo, Danesh, Gaunt, de Lusignan, Gieger, Illig, Jha, Jones, Jowett, Kangas, Kasturiratne, Kato, Kotea, Kowlessur, Pitkäniemi, Punjabi, Saleheen, Schafmayer, Soininen, Tai, Thorand, Tuomilehto, Wickremasinghe, Kyrtopoulos, Aitman, Herder, Hampe, Cauchi, Relton, Froguel, Soong, Vineis, Jarvelin, Scott, Grallert, Bollati, Elliott, McCarthy and Kooner2015). Since epigenetic patterns have vast plasticity, the epigenetic alteration in type 2 diabetes could be targeted for personalized treatment. The non-coding RNAs, especially the short noncoding RNAs (microRNAs), can regulate the expression of protein-coding genes or epigenetic regulators including DNA methyltransferases, histone deacetylases and polycomb protein coding gene (Bushati and Cohen, Reference Bushati and Cohen2007). MicroRNA changes in vivo were associated with the insulin resistance level in type 2 diabetes (Gallagher et al., Reference Gallagher, Scheele, Keller, Nielsen, Remenyi, Fischer, Roder, Babraj, Wahlestedt, Hutvagner, Pedersen and Timmons2010), and its value as biomarkers for type 2 diabetes has been investigated (de Candia et al., Reference de Candia, Spinetti and Specchia2017). Integrating both genetic and epigenetic risk factors might reflect the inheritance-environment interaction, and provide a promising solution for subclassification of type 2 diabetes.

Recently, the complex association between SARS-CoV-2 Infection (COVID-19) and diabetes has emphasized the environmental factors in promoting diabetes (Shin et al., Reference Shin, Toyoda, Nishitani, Fukuhara, Kita, Otsuki and Shimomura2021; Cao et al., Reference Cao, Baranova, Wei, Wang and Zhang2023). Studies showed that critical COVID-19 and hospitalized COVID-19 subjects had an increased risk of type 2 diabetes, and genetic liability to COVID-19 had a causal effect on type 2 diabetes (Cao et al., Reference Cao, Baranova, Wei, Wang and Zhang2023). Mechanistically, the COVID-19 spike protein physically interacted with GRP78 protein in cell surface of adipose tissue, promoting hyperinsulinemia in adipocytes via XBP1, which may be attributed to the development and progress of diabetes (Shin et al., Reference Shin, Toyoda, Nishitani, Fukuhara, Kita, Otsuki and Shimomura2021).

Stratification of diabetes

Various pathogenic mechanisms and outcomes of disease have been observed in the large range of type 2 diabetes (Philipson, Reference Philipson2020). Thus, the stratification of type 2 diabetes may be relevant in the field of precision medicine for diabetes diagnosis. Cluster analysis based on high-dimensional data, such as electronic medical records or omics data (genomics, proteomics, metabolomics, transcriptomics, lipidomics, etc.), has been utilized to identify subtypes of type 2 diabetes (Li et al., Reference Li, Cheng, Glicksberg, Gottesman, Tamler, Chen, Bottinger and Dudley2015; Udler et al., Reference Udler, Kim, von Grotthuss, Bonàs-Guarch, Cole, Chiou, Boehnke, Laakso, Atzmon, Glaser, Mercader, Gaulton, Flannick, Getz and Florez2018; Wagner et al., Reference Wagner, Heni, Tabák, Machann, Schick, Randrianarisoa, Hrabě de Angelis, Birkenfeld, Stefan, Peter, Häring and Fritsche2021). In 2018, a study of the Swedish population with newly diagnosed diabetes used both hierarchical and k-means clustering to identify five subtypes of adult-onset diabetes, named severe autoimmune diabetes (SAID), severe insulin-deficient diabetes (SIDD), severe insulin-resistant diabetes (SIRD), mild obesity-related diabetes (MOD) and mild age-related diabetes (MARD), based on six clinical variables (autoantibodies, age at diagnosis, BMI, HbA1c, C peptide together with glucose for estimation of insulin secretion, HOMA-B and insulin-sensitivity, HOMA-IS) (Ahlqvist et al., Reference Ahlqvist, Storm, Käräjämäki, Martinell, Dorkhan, Carlsson, Vikman, Prasad, Aly, Almgren, Wessman, Shaat, Spégel, Mulder, Lindholm, Melander, Hansson, Malmqvist, Lernmark, Lahti, Forsén, Tuomi, Rosengren and Groop2018). SAID was characterized by the presence of GAD autoantibodies, low insulin secretion and poor metabolic control, SIDD was characterized by low insulin secretion, poor metabolic control and increased risk of retinopathy, SIRD was characterized by severe insulin resistance, obesity, late onset and markedly increased risk of nephropathy, MOD was characterized by obesity, early onset and good metabolic control, and MARD was characterized by late onset and good metabolic control. This classification of adult-onset diabetes has been most frequently replicated in three independent cohorts from different ethnicities (Philipson, Reference Philipson2020). These subgroups differ in genetic predisposition to diabetes, with increased frequency of HLA rs2854275 variant in SAID and TCF7L2 rs7903146 variant in SIDD, MOD and MARD (Lyssenko et al., Reference Lyssenko, Lupi, Marchetti, Del Guerra, Orho-Melander, Almgren, Sjögren, Ling, Eriksson, Lethagen, Mancarella, Berglund, Tuomi, Nilsson, Del Prato and Groop2007). Both SIRD and MOD patients were obese, but SIRD represented the unhealthy obesity with insulin resistance and non-alcoholic fatty liver disease and MOD represented healthy obesity without insulin resistance (Cohen et al., Reference Cohen, Horton and Hobbs2011). More importantly, the new subclassification of diabetes might guide the personalized treatment. The MOD and MARD patients usually had good metabolic control and disease prognosis, thus, it may be that these cases require less frequent glucose monitoring and could be easily managed with metformin and lifestyle intervention. Several medications with confirmed protective effects on specific vital organs, such as sodium-glucose cotransporter 2 (SGLT2) on cardiovascular and renal outcomes, might be especially suitable for SIRD (Wanner et al., Reference Wanner, Inzucchi, Lachin, Fitchett, von Eynatten, Mattheus, Johansen, Woerle, Broedl and Zinman2016). Thus, the attempts on stratification of diabetes by cluster analysis might be promising for precision diagnosis of diabetes.

More recently, the application of artificial intelligence (AI) technology was able to comprehensively ingest all required parameters in supplied formats (text, image/video, biometric data) for analysis, leading to the prediction of incident diabetes (AUROCs: 0.71–0.87) (Ellahham, Reference Ellahham2020) and risk stratification of diabetic populations (Zou et al., Reference Zou, Qu, Luo, Yin, Ju and Tang2018). Recently, a study using genomic and tabular data to predict type 2 diabetes based on Recurrent Neural Networks has been reported (Srinivasu et al., Reference Srinivasu, Shafi, Krishna, Sujatha, Praveen and Ijaz2022). The results showed that the proposed model could predict future diabetes with fair accuracy, which may be used in real-world scenarios (Srinivasu et al., Reference Srinivasu, Shafi, Krishna, Sujatha, Praveen and Ijaz2022). The use of AI in diabetes management could establish a more accurate and objective stratification of type 2 diabetes based on a broad range of candidate parameters.

Precision prevention

Comprehensive management of diabetes includes diet and exercise interventions, patient education, glucose monitoring and drug treatment. By the time the diabetes is diagnosed, diabetes-related tissue damage has occurred in nearly half of the patients (Ambaby and Chamukuttan, Reference Ambaby and Chamukuttan2008). An early intervention in patients with prediabetes, either with lifestyle interventions or pharmacologic interventions, reduces the risk of incident diabetes and improves long-term outcomes (Haw et al., Reference Haw, Galaviz, Straus, Kowalski, Magee, Weber, Wei, Narayan and Ali2017). However, there has been a big variation among the patients diagnosed with prediabetes in their response to lifestyle or drug intervention (Knowler et al., Reference Knowler, Barrett-Connor, Fowler, Hamman, Lachin, Walker and Nathan2002; Knowler et al., Reference Knowler, Fowler, Hamman, Christophi, Hoffman, Brenneman, Brown-Friday, Goldberg, Venditti and Nathan2009). Those who lost the least weight in the early stages of intervention showed the highest risk of incident diabetes (Delahanty et al., Reference Delahanty, Pan, Jablonski, Aroda, Watson, Bray, Kahn, Florez, Perreault and Franks2014). The reduction of body weight in patients with prediabetes after lifestyle or drug intervention was related to genetic variants (Papandonatos et al., Reference Papandonatos, Pan, Pajewski, Delahanty, Peter, Erar, Ahmad, Harden, Chen, Fontanillas, Wagenknecht, Kahn, Wing, Jablonski, Huggins, Knowler, Florez, McCaffery and Franks2015). For instance, the protective effect of metformin in reducing incidence of diabetes was associated with variation in the SLC47A1 gene in the Diabetes Prevention Program (Jablonski et al., Reference Jablonski, McAteer, de Bakker, Franks, Pollin, Hanson, Saxena, Fowler, Shuldiner, Knowler, Altshuler and Florez2010). Therefore, the patients diagnosed with prediabetes who are unlikely to respond well to lifestyle modification might be better served by other therapeutic treatments, but more studies were required to properly identify this subgroup of prediabetes. Meanwhile, efforts have also been made to prevent the incidence of type 1 diabetes in high-risk children with at least two islet autoantibodies using dietary interventions and/or immune-targeting approaches (Skyler et al., Reference Skyler, Kirchner, Becker, Rewers, Cowie and Casagrande2018). Unfortunately, most previous intervention studies were unable to slow, halt or reverse the destruction of beta cells or delay the progression of type 1 diabetes (Hummel et al., Reference Hummel, Pfluger, Hummel, Bonifacio and Ziegler2011; Knip et al., Reference Knip, Åkerblom, al Taji, Becker, Bruining, Castano, Danne, de Beaufort, Dosch, Dupre, Fraser, Howard, Ilonen, Konrad, Kordonouri, Krischer, Lawson, Ludvigsson, Madacsy, Mahon, Ormisson, Palmer, Pozzilli, Savilahti, Serrano-Rios, Songini, Taback, Vaarala, White, Virtanen and Wasikowa2018).

Precision treatment

Drug treatment was recommended to achieve good glucose control and lower the risk of cardiovascular disease and specific diabetic complications in diabetic patients. Although FDA has approved 10 classes of diabetes medications, each of these medications showed great heterogeneity in therapeutic efficacy, being effective for some patients, but less effective for others, with some even experiencing adverse effects (Dennis et al., Reference Dennis, Shields, Jones, Pearson, Hattersley and Henley2018). Trials of medications on diabetes have recognized that different etiologic processes of diabetes would influence the therapeutic effect of antidiabetic medications recently (Dennis et al., Reference Dennis, Shields, Henley, Jones and Hattersley2019). Reanalysis of the data from the ADOPT and RECORD studies found that the subtype of diabetic patients with insulin resistance responded better to treatment with thiazolidinediones and that older patients responded better to sulfonylureas (Dennis et al., Reference Dennis, Shields, Henley, Jones and Hattersley2019). Similar studies using prospective and primary care data in the UK found that the subgroup of diabetics with insulin resistance, obesity or high triglycerides had reduced initial response to DDP4 inhibitor and more rapid failure of therapy (Dennis et al., Reference Dennis, Shields, Hill, McDonald, Knight, Rodgers, Weedon, Henley, Sattar, Holman, Pearson, Hattersley and Jones2018).

There is also an ethnic difference in the individual response to a specific antidiabetic medication. Previous studies indicated that the therapeutic effect of DDP4 inhibitors is greater in Asians than in other demographic groups. Consistently, a subgroup analysis of the Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TE-COS) showed a greater reduction in blood glucose in East Asians (Davis et al., Reference Davis, Mulder, Lokhnygina, Aschner, Chuang, Raffo Grado, Standl, Peterson and Holman2018), and a recent REWIND study found the protective effect of Dulaglutide on cardiovascular disease is significantly stronger in the Asian Pacific area than other regions of the world (Gerstein et al., Reference Gerstein, Colhoun, Dagenais and Diaz2019). Moreover, the effect of metformin also differed among different ethnic groups, with African Americans having a greater response to metformin than was observed for European Americans (Williams et al., Reference Williams, Padhukasahasram, Ahmedani, Peterson, Wells, González Burchard and Lanfear2014).

The use of genetics in guiding the pharmaceutical treatment of diabetes is an important step toward the precision treatment of diabetes. A typical successful example is the application of genetics in treatment of monogenic diabetes. In that case, a specific single gene mutation is causal for the development of diabetes and targeted treatment can well bypass the etiological defect, such as the use of sulfonylurea in MODY3 caused by the mutation of HNF1A gene (Pearson et al., Reference Pearson, Starkey, Powell, Gribble, Clark and Hattersley2003). However, type 2 diabetes is much more complex than MODY, which is influenced by the complex interaction of hundreds of etiological gene variants and environmental risk factors. Traditionally, genetic studies of drug response in type 2 diabetes have focused on candidate genes known to relate to etiological processes or drug transport or metabolism. Studies have shown that a variant in the SLC22A1 gene encoding the organic cation transporter 1 (OCT1) is involved in the cellular transport of metformin and influenced the individual response to metformin (Shu et al., Reference Shu, Sheardown, Brown, Owen, Zhang, Castro, Ianculescu, Yue, Lo, Burchard, Brett and Giacomini2007). Similarly, a variant in MATE1 (Becker et al., Reference Becker, Visser, van Schaik, Hofman, Uitterlinden and Stricker2009) was also associated with metformin response. Studies also found that KCNJ11/ABCC8 risk variant increases, but TCF7L2 risk variant reduces glycemic response to sulfonylureas (Pearson et al., Reference Pearson, Donnelly, Kimber, Whitley, Doney, McCarthy, Hattersley, Morris and Palmer2007; Feng et al., Reference Feng, Mao, Ren, Xing, Tang, Li, Li, Sun, Yang, Ma, Wang and Xu2008). The PPARG risk variant was associated with reduced glycemic response to thiazolidinediones (Kang et al., Reference Kang, Park, Kim, Kim, Ahn, Cha, Lim, Nam and Lee2005). GWAS in the pharmacogenetics of diabetes made no assumptions about the drug mechanism and metabolism, and have therefore provided novel insights into genetic factors related with response to antidiabetic medication, and successfully identified variants at the ATM and SLC2A2 genes as modulators of individual response to metformin (GoDARTS and UKPDS Diabetes Pharmacogenetics Study Group et al., Reference Zhou and Bellenguez2011; Zhou et al., Reference Zhou, Yee, Seiser, van Leeuwen, Tavendale, Bennett, Groves, Coleman, van der Heijden, Beulens, de Keyser, Zaharenko, Rotroff, Out, Jablonski, Chen, Javorský, Židzik, Levin, Williams, Dujic, Semiz, Kubo, Chien, Maeda, Witte, Wu, Tkáč, Kooy, van Schaik, Stehouwer, Logie, Sutherland, Klovins, Pirags, Hofman, Stricker, Motsinger-Reif, Wagner, Innocenti, Hart, Holman, McCarthy, Hedderson, Palmer, Florez, Giacomini and Pearson2016). GWAS of response to other antidiabetic drugs was still necessary to establish the drug response prediction system based on genetics to guide clinical treatment.

The management of diabetes is a comprehensive approach, and glucose monitoring, patient education and lifestyle intervention also played essential roles in diabetes treatment in addition to diabetes medications. Currently, the use of remote continuous glucose monitoring (CGM) enables monitoring of a complete view of glucose control over 24 h (Battelino and Danne, Reference Battelino, Danne, Bergenstal, Amiel, Beck, Biester, Bosi, Buckingham, Cefalu, Close, Cobelli, Dassau, DeVries, Donaghue, Dovc, Doyle, Garg, Grunberger, Heller, Heinemann, Hirsch, Hovorka, Jia, Kordonouri, Kovatchev, Kowalski, Laffel, Levine, Mayorov, Mathieu, Murphy, Nimri, Nørgaard, Parkin, Renard, Rodbard, Saboo, Schatz, Stoner, Urakami, Weinzimer and Phillip2019), and the in-depth insights of glucose and direct feedback provided by CGM system have efficaciously controlled the blood glucose and reduced the incidence of hypoglycemia (Beck et al., Reference Beck, Riddlesworth, Ruedy, Ahmann, Bergenstal, Haller, Kollman, Kruger, McGill, Polonsky, Toschi, Wolpert and Price2017). The accurate CGM also enabled the personalized lifestyle intervention prescription tailored to each diabetic patient. An AI-based decision support system named the Advisor Pro was recently developed. This system sends the data from CGM to a cloud server and uses AI to determine required insulin doses remotely. Studies showed that insulin doses recommended by the Advisor Pro had no significant difference compared with that given by physicians, suggesting this to be a convenient approach in managing diabetes (Nimri et al., Reference Nimri, Dassau, Segall, Muller, Bratina, Kordonouri, Bello, Biester, Dovc, Tenenbaum, Brener, Šimunović, Sakka, Nevo Shenker, Passone, Rutigliano, Tinti, Bonura, Caiulo, Ruszala, Piccini, Giri, Stein, Rabbone, Bruzzi, Omladič, Steele, Beccuti, Yackobovitch-Gavan, Battelino, Danne, Atlas and Phillip2018; Nimri et al., Reference Nimri, Oron, Muller, Kraljevic, Alonso, Keskinen, Milicic, Oren, Christoforidis, den Brinker, Bozzetto, Bolla, Krcma, Rabini, Tabba, Smith, Vazeou, Maltoni, Giani, Atlas and Phillip2022). With the rapid development in the AI field, it is highly possible that AI will introduce a revolutionary shift in management of diabetes from conventional therapeutic strategies to data-driven precision treatment, based on the combination of individual genetic and glucose monitoring information and decision-making systems based on machine learning.

Lifestyle modifications and first-line medication treatment do not prevent the progressive decline of β-cell mass and function in some patients. Therefore, advanced strategies need to be developed to address this issue. Stem cell therapy for the treatment of diabetes has made great progress in recent years (Furuyama et al., Reference Furuyama, Chera, van Gurp, Oropeza, Ghila, Damond, Vethe, Paulo, Joosten, Berney, Bosco, Dorrell, Grompe, Ræder, Roep, Thorel and Herrera2019; Siehler et al., Reference Siehler, Blochinger, Meier and Lickert2021). Both type 1 diabetes and type 2 diabetes can benefit from stem cell therapy. Studies showed that stem cell therapy increased serum C-peptide and reduced glycosylated hemoglobin (HbA1c) in subjects with type 1 diabetes or type 2 diabetes, but had no significant effect on fasting glucose (Zhang et al., Reference Zhang, Chen, Feng and Cao2020). Additionally, stem cell therapy improved insulin requirements in subjects with type 2 diabetes (Zhang et al., Reference Zhang, Chen, Feng and Cao2020). Different types of stem cells affect the clinical efficacy of therapy for diabetes. Bone marrow mononuclear cells were more effective than mesenchymal stem cells in the treatment of type 1 diabetes, whereas both bone marrow mononuclear cells and mesenchymal stem cells had favorable effects on type 2 diabetes (Zhang et al., Reference Zhang, Chen, Feng and Cao2020). This all suggests that stem cell therapy for the treatment of diabetes is an attractive and potential strategy, but is still facing enormous challenges, for instance, the need for greater diversity in the source of stem cells, and also inconsistency in stem cell preparation, evaluation systems and safety.

Future outlook

The heterogeneity of diabetes creates challenges for the wider applicability of precision medicine in successful treatment of this disease. Although the implementation of precision medicine in diabetes is progressing well, more recent developments within precision medicine may benefit both newly diagnosed patients with diabetes, as well as those exposed to glycemic toxicity for years. Multidisciplinary cooperation will be conducive to further in-depth analysis and understanding of diabetes. Currently, our knowledge on the association of individual genetic background with the pathogenesis and drug response of diabetes is increasing rapidly. The application of AI in the management of diabetes enables the objective and comprehensive analysis and process of a large number of individual multi-dimensional genetic, anthropometric, clinical, biochemical and imaging information, and might provide a solution for precision diagnosis and treatment of diabetes. The concept and tools of precision medicine help to accurately predict, diagnose and treat diabetes and its complications. Although there is still a long way to go, precision medicine will become the driving force for early intervention, early prevention and accurate management of diabetes in the future.

Open peer review

To view the open peer review materials for this article, please visit http://doi.org/10.1017/pcm.2023.12.

Acknowledgement

We have obtained financial supports from National Key Research and Development Program of China (2021YFC27 00403), Shanghai Municipal Science and Technology Commission Foundation (23XD1423300).

Competing interest

All authors declare that there is no duality of interest.

References

Ahlqvist, E, Storm, P, Käräjämäki, A, Martinell, M, Dorkhan, M, Carlsson, A, Vikman, P, Prasad, RB, Aly, DM, Almgren, P, Wessman, Y, Shaat, N, Spégel, P, Mulder, H, Lindholm, E, Melander, O, Hansson, O, Malmqvist, U, Lernmark, Å, Lahti, K, Forsén, T, Tuomi, T, Rosengren, AH and Groop, L (2018) Novel subgroups of adult-onset diabetes and their association with outcomes: A data-driven cluster analysis of six variables. The Lancet Diabetes and Endocrinology 6, 361369.CrossRefGoogle ScholarPubMed
Ambaby, R and Chamukuttan, S (2008) Early diagnosis and prevention of diabetes in developing countries. Reviews in Endocrine & Metabolic Disorders 9(3), 193201.CrossRefGoogle Scholar
Battelino, T, Danne, T, Bergenstal, RM, Amiel, SA, Beck, R, Biester, T, Bosi, E, Buckingham, BA, Cefalu, WT, Close, KL, Cobelli, C, Dassau, E, DeVries, JH, Donaghue, KC, Dovc, K, Doyle, FJ III, Garg, S, Grunberger, G, Heller, S, Heinemann, L, Hirsch, IB, Hovorka, R, Jia, W, Kordonouri, O, Kovatchev, B, Kowalski, A, Laffel, L, Levine, B, Mayorov, A, Mathieu, C, Murphy, HR, Nimri, R, Nørgaard, K, Parkin, CG, Renard, E, Rodbard, D, Saboo, B, Schatz, D, Stoner, K, Urakami, T, Weinzimer, SA and Phillip, M (2019) Clinical targets for continuous glucose monitoring data interpretation: Recommendations from the international consensus on time in range. Diabetes Care 42(8), 15931603CrossRefGoogle ScholarPubMed
Beck, RW, Riddlesworth, TD, Ruedy, KJ, Ahmann, A, Bergenstal, R, Haller, S, Kollman, C, Kruger, D, McGill, JB, Polonsky, W, Toschi, E, Wolpert, H, Price, D and for the DIAMOND Study Group (2017) Effect of continuous glucose monitoring on glycemic control in adults with type 1 diabetes using insulin injections: The DIAMOND randomized clinical trial. JAMA 317(4), 371378.CrossRefGoogle ScholarPubMed
Becker, ML, Visser, LE, van Schaik, RH, Hofman, A, Uitterlinden, AG and Stricker, BH (2009) Genetic variation in the multidrug and toxin extrusion 1 transporter protein influences the glucose-lowering effect of metformin in patients with diabetes: A preliminary study. Diabetes 58, 745749.CrossRefGoogle ScholarPubMed
Bhat, S, Chowta, M, Chowta, N, Shastry, R and Kamath, P (2021) The proportion of type 2 diabetic patients achieving treatment goals and the survey of Patients’ attitude towards insulin initiation in patients with inadequate glycaemic control with oral anti-diabetic drugs. Current Diabetes Reviews 17(5), e110620182719.CrossRefGoogle ScholarPubMed
The BLUEPRINT consortium (2016) Quantitative comparison of DNA methylation assays for biomarker development and clinical applications. Nature Biotechnology 34, 726737.CrossRefGoogle Scholar
Bushati, N and Cohen, SM (2007) microRNA functions. Annual Review of Cell and Developmental Biology 23, 175205.CrossRefGoogle ScholarPubMed
Cao, H, Baranova, A, Wei, X, Wang, C and Zhang, F (2023) Bidirectional causal associations between type 2 diabetes and COVID-19. Journal of Medical Virology 95(1), e28100.CrossRefGoogle ScholarPubMed
Chambers, JC, Loh, M, Lehne, B, Drong, A, Kriebel, J, Motta, V, Wahl, S, Elliott, HR, Rota, F, Scott, WR, Zhang, W, Tan, ST, Campanella, G, Chadeau-Hyam, M, Yengo, L, Richmond, RC, Adamowicz-Brice, M, Afzal, U, Bozaoglu, K, Mok, ZY, Ng, HK, Pattou, F, Prokisch, H, Rozario, MA, Tarantini, L, Abbott, J, Ala-Korpela, M, Albetti, B, Ammerpohl, O, Bertazzi, PA, Blancher, C, Caiazzo, R, Danesh, J, Gaunt, TR, de Lusignan, S, Gieger, C, Illig, T, Jha, S, Jones, S, Jowett, J, Kangas, AJ, Kasturiratne, A, Kato, N, Kotea, N, Kowlessur, S, Pitkäniemi, J, Punjabi, P, Saleheen, D, Schafmayer, C, Soininen, P, Tai, ES, Thorand, B, Tuomilehto, J, Wickremasinghe, AR, Kyrtopoulos, SA, Aitman, TJ, Herder, C, Hampe, J, Cauchi, S, Relton, CL, Froguel, P, Soong, R, Vineis, P, Jarvelin, MR, Scott, J, Grallert, H, Bollati, V, Elliott, P, McCarthy, MI and Kooner, JS (2015) Epigenome-wide association of DNA methylation markers in peripheral blood from Indian Asians and Europeans with incident type 2 diabetes: A nested case-control study. The Lancet Diabetes and Endocrinology 3, 526534.CrossRefGoogle ScholarPubMed
Cohen, JC, Horton, JD and Hobbs, HH (2011) Human fatty liver disease: Old questions and new insights. Science 332, 15191523.CrossRefGoogle ScholarPubMed
Cole, JB and Florez, JC (2020) Genetics of diabetes mellitus and diabetes complications. Nature Reviews. Nephrology 16(7): 377390.CrossRefGoogle ScholarPubMed
Cook, MN, Girman, CJ, Stein, PP and Alexander, CM (2007) Initial monotherapy with either metformin or sulphonylureas often fails to achieve ormaintain current glycaemic goals in patients with type 2 diabetes in UK primary care. Diabetic Medicine 24(4), 350358.CrossRefGoogle ScholarPubMed
Davis, TME, Mulder, H, Lokhnygina, Y, Aschner, P, Chuang, LM, Raffo Grado, CA, Standl, E, Peterson, ED, Holman, RR and for the TECOS Study Group (2018) TECOS study group. Effect of race on the glycaemic response to sitagliptin: Insights from the trial evaluating cardiovascular outcomes with Sitagliptin (TECOS). Diabetes, Obesity & Metabolism 20, 14271434.CrossRefGoogle Scholar
de Candia, P, Spinetti, G, Specchia, C et al. (2017) A unique plasma microRNA profile defines type 2 diabetes progression. PLoS One 12, e0188980.CrossRefGoogle ScholarPubMed
Delahanty, LM, Pan, Q, Jablonski, KA, Aroda, VR, Watson, KE, Bray, GA, Kahn, SE, Florez, JC, Perreault, L, Franks, PW and for the Diabetes Prevention Program Research Group (2014) Diabetes prevention program research group. Effects of weight loss, weight cycling, and weight loss maintenance on diabetes incidence and change in cardiometabolic traits in the diabetes prevention program. Diabetes Care 37, 27382745.CrossRefGoogle Scholar
Dennis, JM, Shields, BM, Henley, WE, Jones, AG and Hattersley, AT (2019) Disease progression and treatment response in data-driven subgroups of type 2 diabetes compared with models based on simple clinical features: An analysis using clinical trial data. The Lancet Diabetes and Endocrinology 7, 442451.CrossRefGoogle ScholarPubMed
Dennis, JM, Shields, BM, Hill, AV, McDonald, T, Knight, BA, Rodgers, LR, Weedon, MN, Henley, WE, Sattar, N, Holman, RR, Pearson, ER, Hattersley, AT, Jones, AG and MASTERMIND Consortium (2018) Precision medicine in type 2 diabetes: Clinical markers of insulin resistance are associated with altered short- and long-term glycemic response to DPP-4 inhibitor therapy. Diabetes Care 41, 705712.CrossRefGoogle Scholar
Dennis, JM, Shields, BM, Jones, AG, Pearson, ER, Hattersley, AT, Henley, WE and MASTERMIND Consortium (2018) Evaluating associations between the benefits and risks of drug therapy in type 2 diabetes: A joint modeling approach. Clinical Epidemiology 10, 18691877.CrossRefGoogle ScholarPubMed
Deutsch, AJ, Ahlqvist, E and Udler, MS (2022) Phenotypic and genetic classification of diabetes. Diabetologia 65(11), 17581769.CrossRefGoogle ScholarPubMed
Ellahham, S (2020) Artificial intelligence: The future for diabetes care. The American Journal of Medicine 133(8), 895900.CrossRefGoogle ScholarPubMed
Feng, Y, Mao, G, Ren, X, Xing, H, Tang, G, Li, Q, Li, X, Sun, L, Yang, J, Ma, W, Wang, X and Xu, X (2008) Ser1369Ala variant in sulfonylurea receptor gene ABCC8 is associated with antidiabetic efficacy of gliclazide in Chinese type 2 diabetic patients. Diabetes Care 31, 19391944.CrossRefGoogle ScholarPubMed
Furuyama, K, Chera, S, van Gurp, L, Oropeza, D, Ghila, L, Damond, N, Vethe, H, Paulo, JA, Joosten, AM, Berney, T, Bosco, D, Dorrell, C, Grompe, M, Ræder, H, Roep, BO, Thorel, F and Herrera, PL (2019) Diabetes relief in mice by glucose-sensing insulin-secreting human alpha-cells. Nature 567(7746), 4348.CrossRefGoogle ScholarPubMed
Gallagher, IJ, Scheele, C, Keller, P, Nielsen, AR, Remenyi, J, Fischer, CP, Roder, K, Babraj, J, Wahlestedt, C, Hutvagner, G, Pedersen, BK and Timmons, JA (2010) Integration of microRNA changes in vivo identifies novel molecular features of muscle insulin resistance in type 2 diabetes. Genome Medicine 2(2), 9.CrossRefGoogle ScholarPubMed
GBD 2017 Disease and Injury Incidence and Prevalence Collaborators (2018) Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 392(10159), 17891858.CrossRefGoogle Scholar
Gerstein, HC, Colhoun, HM, Dagenais, GR, Diaz, R, et al. (2019) Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): A double-blind, randomised placebo-controlled trial. Lancet 394(10193), 121130.CrossRefGoogle ScholarPubMed
The GoDARTS and UKPDS Diabetes Pharmacogenetics Study Group, The Wellcome Trust Case Control Consortium 2, Zhou, K, Bellenguez, C et al. (2011 ) Common variants near ATM are associated with glycemic response to metformin in type 2 diabetes. Nature Genetics 43, 117120.Google ScholarPubMed
Haw, JS, Galaviz, KI, Straus, AN, Kowalski, AJ, Magee, MJ, Weber, MB, Wei, J, Narayan, KMV and Ali, MK (2017) Longterm sustainability of diabetes prevention approaches: A systematic review and meta-analysis of randomized clinical trials. JAMA Internal Medicine 177, 18081817.CrossRefGoogle ScholarPubMed
Himsworth, HP and Kerr, RB (1939) Insulin-sensitive and insulininsensitive types of diabetes mellitus. Clinical Science 4, 119152.Google Scholar
Hummel, S, Pfluger, M, Hummel, M, Bonifacio, E and Ziegler, AG (2011) Primary dietary intervention study to reduce the risk of islet autoimmunity in children at increased risk for type 1 diabetes: The BABYDIET study. Diabetes Care 34:13011305.CrossRefGoogle ScholarPubMed
IDF (2021) IDF Diabetes Atlas, 10th Edn.Google Scholar
Ingelfinger, JR and Jarcho, JA (2017) Increase in the incidence of diabetes and its implications. The New England Journal of Medicine 376(15), 14731474.CrossRefGoogle ScholarPubMed
Jablonski, KA, McAteer, JB, de Bakker, PI, Franks, PW, Pollin, TI, Hanson, RL, Saxena, R, Fowler, S, Shuldiner, AR, Knowler, WC, Altshuler, D, Florez, JC and for the Diabetes Prevention Program Research Group (2010) Common variants in 40 genes assessed for diabetes incidence and response to metformin and lifestyle intervention in the diabetes prevention program. Diabetes 59, 26722681.CrossRefGoogle ScholarPubMed
Kahn, SE, Haffner, SM, Heise, MA, Herman, WH, Holman, RR, Jones, NP, Kravitz, BG, Lachin, JM, O’Neill, MC, Zinman, B and Viberti, G (2006) Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. The New England Journal of Medicine 355(23), 24272443.CrossRefGoogle ScholarPubMed
Kang, E, Park, S, Kim, H, Kim, C, Ahn, C, Cha, B, Lim, S, Nam, C and Lee, H (2005) Effects of Pro12Ala polymorphism of peroxisome proliferator-activated receptor gamma2 gene on rosiglitazone response in type 2 diabetes. Clinical Pharmacology and Therapeutics 78, 202208.CrossRefGoogle ScholarPubMed
Khunti, K and Millar-Jones, D (2017) Clinical inertia to insulin initiation and intensification in the UK: A focused literature review. Primary Care Diabetes 11(1), 312CrossRefGoogle Scholar
Kim, YG, Hahn, S, Oh, TJ, Kwak, SH, Park, KS and Cho, YM (2013) Differences in the glucose-lowering efficacy of dipeptidyl peptidase-4 inhibitors between Asians and non-Asians: A systematic review and meta-analysis. Diabetologia 56, 696708.CrossRefGoogle ScholarPubMed
Knip, M, Åkerblom, HK, al Taji, E, Becker, D, Bruining, J, Castano, L, Danne, T, de Beaufort, C, Dosch, HM, Dupre, J, Fraser, WD, Howard, N, Ilonen, J, Konrad, D, Kordonouri, O, Krischer, JP, Lawson, ML, Ludvigsson, J, Madacsy, L, Mahon, JL, Ormisson, A, Palmer, JP, Pozzilli, P, Savilahti, E, Serrano-Rios, M, Songini, M, Taback, S, Vaarala, O, White, NH, Virtanen, SM, Wasikowa, R and Writing Group for the TRIGR Study Group (2018) Effect of hydrolyzed infant formula vs conventional formula on risk of type 1 diabetes: The TRIGR randomized clinical trial. JAMA 319, 3848.Google ScholarPubMed
Knowler, WC, Barrett-Connor, E, Fowler, SE, Hamman, RF, Lachin, JM, Walker, EA, Nathan, DM and Diabetes Prevention Program Research Group (2002) Diabetes prevention program research group. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. The New England Journal of Medicine 346, 393403.Google ScholarPubMed
Knowler, WC, Fowler, SE, Hamman, RF, Christophi, CA, Hoffman, HJ, Brenneman, AT, Brown-Friday, JO, Goldberg, R, Venditti, E, Nathan, DM and Diabetes Prevention Program Research Group (2009) Diabetes prevention program research group. 10-year follow-up of diabetes incidence and weight loss in the diabetes prevention program outcomes study. Lancet 374, 16771686.Google Scholar
Li, L, Cheng, WY, Glicksberg, BS, Gottesman, O, Tamler, R, Chen, R, Bottinger, EP and Dudley, JT (2015) Identification of type 2 diabetes subgroups through topological analysis of patient similarity. Science Translational Medicine 7(311), 311ra174.CrossRefGoogle ScholarPubMed
Lyssenko, V, Lupi, R, Marchetti, P, del Guerra, S, Orho-Melander, M, Almgren, P, Sjögren, M, Ling, C, Eriksson, KF, Lethagen, L, Mancarella, R, Berglund, G, Tuomi, T, Nilsson, P, del Prato, S and Groop, L (2007) Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. The Journal of Clinical Investigation 117, 21552163.CrossRefGoogle ScholarPubMed
Lyssenko, V, Lupi, R, Marchetti, P, Del Guerra, S, Orho-Melander, M, Almgren, P, Sjögren, M, Ling, C, Eriksson, K-F, Lethagen, A-S, Mancarella, R, Berglund, G, Tuomi, T, Nilsson, P, Del Prato, S and Groop, L (2007) Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. The Journal of Clinical Investigation 117, 21552163.CrossRefGoogle ScholarPubMed
Mahajan, A, Wessel, J, Willems, SM, et al. (2018) Refining the accuracy of validated target identification through coding variant finemapping in type 2 diabetes. Nature Genetics 50, 559571.CrossRefGoogle ScholarPubMed
National Diabetes Data Group (1979) Classification and diagnosis of diabetes mellitus and other categories of glucose intolerance. Diabetes 28, 10391057.CrossRefGoogle Scholar
Nimri, R, Dassau, E, Segall, T, Muller, I, Bratina, N, Kordonouri, O, Bello, R, Biester, T, Dovc, K, Tenenbaum, A, Brener, A, Šimunović, M, Sakka, SD, Nevo Shenker, M, Passone, CGB, Rutigliano, I, Tinti, D, Bonura, C, Caiulo, S, Ruszala, A, Piccini, B, Giri, D, Stein, R, Rabbone, I, Bruzzi, P, Omladič, , Steele, C, Beccuti, G, Yackobovitch-Gavan, M, Battelino, T, Danne, T, Atlas, E and Phillip, M (2018) Adjusting insulin doses in patients with type 1 diabetes who use insulin pump and continuous glucose monitoring: Variations among countries and physicians. Diabetes, Obesity & Metabolism 20(10), 24582466.CrossRefGoogle ScholarPubMed
Nimri, R, Oron, T, Muller, I, Kraljevic, I, Alonso, MM, Keskinen, P, Milicic, T, Oren, A, Christoforidis, A, den Brinker, M, Bozzetto, L, Bolla, AM, Krcma, M, Rabini, RA, Tabba, S, Smith, L, Vazeou, A, Maltoni, G, Giani, E, Atlas, E and Phillip, M (2022) Adjustment of insulin pump settings in type 1 diabetes management: Advisor pro device compared to Physicians’ recommendations. Journal of Diabetes Science and Technology 16(2), 364372.CrossRefGoogle ScholarPubMed
Onengut-Gumuscu, S, Chen, WM, Robertson, CC, Bonnie, JK, Farber, E, Zhu, Z, Oksenberg, JR, Brant, SR, Bridges, SL Jr, Edberg, JC, Kimberly, RP, Gregersen, PK, Rewers, MJ, Steck, AK, Black, MH, Dabelea, D, Pihoker, C, Atkinson, MA, Wagenknecht, LE, Divers, J, Bell, RA, SEARCH for Diabetes in Youth, Type 1 Diabetes Genetics Consortium, Erlich, HA, Concannon, P and Rich, SS (2019) Type 1 diabetes risk in African-ancestry participants and utility of an ancestry-specific genetic risk score. Diabetes Care 42, 406415.CrossRefGoogle ScholarPubMed
Papandonatos, GD, Pan, Q, Pajewski, NM, Delahanty, LM, Peter, I, Erar, B, Ahmad, S, Harden, M, Chen, L, Fontanillas, P, GIANT Consortium, Wagenknecht, LE, Kahn, SE, Wing, RR, Jablonski, KA, Huggins, GS, Knowler, WC, Florez, JC, McCaffery, JM, Franks, PW and for the Diabetes Prevention Program and the Look AHEAD Research Groups (2015) GIANT consortium; diabetes prevention program and the look AHEAD research groups. Genetic predisposition to weight loss and regain with lifestyle intervention: Analyses from the diabetes prevention program and the look AHEAD randomized controlled trials. Diabetes 64, 43124321.CrossRefGoogle Scholar
Pearson, ER, Donnelly, LA, Kimber, C, Whitley, A, Doney, ASF, McCarthy, MI, Hattersley, AT, Morris, AD and Palmer, CNA (2007) Variation in TCF7L2 influences therapeutic response to sulfonylureas: A GoDARTs study. Diabetes 56, 21782182CrossRefGoogle ScholarPubMed
Pearson, ER, Starkey, BJ, Powell, RJ, Gribble, FM, Clark, PM, Hattersley, AT (2003) Genetic cause of hyperglycaemia and response to treatment in diabetes. Lancet 362, 12751281.CrossRefGoogle ScholarPubMed
Philipson, LH (2020) Harnessing heterogeneity in type 2 diabetes mellitus. Nature Reviews. Endocrinology 16(2), 7980.CrossRefGoogle ScholarPubMed
Rao Kondapally Seshasai, S, Kaptoge, S, Thompson, A, di Angelantonio, E, Gao, P, Sarwar, N, Whincup, PH, Mukamal, KJ, Gillum, RF, Holme, I, Njølstad, I, Fletcher, A, Nilsson, P, Lewington, S, Collins, R, Gudnason, V, Thompson, SG, Sattar, N, Selvin, E, Hu, FB, Danesh, J and Emerging Risk Factors Collaboration (2011) Diabetes mellitus, fasting glucose, and risk of cause-specific death. The New England Journal of Medicine 364, 829841.Google ScholarPubMed
Riddle, MC, Philipson, LH, Rich, SS, Carlsson, A, Franks, PW, Greeley, SAW, Nolan, JJ, Pearson, ER, Zeitler, PS and Hattersley, AT (2020) Monogenic diabetes: From genetic insights to population-based precision in care. Reflections from a diabetes care editors’ expert forum. Diabetes reviews (Alexandria, Va) Diabetes reviews 43(12), 31173128.Google ScholarPubMed
Sharp, SA, Rich, SS, Wood, AR, Jones, SE, Beaumont, RN, Harrison, JW, Schneider, DA, Locke, JM, Tyrrell, J, Weedon, MN, Hagopian, WA and Oram, RA (2019) Development and standardization of an improved type 1 diabetes genetic risk score for use in newborn screening and incident diagnosis. Diabetes Care 42(2), 200207.CrossRefGoogle ScholarPubMed
Shin, J, Toyoda, S, Nishitani, S, Fukuhara, A, Kita, S, Otsuki, M and Shimomura, I Possible involvement of adipose tissue in patients with older age, obesity, and diabetes with SARS-CoV-2 infection (COVID-19) via GRP78 (BIP/HSPA5): Significance of hyperinsulinemia management in COVID-19. Diabetes 70(12), 27452755.CrossRefGoogle Scholar
Shu, Y, Sheardown, SA, Brown, C, Owen, RP, Zhang, S, Castro, RA, Ianculescu, AG, Yue, L, Lo, JC, Burchard, EG, Brett, CM and Giacomini, KM (2007) Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. The Journal of Clinical Investigation 117, 14221431.CrossRefGoogle ScholarPubMed
Siehler, J, Blochinger, AK, Meier, M and Lickert, H (2021) Engineering islets from stem cells for advanced therapies of diabetes. Nature Reviews. Drug Discovery 20(12), 920940.CrossRefGoogle ScholarPubMed
Skyler, JS, Kirchner, JP, Becker, D, Rewers, M (2018) Chapter 37: Prevention of Type 1 Diabetes in Diabetes in America, 3rd Edn., Cowie, CC, Casagrande, SS, et al., Eds. Bethesda, MD: National Institutes of Health.Google Scholar
Sladek, R, Rocheleau, G, Rung, J, Dina, C, Shen, L, Serre, D, Boutin, P, Vincent, D, Belisle, A, Hadjadj, S, Balkau, B, Heude, B, Charpentier, G, Hudson, TJ, Montpetit, A, Pshezhetsky, AV, Prentki, M, Posner, BI, Balding, DJ, Meyre, D, Polychronakos, C and Froguel, P (2007) A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 445, 881885.CrossRefGoogle ScholarPubMed
Srinivasu, PN, Shafi, J, Krishna, TB, Sujatha, CN, Praveen, SP and Ijaz, MF (2022) Using recurrent neural networks for predicting Type-2 diabetes from genomic and tabular data. Diagnostics (Basel) 12(12).Google ScholarPubMed
Steele, AM, Shields, BM, Wensley, KJ, Colclough, K, Ellard, S and Hattersley, AT (2014) Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA 311: 279–86.CrossRefGoogle ScholarPubMed
Tattersall, RB and Fajans, SS (1975) A difference between the inheritance of classical juvenile-onset and maturity-onset type diabetes of young people. Diabetes 24, 4453.CrossRefGoogle ScholarPubMed
Udler, MS, Kim, J, von Grotthuss, M, Bonàs-Guarch, S, Cole, JB, Chiou, J, Christopher D. Anderson on behalf of METASTROKE and the ISGC, Boehnke, M, Laakso, M, Atzmon, G, Glaser, B, Mercader, JM, Gaulton, K, Flannick, J, Getz, G and Florez, JC (2018) Type 2 diabetes genetic loci informed by multi-trait associations point to disease mechanisms and subtypes: A soft clustering analysis. PLoS Medicine 15, e1002654.CrossRefGoogle ScholarPubMed
Udler, MS, Kim, J, von Grotthuss, M, Bonàs-Guarch, S, Cole, JB, Chiou, J, Christopher D. Anderson on behalf of METASTROKE and the ISGC, Boehnke, M, Laakso, M, Atzmon, G, Glaser, B, Mercader, JM, Gaulton, K, Flannick, J, Getz, G and Florez, JC (2018) Type 2 diabetes genetic loci informed by multi-trait associations point to disease mechanisms and subtypes: A soft clustering analysis. PLoS Medicine 15(9), e1002654.CrossRefGoogle ScholarPubMed
Velásquez-Mieyer, PA, Cowan, PA, Pérez-Faustinelli, S, Nieto-Martínez, R, Villegas-Barreto, C, Tolley, EA, Lustig, RH and Alpert, BS (2008) Racial disparity in glucagon-like peptide 1 and inflammation markers among severely obese adolescents. Diabetes Care 31(4), 770775.CrossRefGoogle ScholarPubMed
Wagner, R, Heni, M, Tabák, AG, Machann, J, Schick, F, Randrianarisoa, E, Hrabě de Angelis, M, Birkenfeld, AL, Stefan, N, Peter, A, Häring, HU and Fritsche, A (2021) Pathophysiology-based subphenotyping of individuals at elevated risk for type 2 diabetes. Nature Medicine 27(1), 4957.CrossRefGoogle ScholarPubMed
Wanner, C, Inzucchi, SE, Lachin, JM, Fitchett, D, von Eynatten, M, Mattheus, M, Johansen, OE, Woerle, HJ, Broedl, UC and Zinman, B (2016) Empagliflozin and progression of kidney disease in type 2 diabetes. The New England Journal of Medicine 375, 323334.CrossRefGoogle ScholarPubMed
Williams, LK, Padhukasahasram, B, Ahmedani, BK, Peterson, EL, Wells, KE, González Burchard, E and Lanfear, DE (2014) Differing effects of metformin on glycemic control by race-ethnicity. The Journal of Clinical Endocrinology and Metabolism 99, 31603168CrossRefGoogle ScholarPubMed
Wu, Y, Ding, Y, Tanaka, Y and Zhang, W. (2014) Risk factors contributing to type 2 diabetes and recent advances in the treatment and prevention. International Journal of Medical Sciences 11(11), 11851200.CrossRefGoogle ScholarPubMed
Zhang, Y, Chen, W, Feng, B and Cao, H (2020) The clinical efficacy and safety of stem cell therapy for diabetes mellitus: A systematic review and meta-analysis. Aging and Disease 11(1), 141153.Google ScholarPubMed
MetGen Investigators, DPP Investigators, ACCORD Investigators, Zhou, K, Yee, SW, Seiser, EL, van Leeuwen, N, Tavendale, R, Bennett, AJ, Groves, CJ, Coleman, RL, van der Heijden, AA, Beulens, JW, de Keyser, CE, Zaharenko, L, Rotroff, DM, Out, M, Jablonski, KA, Chen, L, Javorský, M, Židzik, J, Levin, AM, Williams, LK, Dujic, T, Semiz, S, Kubo, M, Chien, HC, Maeda, S, Witte, JS, Wu, L, Tkáč, I, Kooy, A, van Schaik, RHN, Stehouwer, CDA, Logie, L, Sutherland, C, Klovins, J, Pirags, V, Hofman, A, Stricker, BH, Motsinger-Reif, AA, Wagner, MJ, Innocenti, F, Hart, LM’, Holman, RR, McCarthy, MI, Hedderson, MM, Palmer, CNA, Florez, JC, Giacomini, KM and Pearson, ER (2016) Variation in the glucose transporter gene SLC2A2 is associated with glycemic response to metformin. Nature Genetics 48, 10551059.CrossRefGoogle ScholarPubMed
Zou, Q, Qu, K, Luo, Y, Yin, D, Ju, Y and Tang, H (2018) Predicting diabetes mellitus with machine learning techniques. Frontiers in Genetics 9, 515.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. The precision medicine in diabetes management.

Author comment: Updates of precision medicine in type 2 diabetes — R0/PR1

Comments

Oct 7, 2022

Editorial Office

Cambridge Prisms: Precision Medicine

Dear Editors,

We are writing to submit the invited review entitled “Updates of precision medicine in type 2 diabetes” by Mingfeng Xia and Xiaoying Li, which we hope you will find suitable for publication in Cambridge Prisms: Precision Medicine.

Diabetes mellitus is prevalent worldwide. Despite of the successful development of glucose-lowering drugs, such as GLP-1 agonists and SGLT2 inhibitors recently, the proportion of patients achieving satisfactory glucose control has not risen as expected. Diabetes is undoubtedly more heterogeneous than the conventional subclassification, and obviously a one-size-fits-all strategy is not suitable for all diabetic patients. In this review, we provide an overview of precision medicine in diabetes, focusing on the genetics and epigenetics, clinical stratification and personalized prevention, treatment of this disease and its related complications.

Yours sincerely,

Xiaoying Li, MD, PhD

Professor and Head

Department of Endocrinology and Metabolism

Zhongshan Hospital, Fudan University

Shanghai, 200032, China

Review: Updates of precision medicine in type 2 diabetes — R0/PR2

Conflict of interest statement

The article by Mingfeng Xia and Xiaoying Li describes that despite of the successful development of glucose-lowering drugs, such as GLP-1 receptor agonists and iSGLT-2 recently, the proportion of patients achieving satisfactory glucose control has not risen as expected. They highlight the importance of the heterogeneity of diabetes determines that a one-size-fits-all strategy. The recent progress in genetics and epigenetics of diabetes has gradually unveiled the mechanisms underlying the heterogeneity of diabetes, and cluster analysis has shown promising results in the substratification of the type 2 diabetes, the major form of diabetes. They conclude that the rapid development of the sophisticated glucose monitoring and artificial intelligence technologies further enabled comprehensive consideration of the complex individual genetic and clinical information, and might ultimately realize precision diagnosis and treatment in diabetic patients. I have the following comments: - The introduction should be expanded with more information and more citations. A figure in this section is necessary. - The use of genetics in guiding the pharmaceutical treatment of diabetes is an important step towards the precision treatment of diabetes. This section has to be expanded and make a table with both in vitro and in vivo studies. - The future outlook section has to be expanded and improved. - The authors have to improve all sections of the article with more and more updated information.

Comments

Comments to Author: The article by Mingfeng Xia and Xiaoying Li describes that despite of the successful development of glucose-lowering drugs, such as GLP-1 receptor agonists and iSGLT-2 recently, the proportion of patients achieving satisfactory glucose control has not risen as expected. They highlight the importance of the heterogeneity of diabetes determines that a one-size-fits-all strategy. The recent progress in genetics and epigenetics of diabetes has gradually unveiled the mechanisms underlying the heterogeneity of diabetes, and cluster analysis has shown promising results in the substratification of the type 2 diabetes, the major form of diabetes. They conclude that the rapid development of the sophisticated glucose monitoring and artificial intelligence technologies further enabled comprehensive consideration of the complex individual genetic and clinical information, and might ultimately realize precision diagnosis and treatment in diabetic patients.

I have the following comments:

- The introduction should be expanded with more information and more citations. A figure in this section is necessary.

- The use of genetics in guiding the pharmaceutical treatment of diabetes is an important step towards the precision treatment of diabetes. This section has to be expanded and make a table with both in vitro and in vivo studies.

- The future outlook section has to be expanded and improved.

- The authors have to improve all sections of the article with more and more updated information.

Review: Updates of precision medicine in type 2 diabetes — R0/PR3

Conflict of interest statement

Reviewer declares none.

Comments

Comments to Author: Article needs to be revised completely as it is poorly written and difficult to understand.

Precision Medicine in Diabetes Review

10/29/22

Page/Line Comment

Abstract Suggest changing the term DIABETIC PATIENTS to people with diabetes

Abstract Change this sentence and diabetes is undoubtedly more

heterogeneous than the conventional subclassification, such as type 1,

type 2, monogenic and gestational diabetes.

Place a period after people with diabetes. Then edit the next sentence to: Diabetes is undoubtedly more

heterogeneous than the conventional subclassification, such as type 1,

type 2, monogenic and gestational diabetes.

Abstract. Last sentence. Change in diabetic patients to people with diabetes.

Introduction With the dramatic

escalation in obesity, diabetes has become a fast increased disease worldwide. Actually authors should consider discussing other causes progression from normal glucose regulation to diabetes other than obesity. For example, the COVID pandemic has increased the risk of developing both type 1 and type 2 patients. COVID also increases diabetes risk in YOUNGER patients. (https://www.cdc.gov/mmwr/volumes/71/wr/mm7102e2.htm)

Xie, Y. & Al-Aly, Z. Lancet Diabetes Endocrinol. https://doi.org/10.1016/S2213-8587(22)00044-4 (2022

General note Change all phrases: diabetic patients, to ‘PATIENTS WITH DIABETES.”

Introduction This phrase needs to be edited: However, if blood glucose is well controlled, the clinical outcomes of diabetic patients could be tremendously improved 5. Change to: Early and intensive management of diabetes targeting recommended glycemic and metabolic targets, can reduce long-term diabetes complications. (Khunti K, et al. Prim Care Diabetes. 2017;11(1):3-12)

Page 5 Suggested edit: Moreover, it has been well recognized that not all diabetic patients respond to the

antidiabetic treatment in the same way. Change to: Patients with diabetes do not respond to glucose lowering therapies equally. Thus, pharmacologic intervention should be individualized based on factors such as duration of diabetes, presence of existing comorbidities, expected duration of life, weight, age, family history of diabetes related complications, and ability to cover the cost of prescribed medications and technology.

6 Epigenetics of diabetes. Authors should again consider adding a statement about the effect of COVID infections on activating genes which promote the expression of diabetes.

7 This statement makes no sense. Please revise: Type 2 diabetes is an exclusion diagnosis, and about 90% of diabetes was type 2 diabetes29

7 What is this? Omics data

7 Section about stratification of diabetes.

Look, Im sorry. I read this section 5 times and was totally confused about what the authors are saying or suggesting. And, this is actually a topic that I typically understand.

9 Section precision management:

diabetic drugs should be changed to pharmacologic interventions

9 Change prediabetic patients to patients diagnosed with prediabetes.

9 Do not start a sentence with a preposition: It is not until recently that trials of

medications on diabetes recognized that different etiologic processes of diabetes would

influence the therapeutic effect of antidiabetic medications51

12 Edit this sentence: Although the implementation of precision medicine in diabetes is on the way ahead. The future application of precision medicine may benefit newly diagnosed patients with diabetes, as well as those exposed to glycemic toxicity for years.

Recommendation: Updates of precision medicine in type 2 diabetes — R0/PR4

Comments

Comments to Author: This review has good quality, mainly for individuals with little expertise in the area or as an overview for beginners. There is a large amount of dense studies in the literature on about genomics of diabetes, and perhaps this is the main challenge for the authors. Perhaps a better delimitation of the study object could be considered to open writing space for an adequate deepening of the results obtained in each study cited in the text. Data on conclusions about each study could be more explained, sometimes it was too superficial. For example, addressing AI and genomics could have been a clipping error. If the authors propose a general review on the topic, I still believe that other relevant topics in the area were missing, as suggested below.

1. In the introduction, authors have cited that DM affects 141 million adults in China. The following sentence in the same paragraph describes “diabetes is also the ninth leading cause of population death”. Is this position in China, or globally? Please remove the numbers about your country from the text, as our journal is aimed at research groups worldwide. Your article is not exclusively about China.

2. In the section, Heterogeneity of Diabetes. The sentence: “However, it was not until the introduction of genetics in the recent 20 years, that the molecular mechanisms of the monogenic diabetes were uncovered.” Several studies on medical genetics have been performed about Mody before 2002 (for example Yagamata et al 1996). I think that the word “genetics” should be changed to “genomic medicine”.

3. In the section, Heterogeneity of Diabetes. In the last sentence, the word “crucial” should be changed to “might be considered”, since the genomic stratification of DM-patients is still a promise in personalized medicine. There is no robust recommendation for this medical approach into clinical practice to date, such as you have described in the following sections.

4. In the last sentence of the section Genetics of Diabetes you have described the value of genetic risk scores for type 1 DM. References 21 and 22 report findings in neonatal and african-descendents populations specifically. You should describe in the text that these scores were performed in specific populations, neonatal and african-ancestry populations with more details about these cohorts. Genetic risk scores are dependent on the genetic variation identified among the different populations.

5. Again, in the section Stratification of Diabetes, the word “crucial” in the text should be changed because it is very emphatic. “Thus, the stratification of type 2 diabetes is crucial in the field of precision medicine for diabetes diagnosis”. Terms such as “may be relevant”, or “may be considered” is preferably in this incipient medical context.

6. There is no reference supporting the sentence “The MOD and MARD patients usually had good metabolic control and disease prognosis, thus required less frequent glucose monitoring and could be easily managed with metformin and lifestyle intervention”. Please cite the reference(s) or adjust the text, for example; “thus, in these cases it might require less frequent…” and cite your reference, or “thus, in our opinion, in these cases it might require less frequent…” although this last suggestion maybe would be inappropriate.

7. In the last paragraph of the section Stratification of Diabetes, is there some article about AI and diabetes taking into consideration genomic variants? If yes, you should cite these articles, describing if the results were consistent or not. If there is no publication on it, please report that there is no evidence to date.

8. Several continuous glucose monitoring devices have been approved by FDA, not only Advisor Pro as cited in the last section of your revision. Further, you have not cited any trial or study showing a particular relevance of this particular device against the others. How was the reason to cite this specific device? You can cite that several devices have been approved by FDA and that in general the system sends the GCM data for a cloud server…

9. I missed some reference in your article regarding stem cell therapy and diabetes. Several recent studies have demonstrated consistent clinical efficacy in controlling diabetes, including in children. I believe that in your review there may be something about this. You mentioned AI as an additional theme in your revision about genomic medicine, so I believe that stem cells is a topic related to precision medicine that deserves some explanation in your revision.

Decision: Updates of precision medicine in type 2 diabetes — R0/PR5

Comments

No accompanying comment.

Author comment: Updates of precision medicine in type 2 diabetes — R1/PR6

Comments

Feb 2, 2023

Editorial Office

Cambridge Prisms: Precision Medicine

Dear Editors,

We are writing to submit our revised review entitled “Updates of precision medicine in type 2 diabetes” (ID: PCM-22-023) by Mingfeng Xia and Xiaoying Li, which we hope you will find suitable for publication in Cambridge Prisms: Precision Medicine.

We thank you and reviewers for the helpful comments, and we have addressed them one by one in the accompanied point-by-point rebuttal file. We have highlighted the changes in red in the revised manuscript. We welcome any further comments, and hope you and the reviewers will agree on our response.

Thanks for your time in examining our manuscript for publication and do not hesitate to contact me if you have any questions.

Yours sincerely,

Xiaoying Li, MD, PhD

Professor and Head

Department of Endocrinology and Metabolism

Zhongshan Hospital, Fudan University

Shanghai, 200032, China

Review: Updates of precision medicine in type 2 diabetes — R1/PR7

Conflict of interest statement

Reviewer declares none.

Comments

Comments to Author: No more comments

Review: Updates of precision medicine in type 2 diabetes — R1/PR8

Conflict of interest statement

Reviewer declares none.

Comments

Comments to Author: This is a poorly written manuscript which requires editing. I am not going to spend time editing this paper. I do believe, based on the first 7 pages I reviewed, that the manuscript is not fit for publication. I spent an hour looking at the first 5 pages then gave up.

Precision Medicine Manuscript Review # 2

Jeff Unger, MD

Page number/Line Comment

3/3 This line is poorly written. Just remove this line and the reference: Diabetes has become a fast increased disease worldwide1

5/4 This sentence needs to be edited: The heterogeneity of diabetes has been recognized several decades ago, which divided people with diabetes into insulin-sensitive and insulin-insensitive subgroups based on the oral glucose tolerance test (OGTT):

Several decades ago, the heterogeneity of glucose intolerance was recognized as a disease state dividing individuals into insulin-sensitive and insulin- insensitive subgroups based on one’s oral glucose tolerance test (OGTT). 7

5/20 This paragraph, again, is poorly written and needs to be edited. I am NOT going to edit this paper.

However, it was not until the introduction of genomic 10 medicine in the recent 20 years, that the molecular mechanisms of the monogenic diabetes were uncovered.

6/12 Change “might be considered” to SHOULD be considered

6/18 Change super sensitive to “highly sensitive”

6/19 Another poorly written sentence:

While individuals with loss-of19 function mutations in the GCK gene are unlikely to develop diabetic complications, and have no need for unnecessary treatment

Suggest removing the word: WHILE

7/20 Poorly written: In 2007, the first genome-wide association study (GWAS) in type 2 diabetes was reported

Should say: The first genome-wide association study (GWAS) in type 2 diabetes was published in…

Recommendation: Updates of precision medicine in type 2 diabetes — R1/PR9

Comments

Comments to Author: Dear colleagues,

We would like to congratulate the dedicated effort on this manuscript. However, we believe that the article does not fit within the proposal and scientific rigor of our journal.

The article is not accepted for publication and we wish you success in future submissions.

Sincerely.

Decision: Updates of precision medicine in type 2 diabetes — R1/PR10

Comments

No accompanying comment.