Skip to main content
The Journal of Clinical Endocrinology and Metabolism logoLink to The Journal of Clinical Endocrinology and Metabolism
. 2021 Jul 22;106(12):e4822–e4833. doi: 10.1210/clinem/dgab545

Toward an Improved Classification of Type 2 Diabetes: Lessons From Research into the Heterogeneity of a Complex Disease

Maria J Redondo 1,2, Ashok Balasubramanyam 3,
PMCID: PMC8787852  PMID: 34291809

Abstract

Context

Accumulating evidence indicates that type 2 diabetes (T2D) is phenotypically heterogeneous. Defining and classifying variant forms of T2D are priorities to better understand its pathophysiology and usher clinical practice into an era of “precision diabetes.”

Evidence Acquisition and Methods

We reviewed literature related to heterogeneity of T2D over the past 5 decades and identified a range of phenotypic variants of T2D. Their descriptions expose inadequacies in current classification systems. We attempt to link phenotypically diverse forms to pathophysiology, explore investigative methods that have characterized “atypical” forms of T2D on an etiological basis, and review conceptual frameworks for an improved taxonomy. Finally, we propose future directions to achieve the goal of an etiological classification of T2D.

Evidence Synthesis

Differences among ethnic and racial groups were early observations of phenotypic heterogeneity. Investigations that uncover complex interactions of pathophysiologic pathways leading to T2D are supported by epidemiological and clinical differences between the sexes and between adult and youth-onset T2D. Approaches to an etiological classification are illustrated by investigations of atypical forms of T2D, such as monogenic diabetes and syndromes of ketosis-prone diabetes. Conceptual frameworks that accommodate heterogeneity in T2D include an overlap between known diabetes types, a “palette” model integrated with a “threshold hypothesis,” and a spectrum model of atypical diabetes.

Conclusion

The heterogeneity of T2D demands an improved, etiological classification scheme. Excellent phenotypic descriptions of emerging syndromes in different populations, continued clinical and molecular investigations of atypical forms of diabetes, and useful conceptual models can be utilized to achieve this important goal.

Keywords: atypical diabetes, pediatric diabetes, LADA, monogenic diabetes, ketosis-prone diabetes, palette model, spectrum model, cluster analysis


Current definitions of type 2 diabetes (T2D) are based on broad pathophysiological concepts that provide little practical guidance to clinicians grappling with an expanding array of phenotypic presentations that elude precise diagnosis, and therapeutic options guided by consensus rather than targeted recommendations (1). Intensive research over decades has revealed that the pathophysiology of T2D is complex, variable, and multifactorial (2-5). It is now becoming increasingly clear that the phenotypes of patients diagnosed with “T2D” are equally complex and varied, likely reflecting heterogeneity of epigenetic, genetic, and acquired defects in intersecting pathways and multiple tissues leading to the final common pathway of hyperglycemia (4). Definitions of T2D as “[a condition]... due to a progressive loss of adequate β-cell insulin secretion frequently on the background of insulin resistance” (1), or “[a pathophysiologic process that]... ranges from predominantly insulin resistance with relative insulin deficiency to a predominantly secretory defect with or without insulin resistance” (6) could enfold virtually any type of diabetes and do not help to specify the varieties of patients around the world diagnosed with “T2D.” An etiological classification of T2D would facilitate precision medicine in diabetes, a highly desirable goal.

Here, we examine evidence for the emerging clinical heterogeneity of T2D, attempt to link its phenotypically diverse forms to pathophysiologic processes, review investigative methods that have proven successful in characterizing variant or atypical forms of diabetes from an etiological perspective, present conceptual frameworks to understand the heterogeneity of T2D, and suggest future directions to achieve the goal of an etiological classification of T2D.

The Phenotypic Heterogeneity of Diabetes Exposes Inadequacies of the Current Classification System

The heterogeneity and complexity of T2D have long been recognized (5, 6). Early observations of phenotypic heterogeneity and corresponding pathophysiologic complexity were made through comparisons of different ethnic/racial groups in diverse geographic regions (4). Beginning with case series in the 1960s through comparative cohort analyses in the 1970s and 1980s, clinical and phenotypic differences were reported in populations with varying risk factors and prevalence rates for T2D (4). Studies then sought to link these variations to differences in genetic susceptibility (7-9). For example, high rates of T2D were reported in “lean” East Asian and South Asian populations, suggesting a phenotypic contrast with obesity-associated T2D in Europid populations. The biological components measured in these studies included body mass index (BMI), waist circumference, dyslipidemia, hypertension, family history of diabetes, insulin sensitivity, and beta cell function, and the analyses of the study data demonstrated that these factors differentially affect population risk of T2D development and rate of progression to chronic complications (7-9). Comparative genetic analyses of the geographic or ethnic groups demonstrated variation in T2D risk loci based on sex (10), body fat distribution (11), and race or ethnicity (12-14). Guided by these findings, physiological investigations subsequently revealed the differences were linked to quantitative differences in insulin sensitivity of different adipose tissue depots (15, 16) or in the mass or function of beta cells (17, 18) between persons belonging to these different groups.

Such comparative studies continue to illuminate mechanisms responsible for phenotypic group differences and inform more complex gene-environment analyses. Minority ethnic groups in the United States have been shown to have higher rates of developing T2D (https://www.cdc.gov/diabetes/pdfs/data/statistics/national-diabetes-statistics-report.pdf) as the result of complex interactions between genetics, environment, and lifestyle, with large influences derived from social determinants of health (19). The observations include a propensity to greater degrees of insulin resistance in African Americans across BMI strata, and glucose metabolic abnormalities at lower levels of BMI in Chinese and South Asian individuals (20-22). Recently, the Mediators of Atherosclerosis if South Asians Living in America (MASALA) and the Multi-Ethnic Study of Atherosclerosis (MESA) studies demonstrated that race and ethnicity are markers of heterogeneity and differential risk for complications in T2D (23). South Asian individuals with T2D have higher glycated hemoglobin A1c (HbA1c), lower beta cell function (20, 21), younger age of onset, and higher incidence of coronary artery disease compared with non-Hispanic White, Chinese, African American, and Hispanic groups. Besides biological considerations, social disparities that influence diet, lifestyle, and access to care are important contributors to the phenotypic heterogeneity of the disease.

Beyond genetic susceptibility, a host of epigenetic influences related to prenatal exposures (24) and pregnancy “programming” (25), lifestyle factors and environmental exposures that affect metabolism both directly and through the mediation of the intestinal microbiome (26, 27), variations in lifestyle and socioeconomic status, as well as “life course” evolution of many of these factors (4, 28) are known to modulate the pathophysiology of T2D, and hence the phenotypic presentations of patients.

There are differences in T2D prevalence among the sexes, with women at younger ages having a higher predisposition to diabetes than men (29) (in contrast with type 1 diabetes [T1D], wherein there is a higher overall male predominance among youth). Importantly, from the standpoint of a useful etiological classification, the pathophysiology itself may be different on the basis of sex. For example, although the metabolic syndrome is considered almost invariably associated with T2D, inasmuch as insulin resistance is a foundation of both conditions, a recent report suggests that a substantial proportion (45%) of women with T2D have no generally accepted metabolic syndrome components (other than dysglycemia) (30). Males and females clearly have different pathophysiologic interactions of biological pathways leading to T2D.

T2D was once synonymous with “adult-onset” diabetes, but this concept was rendered obsolete in the late twentieth century by a rising epidemic of the condition in pediatric populations. The incidence of childhood-onset T2D has risen by 4.8% over the past decade (31-33), associated with the increasing prevalence of childhood obesity. Compared to adults with T2D, children with T2D lose insulin secretory capacity earlier in the course of the disease (34-36) and have more pronounced decrease in insulin sensitivity, higher rates of failure to respond to metformin alone or with rosiglitazone (37), and overall worse health outcomes due to a range of social and genetic factors (38-40). There is emerging heterogeneity within pediatric T2D as well. The small but distinct fraction of children who develop a T2D phenotype prior to or early in puberty (41) have unique features and pathophysiologic drivers distinct from the rise of pubertal hormones (such as growth hormone, insulin-like growth factor-1, and sex hormones) often implicated as a mechanism underlying T2D in older children and adolescents (41-43). Productive investigation toward etiologic definition of pre-pubertal T2D may involve the roles of exposures in the prenatal period (eg, due to maternal diabetes) or in early infancy (24, 44). Defining variations in pediatric T2D phenotype and course due to sex, ancestry, and geography also remain to be explored. Some children with T2D present with marked ketosis or diabetic ketoacidosis (DKA) even absent circulating islet autoantibodies, while others do not (45). Do the former simply represent a more rapid deterioration of beta cell function (and if so, why?), or as with adult A−ß+ ketosis-prone diabetes (see below), do they have novel defects in intermediary metabolism that accelerate ketone production or inhibit its oxidation?

As in adult T2D, pediatric natural history studies, as well as clinical trials coupled with population data analyses, such as the Search for Diabetes in Youth (SEARCH) (46), Treatment Options for type 2 Diabetes in Adolescents and Youth (TODAY) (47), or Restoring Insulin Secretion (RISE) (48) studies have contributed much to understanding pediatric T2D heterogeneity, and collaborations between the study groups have been useful in this regard (49). Progression to insulin dependence is variable in children with T2D (47), and one relevant factor is islet autoimmunity manifested by autoantibodies in some T2D children. These children have a higher likelihood of rapid progression to insulin dependence (50) due to early beta cell function loss (51). Terms such as diabetes 1.5, double diabetes (52), or latent autoimmune diabetes in youth (50) have sprouted to indicate these variants, but we should avoid the easy or eponymous attachment of labels that bring us no closer to an etiological classification than type 1 or type 2 diabetes.

Approach to an Etiological Classification Based on Understanding Atypical Forms of T2D

The process of defining the currently known monogenic syndromes of diabetes serves as a paradigm for an etiological classification of T2D diabetes based on careful linkage of “atypical” phenotypes with specific genetic causes. The clinical utility of this classification has been demonstrated in the manner in which it permits precise diagnosis and targeted therapy (53-56). Historically, the recognition of monogenic diabetes began with careful delineation of phenotypic characteristics such as early onset or autosomal dominant transmission of the trait and their initial classification as a separate entity denoted maturity-onset diabetes of youth (MODY). Aggregation of cases and families together with prospective population studies by dedicated expert groups (eg, at the Universities of Exeter and Chicago) have led to accurate screening modalities such as the phenotypic MODY risk calculator (57), and a genetic risk score that can differentiate adults and young adults with T1D from those with potentially monogenic diabetes (58). Following such screening or upon clinical suspicion, one can readily perform “MODY testing” using commercially available gene panels or rapid exome sequencing. In nononcologic adult clinical practice, this is one of the early examples of precision diagnosis made feasible by an etiological classification, and its benefits are clear whenever an apparently insulin-dependent patient is successfully transitioned to sulfonylurea (eg, persons with pathogenic variants in the hepatocyte nuclear factor-4α [HNF4-alpha] or hepatocyte nuclear factor-1α [HNF1-alpha] genes), or a person diagnosed with “diabetes” may dispense with diabetes medications altogether (eg, persons with variants in the glucokinase [GCK] gene), or a family member at risk is identified at a preclinical stage.

However, the rarity of known monogenic diabetes (under 4% in selected patient cohorts (59), thus even lower in the general diabetes population) makes this model less useful as a guide to classifying other atypical T2D phenotypes that may arise from more complex genetics or epigenetics. Longitudinal characterization of patients with such phenotypes and assessment of their natural histories have proven productive in delineating subgroups among these T2D variants, anticipating the development of “omics” tools for mechanistic analysis of their multifactorial etiologies. A good example lies in the evolution of our understanding of syndromes of ketosis-prone diabetes (KPD) (60).

KPD is defined by the striking presentation with DKA of patients lacking clinical evidence of autoimmune T1D. It is an emerging cluster of conditions recognized in larger numbers over the last 3 decades, among Asian, Afro-Caribbean, African American, US Hispanic, and South American populations (61-76). Based on longitudinal descriptions and multivariable analysis of a large database, an algorithm utilizing 2 biomarkers (islet autoantibodies and a measure of beta cell functional reserve, each present or absent by validated cutoffs) defines different forms of KPD with distinct pathophysiologic mechanisms (64). Of the 4 subgroups defined by this 2 × 2 factorial approach, the 2 autoantibody-negative (A−) groups represent complex atypical T2D syndromes. A−ß+ KPD (autoantibody absent, beta cell function present) comprises ~50% of patients presenting with DKA in a US urban multiethnic population. These patients can discontinue insulin therapy following the index DKA episode and about half can maintain excellent glycemic control on oral medications for many years (64). The latter subgroup of patients develop unprovoked DKA and are characterized by late onset, male predominance, obesity, DKA at initial diagnosis of diabetes, and lack of HLA class II T1D susceptibility alleles or T-cell reactivity to islet autoantigens (77, 78). Plasma metabolomics, validated by kinetic studies utilizing stable isotopes and mass spectrometry, has revealed that these patients have variable defects in branch-chain amino acid and arginine metabolism that may lead to abnormal ketogenesis but defective ketone oxidation (79, 80), and inability to maintain sufficient insulin secretion during hyperglycemic crises (81).

The clinical phenotype of A−β− (autoantibody absent, beta cell function absent) KPD resembles lean, late-onset T1D but the patients lack evidence for islet autoimmunity (absent islet autoantibodies and low frequency of HLA risk alleles for T1D) (64). In addition to identifying as patients who require life-long insulin therapy following the index DKA, they represent a valuable reservoir for discovery of novel monogenic or oligogenic mechanisms of severe beta cell failure using physiologic insulin secretion studies and more detailed analysis of patient-specific inducible pluripotent stem cells differentiated along the pancreatic islet lineage. Preliminary evidence indicates approximately a quarter have potentially pathogenic variants in the HNF-1α and pancreas-duodenum homeobox-1 (PDX-1) genes, encoding critical transcription factors for beta cell development (82).

The potential utility of the Aß classification has been demonstrated more broadly in application to pediatric diabetes (83), permitting identification of 4 groups with distinct characteristics at diagnosis and clinical course. The SEARCH investigators have used a similar 2 × 2 factorial analysis (presence/absence of islet autoimmunity and presence/absence of insulin resistance) to characterize different forms of diabetes, including T2D, in their childhood and adolescent population. The SEARCH classification of pediatric diabetes has genetic associations (84) and has proven useful for epidemiological discovery (85). However, this classification was not designed for individual diagnosis, nor has it been tested for clinical management. Development and application of similar classification algorithms in different prospectively followed cohorts with new-onset T2D should be a priority for translational research. If validated, especially with predictive capacity (as with the Aß classification for KPD), they would fulfill an important unmet need in clinical practice (86).

Conceptual Frameworks of Diabetes Heterogeneity

Heterogeneity as Reflecting an Overlap Between Diabetes Types

An acknowledged model to accommodate variant forms of T2D is to consider them as “overlaps” with other diabetes types. The most established example of this is latent autoimmune diabetes in adults (LADA), which defines patients who display key characteristics of both T1D and T2D, such as presentation in middle-late adulthood, positivity for a single islet autoantibody, risk alleles for both T1D and T2D, and a faster rate of progression to insulin requirement than autoantibody-negative T2D patients (87, 88). The prevalence of LADA ranges from 6% to 12% depending on the T2D cohort studied (89-96). However, the scope and variety of the overlapping phenotypes may be much greater if T-cell reactivity to islet autoantigens (ie, cellular autoimmunity) rather than autoantibodies (ie, humoral autoimmunity) alone were used to define the autoimmune basis of beta cell dysfunction. In a representative subset of T2D participants in the Glycemic Reduction Approaches in Diabetes–A Comparative Effectiveness (GRADE) study, 41% of participants had evidence for cellular islet autoimmunity, while 13% had humoral islet autoimmunity, and only 5% had evidence for both (97). These differences in autoimmune processes, apart from revealing a much higher prevalence of islet autoimmunity than previously considered in T2D, had physiologic and clinical relevance as participants with cellular islet autoimmunity manifested decreased beta cell function and worse glycemic control (97). These data support the case that the overlap model is not a simple construct but encompasses heterogeneous pathophysiologic pathways and clinical syndromes of T2D.

Clinically accessible tools now exist to identify patients with phenotypes that lie within the T1D-T2D overlap. A genetic risk score (GRS) for T1D (98) that incorporates T1D-associated genetic risk variants can differentiate autoimmune T1D from monogenic diabetes among adults and young adults (58, 99). A shortcoming of this GRS is its use of gene variants predominant in persons of European ancestry, so recently others, such as an African ancestry-specific GRS, have been developed (100). The predictive value of these GRSs makes them useful for diagnosis, prognosis and screening of patients’ family members (101). Studies are ongoing to test the role of GRS in classifying pediatric diabetes.

Phenotypic heterogeneity in T1D and KPD has been dissected using markers of insulin secretion, and this approach may also help to place the phenotypic heterogeneity in T2D on an etiological footing. In T1D, clinical trials of immune modulation to preserve residual beta cell function may have been confounded by pathogenic heterogeneity not well reflected by the current diagnostic tests. A measure of insulin secretion may be built into patient characterization at the time of diagnosis. For example, Index60 combines glucose and C-peptide values measured during an oral glucose tolerance test (102). In autoantibody-positive individuals with dysglycemia, those in the upper quartile of the Index60 distribution were significantly younger, with lower BMI and higher frequency of autoantibody positivity compared with those in the lower quartile (103), suggesting that the latter group may have a different pathogenesis, not adequately addressed by immunomodulatory therapies. Tools like Index60 and GRS may be effective at resolving heterogeneity within the T1D/T2D overlap.

The Palette and Threshold Models of Heterogeneity

Diabetes may result from multiple possible mechanisms that combine and interact to disrupt metabolic homeostasis. These mechanisms may vary by individual and, within the same individual, as time goes by. This has led to the concept that genetic mechanisms that are typically associated with either T1D or T2D may operate in the manner of a palette, ie, with varying combinations in different individuals to generate a range of phenotypes (104-106). A parallel concept lies in the “threshold hypothesis” (107) wherein interaction of both genetic and environmental factors may force an individual’s metabolic pathways to exceed a normal threshold and progress to diabetes. The palette model and the threshold hypothesis may be integrated to explain a wide, combinatorial variety of diabetes phenotypes (108). For instance, excess BMI combines with autoantibody positivity in both children (109) and adults (110), with different thresholds by sex and age, to elevate the risk of T1D and modify the rate of progression of islet autoimmunity (111). As another example, T2D-associated genetic variants in the TCF7L2 locus are more frequent in both children and adults with new-onset autoimmune T1D who carry a single islet autoantibody (a marker of only mild islet autoimmunity) or lack T1D-associated HLA genotypes (112-114). In these individuals, diabetes results from the combination of relatively slow progression of autoimmune islet destruction and additional diabetogenic mechanisms that are typically associated with T2D, resulting in atypical T1D characteristics (115, 116). This conceptual model shows that diabetes heterogeneity at the individual level requires recognition of multiple dynamic pathophysiological processes in the same person. While it does not readily evolve into an etiological classification, it provides useful guidance in clinical and research practice, such as including BMI as an important predictor for progression from preclinical to clinical T1D (117), using of metformin in pediatric T1D to improve insulin sensitivity (118), and recognition that islet autoantibody positivity accelerates progression to insulin dependence in T2D (50).

The Spectrum Model of Heterogeneity

As described in an earlier review by one of us (A.B.), variant forms of diabetes could fall into a spectrum of diseases or syndromes between the 2 poles of “typical” autoimmune T1D and “typical” T2D (119). The spectrum includes the currently known, etiological forms of atypical diabetes, including monogenic diabetes and the atypical subgroups of KPD, as well as LADA and T2D associated with T-cell mediated islet autoimmunity. The spectrum would also accommodate a range of currently unknown atypical forms of diabetes. Approaches to identify these unknown forms would require a concerted and dedicated effort beginning with selection of patients (from specialty clinics, registries of monogenic, lipodystrophic, syndromic, and KPD, cohorts of relatively undifferentiated “T2D” patients enrolled in clinical trials, as well as from patient self-referral or clinician referral), moving through a process to identify variant T2D phenotypes by clinical and biochemical criteria (eg, early or pre-pubertal onset, leanness, Mendelian inheritance of the trait, absence of autoantibodies in the face of severely deficient insulin production) and employing the full technological force of comprehensive “omics” analysis and physiological testing on the patients and their biosamples to delineate the pathophysiologic mechanisms. The outcome would be a clustered compendium of T2D variants classified by etiologic mechanisms. Such is the approach and goal of the Rare and Atypical Diabetes Network (RADIANT) (see below).

Future Directions Toward an Etiological Classification of T2D

To prepare the ground for a new, etiological classification of T2D, research is needed to sharpen our understanding of the pathogenic basis of the phenotypically variant forms of the condition. If the study of atypical forms of T2D could open windows into distinct pathophysiologic pathways leading to T2D, then comprehensive strategies to identify patients and families with these syndromes should be scientific priorities for clinical investigation. One of the challenges in this effort is the potential for wide variation in genetic contributions to pathogenesis within each form. If the patients were identified based on early onset of diabetes without evidence of islet autoimmunity, a Mendelian pattern of inheritance, or syndromic features that have failed to produce a clear diagnosis despite standard clinical evaluations (including MODY genetic testing), the cause of the diabetes might be pinpointed to novel, rare variants in 1 or 2 genes in the nuclear or mitochondrial genome that have a strong phenotypic effect. At specialized centers to which such patients may be referred, genetic testing is often recommended. The patients and their families are enrolled in genetics registries, and their data made available to researchers. If the phenotype is more complex, including late onset of diabetes and an uninformative pedigree in adult patients, then rare gene variants are less likely to contribute significantly to the etiology. In these cases, complex genetics, epigenetics, environmental factors, or combinations therein may underlie the pathogenesis. Identifying atypical forms within this group requires different methods that account for this complexity. Electronic searches and clustering analyses of extensive datasets from very large numbers of diabetes patients may discern discrete forms of atypical diabetes based on both genetic and phenotypic data.

Multiple approaches could help identify and define forms of atypical diabetes, investigations of which are likely to uncover pathogenic mechanisms relevant to more common forms of T2D. These include:

Population Diabetes Cohort/Diabetes Study Clinic Approach

Existing diabetes cohorts, registries, and studies have large longitudinal databases, stored biosamples of fasting serum, DNA, or peripheral blood mononuclear cells, broad geographic spread, and a wide range of age and ethnic groups likely enriched in atypical forms of diabetes. Unique study designs could further enrich some cohorts in patients with atypical forms of T2D—for example, the SEARCH cohort is exceptionally large and diverse and includes every form of pediatric and adolescent diabetes from across the United States. The TODAY cohort focused on new-onset “T2D” in children, which is likely enriched in atypical forms of higher complexity. Cohorts of recent-onset diabetic patients from North and South India include different regional ethnic populations who likely harbor unique forms of “lean” T2D. “Lean T2D” has proven, even with clinical and biochemical measures, to aggregate into atypical subgroups, such as ketosis-resistant diabetes of youth (27, 28) and the more recently described insulin-resistant obese diabetes (IROD) and combined insulin-resistant and deficient diabetes (CIRDD) (120). A filtering process, such as has been developed by the RADIANT study (see below), could serve to cull out from these databases persons with atypical features. These processes could be adapted to perform similar searches among in electronic medical records (especially if natural language processing software are available) of clinics and health systems and very large national or international databases such as the Botnia project, UK Biobank, the US Department of Veterans Affairs’ Million Veterans Program, or the DPV cohort in Germany. More circumscribed existing registries for specific atypical forms of diabetes (monogenic diabetes registry at the University of Chicago, lipodystrophy registry at the University of Michigan, Ketosis-Prone Diabetes registry at Baylor College of Medicine) are smaller but have potential for a higher yield of such patients.

Genetics Registries Approach

Patients referred to clinical geneticists and genetic testing laboratories are a potentially rich source to mine for patients with atypical T2D. A review of patient phenotypes referred to the Baylor College of Medicine clinical and research genetics clinics (comprising the Clinical Genetics laboratory, the mitochondrial genetics laboratory, the nuclear whole-exome sequencing laboratory, and the Center for Mendelian Genetics) illustrates this possibility. In the >100 000 cases referred to these laboratories collectively for genetic testing, approximately 800 were referred for phenotypic characteristics that included diabetes. Diabetes patients in the nuclear exome sequencing laboratory and mitochondrial sequencing laboratory data sets were reviewed for the following atypical criteria: age at testing (early-onset or later-onset with features atypical of T2D), strong family history (multigenerational), syndromic features, existing variant interpretations and genotype-phenotype associations, and expert review by clinical diabetologists (121). Suspicion for atypical diabetes was raised in 31 of the 287 nuclear exome sequencing referrals and 87 of the 115 mitochondrial DNA sequencing referrals. Since genetic testing is often ordered for atypical presentation of a common disease, clinical genetic diagnostic lab data are a rich source for discovery of patients with atypical forms of diabetes.

Machine Learning Approaches for Subgrouping T2D

Machine learning models and statistical techniques offer exciting opportunities to uncover heterogeneous forms of diabetes within T2D by clustering individuals with similar biochemical, genetic, and clinical characteristics. The limitations are largely related to the nature and coding of the stored data and the selection of variables to circumscribe the phenotypes.

Alqvist et al performed unsupervised hierarchical cluster analysis on clinical data from ~9000 Swedish adult patients with newly diagnosed diabetes (122), using 6 variables collected systematically over many years in longitudinal Scandinavian registries. The variables were GAD65-Ab, age at diagnosis, BMI, and measures of insulin sensitivity and insulin secretion. The analysis uncovered 5 clusters of T2D: severe autoimmune diabetes (SAID), severe insulin-dependent diabetes (SIDD), severe insulin-resistant diabetes (SIRD), mild obesity-related diabetes (MOD) and mild age-related diabetes (MARD). The clinical relevance of the clusters (each representing a distinctive syndrome of T2D) was evident in their natural histories and risks of complications. This clustering analysis demonstrates that the heterogeneity of T2D is susceptible to useful, predictive subclassification based on key differences in phenotypic markers such as insulin sensitivity, metabolic syndrome markers, BMI, and age of onset. SAID and SIDD patients had the poorest glycemic control and became insulin dependent over the shortest period. SIDD patients had higher frequency of diabetic retinopathy than other clusters. SIRD patients had the highest prevalence of nonalcoholic fatty liver disease, and, despite a relatively low HbA1c, developed diabetic kidney disease at the fastest rate. Limited analysis of T2D risk alleles indicated that SIRD might be genetically distinct from other forms of T2D; the TCF7L2 rs7903146 variant that is strongly associated with T2D overall was associated with SIDD, MOD, and MARD but not with SIRD, and the T2D genetic risk score was associated with all the clusters but not with SIRD.

Udler et al used nonnegative matrix factorization (123) in over 17 000 patients with T2D genetic variants and 47 traits from genome-wide association studies of T2D cohorts (64). They also identified 5 clusters, each differentially enriched for tissue-specific regulators of gene expression (enhancers and promoters). Recently a large T2D cohort database in South India was utilized to identify 4 clusters prone to young-onset T2D (119). Two clusters (SIDD and MARD) were similar to those identified in the Scandinavian study. Two new clusters unique to this ethnic group were identified, denoted IROD and CIRDD. Bancks et al (124) analyzed T2D subgroups in participants in the Look AHEAD weight loss study and observed that intensive lifestyle intervention was associated with higher risk of cardiovascular disease in the subgroup with poorest glucose control. Wagner et al recently proposed that clusters can be identified even in the prediabetic period, drawing upon variables that would not be confounded by secondary effects of gluco- and lipotoxicity (125). In adults at risk for T2D, these authors identified 6 clusters with different risk of progression to T2D despite, in some cases, similar glycemic levels. Thus, although there is not a straight path from current cluster analysis to an etiological classification scheme for T2D, the clusters permit clinicians to refine screening approaches and provide better informed therapy to different groups of individuals with T2D.

Another limitation of extant clustering / bioinformatics analyses arises from the inclusion of more or less undifferentiated forms of T2D in the cohorts studied. Rare or atypical forms of diabetes may not find a taxonomic home using this approach. There is an ongoing attempt to first filter a T2D population for atypical features, and then perform cluster analysis in those persons, in a Mexican American cohort in South Texas. The approach is to manually identify patients who do not fit the conventional criteria of T1D or T2D and then use a machine learning procedure with a filtering process utilizing variables such as lean habitus, islet autoantibody, age of onset, insulin dependence, and C-peptide, followed by a robust spatial clustering algorithm (Parikh H et al, unpublished). Validation of this and similar approaches in larger and more geographically and ethnically varied cohorts are needed. Electronic medical records can be leveraged for the purpose of identifying atypical diabetes but this requires the development of algorithms that are adapted to the specific target population (126). Overall, a variety of approaches will be needed to determine the true prevalence of atypical diabetes and their forms. This is a goal of RADIANT, a multicenter collaborative project funded by the National Institutes of Health, launched in October 2020. This innovative project should provide detailed insight into the pathophysiology of a wide range of atypical forms of diabetes, with the promise through clustering analyses of these mechanistically defined phenotypes to better understand and develop an etiological classification of T2D.

Acknowledgments

Financial Support: National Institute of Diabetes and Digestive and Kidney Diseases R01 DK124395 (M.J.R.), R01 DK121843 (M.J.R.), R01 DK101411 (A.B.) and R01 DK104832 (A.B.).

Glossary

Abbreviations

BMI

body mass index

CIRDD

combined insulin-resistant and deficient diabetes

DKA

diabetic ketoacidosis

GRS

genetic risk score

HbA1c

glycated hemoglobin A1c

IROD

insulin resistant obese diabetes

KPD

ketosis-prone diabetes

LADA

latent autoimmune diabetes in adults

MARD

mild age-related diabetes

MOD

mild obesity-related diabetes

MODY

maturity onset diabetes of the young

SAID

severe autoimmune diabetes

SIDD

severe insulin-dependent diabetes

SIRD

severe insulin-resistant diabetes

T1D

type 1 diabetes

T2D

type 2 diabetes

Contributor Information

Maria J Redondo, Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children’s Hospital, Houston, TX 77030, USA.

Ashok Balasubramanyam, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA.

Additional Information

Disclosures: The authors affirm no conflicts of interest related to the contents of this paper.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

  • 1. American Diabetes Association. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2021 [published correction appears in Diabetes Care. 2021 Jun 16;dc21ad09]. Diabetes Care. 2021;44(Suppl 1):S15-S33. [DOI] [PubMed] [Google Scholar]
  • 2. Defronzo  RA. Banting Lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes. 2009;58(4):773-795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Kahn  SE, Cooper ME, Del Prato S. Pathophysiology and treatment of type 2 diabetes: perspectives on the past, present, and future. Lancet. 2014;383(9922):1068-1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Balasubramanyam  A, Tandon N, Yajnik C. Non-traditional forms of diabetes worldwide: implications for translational investigation. In: Powers AC, ed. Translational Endocrinology & Metabolism: Type 2 Diabetes Update. Washington, DC: Endocr Soc; 2011:43-67. [Google Scholar]
  • 5. Report of the expert committee on the diagnosis and classification of diabetes mellitus. Diabetes Care. 1997;20(7):1183-1197. [DOI] [PubMed] [Google Scholar]
  • 6. World Health Organization. Definition, Diagnosis and Classification of Diabetes Mellitus and Its Complications: Report of a WHO Consultation. Part 1, Diagnosis and Classification of Diabetes Mellitus. WHO/NCD/NCS/99.2. World Health Organization; 1999. [Google Scholar]
  • 7. Rostambeigi  N, Shaw JE, Atkins RC, et al.  Waist circumference has heterogeneous impact on development of diabetes in different populations: longitudinal comparative study between Australia and Iran. Diabetes Res Clin Pract. 2010;88(1):117-124. [DOI] [PubMed] [Google Scholar]
  • 8. Birkeland  KI, Kilhovd B, Thorsby P, et al.  Heterogeneity of non-insulin-dependent diabetes expressed as variability in insulin sensitivity, beta-cell function and cardiovascular risk profile. Diabet Med. 2003;20(1):37-45. [DOI] [PubMed] [Google Scholar]
  • 9. Cho  SB, Kim SC, Chung MG. Identification of novel population clusters with different susceptibilities to type 2 diabetes and their impact on the prediction of diabetes. Sci Rep. 2019;9(1):3329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Ryoo  H, Woo J, Kim Y, Lee C. Heterogeneity of genetic associations of CDKAL1 and HHEX with susceptibility of type 2 diabetes mellitus by gender. Eur J Hum Genet. 2011;19(6):672-675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Timpson  NJ, Lindgren CM, Weedon MN, et al.  Adiposity-related heterogeneity in patterns of type 2 diabetes susceptibility observed in genome-wide association data. Diabetes. 2009;58(2):505-510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Kong  X, Xing X, Hong J, Zhang X, Yang W. Genetic variants associated with lean and obese type 2 diabetes in a Han Chinese population: a case-control study. Medicine (Baltimore). 2016;95(23):e3841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ng  MC, Park KS, Oh B, et al.  Implication of genetic variants near TCF7L2, SLC30A8, HHEX, CDKAL1, CDKN2A/B, IGF2BP2, and FTO in type 2 diabetes and obesity in 6719 Asians. Diabetes. 2008;57(8):2226-2233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Perry  JR, Voight BF, Yengo L, et al. ; MAGIC; DIAGRAM Consortium; GIANT Consortium . Stratifying type 2 diabetes cases by BMI identifies genetic risk variants in LAMA1 and enrichment for risk variants in lean compared to obese cases. Plos Genet. 2012;8(5):e1002741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Wium  C, Eggesbø HB, Ueland T, et al.  Adipose tissue distribution in relation to insulin sensitivity and inflammation in Pakistani and Norwegian subjects with type 2 diabetes. Scand J Clin Lab Invest. 2014;74(8):700-707. [DOI] [PubMed] [Google Scholar]
  • 16. Møller  JB, Pedersen M, Tanaka H, et al.  Body composition is the main determinant for the difference in type 2 diabetes pathophysiology between Japanese and Caucasians. Diabetes Care. 2014;37(3):796-804. [DOI] [PubMed] [Google Scholar]
  • 17. Møller  JB, Dalla Man C, Overgaard RV, et al.  Ethnic differences in insulin sensitivity, β-cell function, and hepatic extraction between Japanese and Caucasians: a minimal model analysis. J Clin Endocrinol Metab. 2014;99(11):4273-4280. [DOI] [PubMed] [Google Scholar]
  • 18. Hasson  BR, Apovian C, Istfan N. Racial/ethnic differences in insulin resistance and beta cell function: relationship to racial disparities in type 2 diabetes among African Americans versus Caucasians. Curr Obes Rep. 2015;4(2):241-249. [DOI] [PubMed] [Google Scholar]
  • 19. Spanakis  EK, Golden SH. Race/ethnic difference in diabetes and diabetic complications. Curr Diab Rep. 2013;13(6):814-823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Kanaya  AM, Herrington D, Vittinghoff E, et al.  Understanding the high prevalence of diabetes in U.S. south Asians compared with four racial/ethnic groups: the MASALA and MESA studies. Diabetes Care. 2014;37(6):1621-1628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Gujral  UP, Vittinghoff E, Mongraw-Chaffin M, et al.  Cardiometabolic abnormalities among normal-weight persons from five racial/ethnic groups in the United States: a cross-sectional analysis of two cohort studies. Ann Intern Med. 2017;166(9):628-636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Gujral  UP, Narayan KM, Kahn SE, Kanaya AM. The relative associations of β-cell function and insulin sensitivity with glycemic status and incident glycemic progression in migrant Asian Indians in the United States: the MASALA study. J Diabetes Complications. 2014;28(1):45-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Bancks  MP, Bertoni AG, Carnethon M, et al.  Association of diabetes subgroups with race/ethnicity, risk factor burden and complications: the MASALA and MESA studies. J Clin Endocrinol Metab. 2021;106(5):e2106-e2115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Dabelea  D, Crume T. Maternal environment and the transgenerational cycle of obesity and diabetes. Diabetes. 2011;60(7):1849-1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Howe  CG, Cox B, Fore R, et al.  Maternal gestational diabetes mellitus and newborn DNA methylation: findings from the pregnancy and childhood epigenetics consortium. Diabetes Care. 2020;43(1):98-105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Díaz-Rizzolo  DA, Kostov B, López-Siles M, et al.  Healthy dietary pattern and their corresponding gut microbiota profile are linked to a lower risk of type 2 diabetes, independent of the presence of obesity. Clin Nutr. 2020;39(2):524-532. [DOI] [PubMed] [Google Scholar]
  • 27. Massey  W, Brown JM. The gut microbial endocrine organ in type 2 diabetes. Endocrinology. 2021;162(2):bqaa235. doi:10.1210/endocr/bqaa235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Sebert  S, Lowry E, Aumüller N, et al.  Cohort profile: the DynaHEALTH consortium - a European consortium for a life-course bio-psychosocial model of healthy ageing of glucose homeostasis. Int J Epidemiol. 2019;48(4):1051-1051k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Mauvais-Jarvis  F, Bairey Merz N, Barnes PJ, et al.  Sex and gender: modifiers of health, disease, and medicine. Lancet. 2020;396(10250):565-582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Rottenkolber  M, Gar C, Then C, et al.  A pathophysiology of type 2 diabetes unrelated to metabolic syndrome. J Clin Endocrinol Metab. 2021;106(5):1460-1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Dabelea  D. Diabetes in youth-looking backwards to inform the future: kelly west award lecture 2017. Diabetes Care. 2018;41(2):233-240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Mayer-Davis  EJ, Lawrence JM, Dabelea D, et al. ; SEARCH for Diabetes in Youth Study . Incidence trends of type 1 and type 2 diabetes among youths, 2002-2012. N Engl J Med. 2017;376(15):1419-1429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Dabelea  D, Hamman R, Knowler W. Diabetes in youth. In: Cowie CC, Casagrande SS, Menke A, et al., eds. Diabetes in America. 3rd ed. National Institute of Diabetes and Digestive and Kidney Diseases (US); 2017:11-15. [PubMed] [Google Scholar]
  • 34. Zeitler  P, Hirst K, Pyle L, et al.  A clinical trial to maintain glycemic control in youth with type 2 diabetes. N Engl J Med. 2012;366:2247-2256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Consortium  R. Impact of insulin and metformin versus metformin alone on β-cell function in youth with impaired glucose tolerance or recently diagnosed type 2 diabetes. Diabetes Care. 2018;41(8): 1717-1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Arslanian  SA, El Ghormli L, Kim JY, et al. ; RISE Consortium . OGTT glucose response curves, insulin sensitivity, and β-cell function in RISE: comparison between youth and adults at randomization and in response to interventions to preserve β-cell function. Diabetes Care. 2021;44(3):817-825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Nadeau  KJ, Anderson BJ, Berg EG, et al.  Youth-onset type 2 diabetes consensus report: current status, challenges, and priorities. Diabetes Care. 2016;39(9):1635-1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Savic Hitt  TA, Katz LEL. Pediatric type 2 diabetes: not a mini version of adult type 2 diabetes. Endocrinol Metab Clin North Am. 2020;49(4):679-693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Constantino  MI, Molyneaux L, Limacher-Gisler F, et al.  Long-term complications and mortality in young-onset diabetes: type 2 diabetes is more hazardous and lethal than type 1 diabetes. Diabetes Care. 2013;36(12):3863-3869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Maahs  DM, Snively BM, Bell RA, et al.  Higher prevalence of elevated albumin excretion in youth with type 2 than type 1 diabetes: the SEARCH for Diabetes in Youth study. Diabetes Care. 2007;30(10):2593-2598. [DOI] [PubMed] [Google Scholar]
  • 41. Astudillo  M, Tosur M, Castillo C, et al.  Type 2 diabetes in prepubertal children. Pediatr Diabetes. 2021. (in press). [DOI] [PubMed] [Google Scholar]
  • 42. Amutha  A, Unnikrishnan R, Anjana RM, Mohan V. Prepubertal childhood onset type 2 diabetes mellitus: four case reports. J Assoc Physicians India. 2017;65(2):43-46. [PubMed] [Google Scholar]
  • 43. Hutchins  J, Barajas RA, Hale D, Escaname E, Lynch J. Type 2 diabetes in a 5-year-old and single center experience of type 2 diabetes in youth under 10. Pediatr Diabetes. 2017;18(7):674-677. [DOI] [PubMed] [Google Scholar]
  • 44. Dabelea  D, Mayer-Davis EJ, Lamichhane AP, et al.  Association of intrauterine exposure to maternal diabetes and obesity with type 2 diabetes in youth: the SEARCH Case-Control Study. Diabetes Care. 2008;31(7):1422-1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Praveen  PA, Hockett CW, Ong TC, et al.  Diabetic ketoacidosis at diagnosis among youth with type 1 and type 2 diabetes: results from SEARCH (United States) and YDR (India) registries. Pediatr Diabetes. 2021;22(1):40-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Divers  J, Mayer-Davis EJ, Lawrence JM, et al.  Trends in incidence of type 1 and type 2 diabetes among youths—selected counties and Indian reservations, United States, 2002-2015. MMWR Morb Mortal Wkly Rep. 2020;69(6):161-165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Bacha  F, El Ghormli L, Arslanian S, et al. ; TODAY Study Group . Predictors of response to insulin therapy in youth with poorly-controlled type 2 diabetes in the TODAY trial. Pediatr Diabetes. 2019;20(7):871-879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Mokhlesi  B, Tjaden AH, Temple KA, et al. ; RISE Consortium . Obstructive sleep apnea, glucose tolerance, and β-cell function in adults with prediabetes or untreated type 2 diabetes in the restoring insulin secretion (RISE) study. Diabetes Care. 2021;44(4):993-1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Srinivasan  S, Chen L, Todd J, et al. ; ProDiGY Consortium . The first genome-wide association study for type 2 diabetes in youth: the progress in diabetes genetics in youth (ProDiGY) consortium. Diabetes. 2021;70(4):996-1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Awa  WL, Boehm BO, Rosinger S, et al. ; DPV Initiative and the German BMBF Competence Networks Diabetes Mellitus and Obesity . HLA-typing, clinical, and immunological characterization of youth with type 2 diabetes mellitus phenotype from the German/Austrian DPV database. Pediatr Diabetes. 2013;14(8):562-574. [DOI] [PubMed] [Google Scholar]
  • 51. Syed  F, Tersey SA, Turatsinze JV, et al.  Circulating unmethylated CHTOP and INS DNA fragments provide evidence of possible islet cell death in youth with obesity and diabetes. Clin Epigenetics. 2020;12(1):116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Libman  IM, Becker DJ. Coexistence of type 1 and type 2 diabetes mellitus: “double” diabetes? Pediatr Diabetes. 2003;4(2):110-113. [DOI] [PubMed] [Google Scholar]
  • 53. Hattersley  AT, Patel KA. Precision diabetes: learning from monogenic diabetes. Diabetologia. 2017;60(5):769-777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Yang  Y, Chan L. Monogenic diabetes: what it teaches Us on the common forms of type 1 and type 2 diabetes. Endocr Rev. 2016;37(3):190-222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Chung  WK, Erion K, Florez JC, et al.  Precision medicine in diabetes: a consensus report from the american diabetes association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2020;43(7):1617-1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Chung  WK, Erion K, Florez JC, et al.  Precision medicine in diabetes: a Consensus Report from the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia. 2020;63(9):1671-1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. 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):1265-1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Patel  KA, Oram RA, Flanagan SE, et al.  Type 1 diabetes genetic risk score: a novel tool to discriminate monogenic and type 1 diabetes. Diabetes. 2016;65(7):2094-2099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Shields  BM, Shepherd M, Hudson M, et al. ; UNITED study team . Population-based assessment of a biomarker-based screening pathway to aid diagnosis of monogenic diabetes in young-onset patients. Diabetes Care. 2017;40(8):1017-1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Balasubramanyam  A, Nalini R, Hampe CS, Maldonado M. Syndromes of ketosis-prone diabetes mellitus. Endocr Rev. 2008;29(3):292-302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Winter  WE, Maclaren NK, Riley WJ, Clarke DW, Kappy MS, Spillar RP. Maturity-onset diabetes of youth in black Americans. N Engl J Med. 1987;316(6):285-291. [DOI] [PubMed] [Google Scholar]
  • 62. Banerji  MA, Chaiken RL, Huey H, et al.  GAD antibody negative NIDDM in adult black subjects with diabetic ketoacidosis and increased frequency of human leukocyte antigen DR3 and DR4. Flatbush diabetes. Diabetes. 1994;43(6):741-745. [DOI] [PubMed] [Google Scholar]
  • 63. Umpierrez  GE, Casals MM, Gebhart SP, Mixon PS, Clark WS, Phillips LS. Diabetic ketoacidosis in obese African-Americans. Diabetes. 1995;44(7):790-795. [DOI] [PubMed] [Google Scholar]
  • 64. Maldonado  M, Hampe CS, Gaur LK, et al.  Ketosis-prone diabetes: dissection of a heterogeneous syndrome using an immunogenetic and beta-cell functional classification, prospective analysis, and clinical outcomes. J Clin Endocrinol Metab. 2003;88(11):5090-5098. [DOI] [PubMed] [Google Scholar]
  • 65. Mauvais-Jarvis  F, Sobngwi E, Porcher R, et al.  Ketosis-prone type 2 diabetes in patients of sub-Saharan African origin: clinical pathophysiology and natural history of beta-cell dysfunction and insulin resistance. Diabetes. 2004;53(3):645-653. [DOI] [PubMed] [Google Scholar]
  • 66. Ramos-Román  MA, Piñero-Piloña A, Adams-Huet B, Raskin P. Comparison of type 1, type 2, and atypical ketosis-prone diabetes at 4 years of diabetes duration. J Diabetes Complications. 2006;20(3):137-144. [DOI] [PubMed] [Google Scholar]
  • 67. Aizawa  T, Katakura M, Taguchi N, et al.  Ketoacidosis-onset noninsulin dependent diabetes in Japanese subjects. Am J Med Sci. 1995;310(5):198-201. [DOI] [PubMed] [Google Scholar]
  • 68. Wilson  C, Krakoff J, Gohdes D. Ketoacidosis in Apache Indians with non-insulin-dependent diabetes mellitus. Arch Intern Med. 1997;157(18):2098-2100. [PubMed] [Google Scholar]
  • 69. Balasubramanyam  A, Zern JW, Hyman DJ, Pavlik V. New profiles of diabetic ketoacidosis: type 1 vs type 2 diabetes and the effect of ethnicity. Arch Intern Med. 1999;159(19):2317-2322. [DOI] [PubMed] [Google Scholar]
  • 70. Westphal  SA. The occurrence of diabetic ketoacidosis in non-insulin-dependent diabetes and newly diagnosed diabetic adults. Am J Med. 1996;101(1):19-24. [DOI] [PubMed] [Google Scholar]
  • 71. Pinto  ME, Villena JE, Villena AE. Diabetic ketoacidosis in Peruvian patients with type 2 diabetes mellitus. Endocr Pract. 2008;14(4):442-446. [DOI] [PubMed] [Google Scholar]
  • 72. Pitteloud  N, Philippe J. Characteristics of Caucasian type 2 diabetic patients during ketoacidosis and at follow-up. Schweiz Med Wochenschr. 2000;130(16):576-582. [PubMed] [Google Scholar]
  • 73. Jabbar  A, Farooqui K, Habib A, Islam N, Haque N, Akhter J. Clinical characteristics and outcomes of diabetic ketoacidosis in Pakistani adults with Type 2 diabetes mellitus. Diabet Med. 2004;21(8):920-923. [DOI] [PubMed] [Google Scholar]
  • 74. Gupta  RD, Ramachandran R, Gangadhara P, et al.  Clinical characteristics, beta-cell dysfunction and treatment outcomes in patients with A-β+ Ketosis-Prone Diabetes (KPD): The first identified cohort amongst Asian Indians. J Diabetes Complications. 2017;31(9):1401-1407. [DOI] [PubMed] [Google Scholar]
  • 75. Tan  KC, Mackay IR, Zimmet PZ, Hawkins BR, Lam KS. Metabolic and immunologic features of Chinese patients with atypical diabetes mellitus. Diabetes Care. 2000;23(3):335-338. [DOI] [PubMed] [Google Scholar]
  • 76. Kim  MK, Lee SH, Kim JH, et al.  Clinical characteristics of Korean patients with new-onset diabetes presenting with diabetic ketoacidosis. Diabetes Res Clin Pract. 2009;85(1):e8-11. [DOI] [PubMed] [Google Scholar]
  • 77. Brooks-Worrell  BM, Iyer D, Coraza I, et al.  Islet-specific T-cell responses and proinflammatory monocytes define subtypes of autoantibody-negative ketosis-prone diabetes. Diabetes Care. 2013;36(12):4098-4103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Nalini  R, Ozer K, Maldonado M, et al.  Presence or absence of a known diabetic ketoacidosis precipitant defines distinct syndromes of “A-β+” ketosis-prone diabetes based on long-term β-cell function, human leukocyte antigen class II alleles, and sex predilection. Metabolism. 2010;59(10):1448-1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Patel  SG, Hsu JW, Jahoor F, et al.  Pathogenesis of Aβ + ketosis-prone diabetes. Diabetes. 2013;62(3):912-922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Jahoor  F, Hsu JW, Mehta PB, et al.  Metabolomic profiling of patients with A-β+ ketosis-prone diabetes during diabetic ketoacidosis. Diabetes. 2021:db210066. doi: 10.2337/db21-0066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Mulukutla  SN, Hsu JW, Gaba R, et al.  Arginine metabolism is altered in adults with A-β + ketosis-prone diabetes. J Nutr. 2018;148(2):185-193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Haaland  WC, Scaduto DI, Maldonado MR, et al.  A-beta-subtype of ketosis-prone diabetes is not predominantly a monogenic diabetic syndrome. Diabetes Care. 2009;32(5):873-877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Redondo  MJ, Rodriguez LM, Escalante M, Smith EO, Balasubramanyam A, Haymond MW. Types of pediatric diabetes mellitus defined by anti-islet autoimmunity and random C-peptide at diagnosis. Pediatr Diabetes. 2013;14(5):333-340. [DOI] [PubMed] [Google Scholar]
  • 84. Oram  RR, Sharp S, Pihoke C, et al.  A Type 1 diabetes genetic risk score combined with clinical features and autoantibodies enables accurate diabetes classification in a racial/ethnically diverse population: the search for diabetes in youth study. Diabetes 2019;68(Suppl. 1). doi: 10.2337/db19-82-OR [DOI] [Google Scholar]
  • 85. Dabelea  D, Pihoker C, Talton JW, et al. ; SEARCH for Diabetes in Youth Study . Etiological approach to characterization of diabetes type: the SEARCH for Diabetes in Youth Study. Diabetes Care. 2011;34(7):1628-1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Siller  AF, Tosur M, Relan S, et al.  Challenges in the diagnosis of diabetes type in pediatrics. Pediatr Diabetes. 2020;21(7):1064-1073. [DOI] [PubMed] [Google Scholar]
  • 87. Pieralice  S, Pozzilli P. Latent autoimmune diabetes in adults: a review on clinical implications and management. Diabetes Metab J. 2018;42(6):451-464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Andersen  MK, Hansen T. Genetic aspects of latent autoimmune diabetes in adults: a mini-review. Curr Diabetes Rev. 2019;15(3):194-198. [DOI] [PubMed] [Google Scholar]
  • 89. Turner  R, Stratton I, Horton V, et al.  UKPDS 25: autoantibodies to islet-cell cytoplasm and glutamic acid decarboxylase for prediction of insulin requirement in type 2 diabetes. UK Prospective Diabetes Study Group. Lancet. 1997;350(9087):1288-1293. [DOI] [PubMed] [Google Scholar]
  • 90. Davis  TM, Wright AD, Mehta ZM, et al.  Islet autoantibodies in clinically diagnosed type 2 diabetes: prevalence and relationship with metabolic control (UKPDS 70). Diabetologia. 2005;48(4):695-702. [DOI] [PubMed] [Google Scholar]
  • 91. Zinman  B, Kahn SE, Haffner SM, O’Neill MC, Heise MA, Freed MI; ADOPT Study Group . Phenotypic characteristics of GAD antibody-positive recently diagnosed patients with type 2 diabetes in North America and Europe. Diabetes. 2004;53(12):3193-3200. [DOI] [PubMed] [Google Scholar]
  • 92. Pilla  SJ, Balasubramanyam A, Knowler WC, et al. ; Look AHEAD Research Group . Islet autoantibody positivity in overweight and obese adults with type 2 diabetes. Autoimmunity. 2018;51(8):408-416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. van Deutekom  AW, Heine RJ, Simsek S. The islet autoantibody titres: their clinical relevance in latent autoimmune diabetes in adults (LADA) and the classification of diabetes mellitus. Diabet Med. 2008;25(2):117-125. [DOI] [PubMed] [Google Scholar]
  • 94. Monge  L, Bruno G, Pinach S, et al.  A clinically orientated approach increases the efficiency of screening for latent autoimmune diabetes in adults (LADA) in a large clinic-based cohort of patients with diabetes onset over 50 years. Diabet Med. 2004;21(5):456-459. [DOI] [PubMed] [Google Scholar]
  • 95. Shah  VN, Yu L. Comment on Liu et al. Latent autoimmune diabetes in adults with low-titer GAD antibodies: similar disease progression with type 2 diabetes: a nationwide, multicenter prospective study (LADA China Study 3). Diabetes Care 2015;38:16-21. Diabetes Care. 2015;38(3):e43. [DOI] [PubMed] [Google Scholar]
  • 96. Liu  L, Li X, Xiang Y, et al. ; LADA China Study Group . Latent autoimmune diabetes in adults with low-titer GAD antibodies: similar disease progression with type 2 diabetes: a nationwide, multicenter prospective study (LADA China Study 3). Diabetes Care. 2015;38(1):16-21. [DOI] [PubMed] [Google Scholar]
  • 97. Brooks-Worrell  B, Hampe C, Hattery EG, et al.  Islet autoimmunity is highly prevalent and associated with diminished beta cell function in type 2 diabetes patients in the GRADE Study [Abstract]. Diabetes. 2020;69(Suppl. 1). doi: 10.2337/db20-254-OR [DOI] [PMC free article] [PubMed]
  • 98. Oram  RA, Patel K, Hill A, et al.  A type 1 diabetes genetic risk score can aid discrimination between type 1 and type 2 diabetes in young adults. Diabetes Care. 2016;39(3):337-344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Sharp  SA, Rich SS, Wood AR, et al.  Development and standardization of an improved type 1 diabetes genetic risk score for use in newborn screening and incident diagnosis. Diabetes Care. 2019;42(2):200-207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Onengut-Gumuscu  S, Chen WM, Robertson CC, et al. ; SEARCH for Diabetes in Youth; Type 1 Diabetes Genetics Consortium . Type 1 diabetes risk in african-ancestry participants and utility of an ancestry-specific genetic risk score. Diabetes Care. 2019;42(3):406-415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Redondo  MJ, Geyer S, Steck AK, et al. ; Type 1 Diabetes TrialNet Study Group . A type 1 diabetes genetic risk score predicts progression of islet autoimmunity and development of type 1 diabetes in individuals at risk. Diabetes Care. 2018;41(9):1887-1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Sosenko  JM, Skyler JS, DiMeglio LA, et al. ; Type 1 Diabetes TrialNet Study Group; Diabetes Prevention Trial-Type 1 Study Group . A new approach for diagnosing type 1 diabetes in autoantibody-positive individuals based on prediction and natural history. Diabetes Care. 2015;38(2):271-276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Redondo  MJ, Nathan BM, Jacobsen LM, et al. ; Type 1 diabetes TrialNet Study Group . Index60 as an additional diagnostic criterion for type 1 diabetes. Diabetologia. 2021;64(4):836-844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Leslie  RD, Kolb H, Schloot NC, et al.  Diabetes classification: grey zones, sound and smoke: action LADA 1. Diabetes Metab Res Rev. 2008;24(7):511-519. [DOI] [PubMed] [Google Scholar]
  • 105. Udler  MS. Type 2 diabetes: multiple genes, multiple diseases. Curr Diab Rep. 2019;19(8):55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. McCarthy  MI. Painting a new picture of personalised medicine for diabetes. Diabetologia. 2017;60(5):793-799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Wasserfall  C, Nead K, Mathews C, Atkinson MA. The threshold hypothesis: solving the equation of nurture vs nature in type 1 diabetes. Diabetologia. 2011;54(9):2232-2236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Redondo  MJ, Hagopian WA, Oram R, et al.  The clinical consequences of heterogeneity within and between different diabetes types. Diabetologia. 2020;63(10):2040-2048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Ferrara  CT, Geyer SM, Liu YF, et al. ; Type 1 Diabetes TrialNet Study Group . Excess BMI in childhood: a modifiable risk factor for type 1 diabetes development? Diabetes Care. 2017;40(5):698-701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Ferrara  CT, Geyer SM, Evans-Molina C, et al. ; Type 1 Diabetes TrialNet Study Group . The role of age and excess body mass index in progression to type 1 diabetes in at-risk adults. J Clin Endocrinol Metab. 2017;102(12):4596-4603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Ferrara-Cook  C, Geyer SM, Evans-Molina C, et al. ; Type 1 Diabetes TrialNet Study Group . Excess BMI accelerates islet autoimmunity in older children and adolescents. Diabetes Care. 2020;43(3):580-587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Redondo  MJ, Muniz J, Rodriguez LM, et al.  Association of TCF7L2 variation with single islet autoantibody expression in children with type 1 diabetes. BMJ Open Diabetes Res Care. 2014;2(1):e000008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Redondo  MJ, Grant SF, Davis A, Greenbaum C; T1D Exchange Biobank . Dissecting heterogeneity in paediatric Type 1 diabetes: association of TCF7L2 rs7903146 TT and low-risk human leukocyte antigen (HLA) genotypes. Diabet Med. 2017;34(2):286-290. [DOI] [PubMed] [Google Scholar]
  • 114. Redondo  MJ, Geyer S, Steck AK, et al. ; Type 1 Diabetes TrialNet Study Group . TCF7L2 genetic variants contribute to phenotypic heterogeneity of type 1 diabetes. Diabetes Care. 2018;41(2):311-317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Redondo  MJ, Sosenko J, Libman I, et al.  Single islet autoantibody at diagnosis of clinical type 1 diabetes is associated with older age and insulin resistance. J Clin Endocrinol Metab. 2020;105(5):1629-1640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Redondo  MJ, Evans-Molina C, Steck AK, Atkinson MA, Sosenko J. The influence of type 2 diabetes-associated factors on type 1 diabetes. Diabetes Care. 2019;42(8):1357-1364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Sosenko  JM, Skyler JS, Mahon J, et al. ; Type 1 Diabetes TrialNet and Diabetes Prevention Trial-Type 1 Study Groups . Validation of the diabetes prevention trial-type 1 risk score in the TrialNet natural history study. Diabetes Care. 2011;34(8):1785-1787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Cree-Green  M, Bergman BC, Cengiz E, et al.  Metformin improves peripheral insulin sensitivity in youth with type 1 diabetes. J Clin Endocrinol Metab. 2019;104(8):3265-3278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Balasubramanyam  A. Defining and classifying new subgroups of diabetes. Annu Rev Med. 2021;72:63-74. [DOI] [PubMed] [Google Scholar]
  • 120. Anjana  RM, Baskar V, Nair ATN, et al.  Novel subgroups of type 2 diabetes and their association with microvascular outcomes in an Asian Indian population: a data-driven cluster analysis: the INSPIRED study. BMJ Open Diabetes Res Care. 2020;8(1):e001506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Fang  ME, Rosenfeld JA, Redondo MJ, et al. 1136-P: Utility of mining a genetic diagnostic lab for atypical diabetes cases. Abstract presented at the American Diabetes Association Scientific Meeting June 2021. Diabetes. 2021;70(Suppl 1): 1136-P. doi:10.2337/db21-1136-P [Google Scholar]
  • 122. Ahlqvist  E, Storm P, Käräjämäki A, et al.  Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol. 2018;6(5):361-369. [DOI] [PubMed] [Google Scholar]
  • 123. Udler  MS, Kim J, von Grotthuss M, et al. ; Christopher D. Anderson on behalf of METASTROKE and the ISGC. Type 2 diabetes genetic loci informed by multi-trait associations point to disease mechanisms and subtypes: a soft clustering analysis. Plos Med. 2018;15(9):e1002654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Bancks  MP, Chen H, Balasubramanyam A, et al. ; Look AHEAD Research Group . Type 2 diabetes subgroups, risk for complications, and differential effects due to an intensive lifestyle intervention. Diabetes Care. 2021;44(5):1203-1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Wagner  R, Heni M, Tabák AG, et al.  Pathophysiology-based subphenotyping of individuals at elevated risk for type 2 diabetes. Nat Med. 2021;27(1):49-57. [DOI] [PubMed] [Google Scholar]
  • 126. Astudillo  M, Tosur M, Siller A, Balasubramanyam A, Redondo MJ. 1286-P: Identification of atypical pediatric diabetes using electronic medical records (EMR). Diabetes. 2020;69(Suppl 1). doi: 10.2337/db20-1286-p [DOI] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.


Articles from The Journal of Clinical Endocrinology and Metabolism are provided here courtesy of The Endocrine Society

RESOURCES