Next Article in Journal
Key Questions on the Long-Term Utility of Methylphenidate in Paediatric Brain Tumour Survivorship: A Retrospective Clinical Case Series
Previous Article in Journal
Atraumatic Limping Child, a Challenge for Pediatricians: An Observational Age-Related Study in a Pediatric Emergency Department
Previous Article in Special Issue
The COVID-19 Pandemic Increased the Incidence of New-Onset Type One Diabetes in Children
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Approaches to Measuring Beta Cell Reserve and Defining Partial Clinical Remission in Paediatric Type 1 Diabetes

by
Elaine C. Kennedy
1,2 and
Colin P. Hawkes
1,2,3,4,*
1
Department of Paediatrics and Child Health, University College Cork, T12 DC4A Cork, Ireland
2
INFANT Research Centre, University College Cork, T12 DC4A Cork, Ireland
3
Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
4
Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
*
Author to whom correspondence should be addressed.
Children 2024, 11(2), 186; https://doi.org/10.3390/children11020186
Submission received: 27 December 2023 / Revised: 26 January 2024 / Accepted: 1 February 2024 / Published: 2 February 2024
(This article belongs to the Special Issue Advances in Childhood Diabetes)

Abstract

:
Context: Type 1 diabetes (T1D) results from the autoimmune T-cell mediated destruction of pancreatic beta cells leading to insufficient insulin secretion. At the time of diagnosis of T1D, there is residual beta cell function that declines over the subsequent months to years. Recent interventions have been approved to preserve beta cell function in evolving T1D. Objective: The aim of this review is to summarise the approaches used to assess residual beta cell function in evolving T1D, and to highlight potential future directions. Methods: Studies including subjects aged 0 to 18 years were included in this review. The following search terms were used; “(type 1 diabetes) and (partial remission)” and “(type 1 diabetes) and (honeymoon)”. References of included studies were reviewed to determine if additional relevant studies were eligible. Results: There are numerous approaches to quantifying beta cell reserve in evolving T1D. These include c-peptide measurement after a mixed meal or glucagon stimuli, fasting c-peptide, the urinary c-peptide/creatinine ratio, insulin dose-adjusted haemoglobin A1c, and other clinical models to estimate beta cell function. Other biomarkers may have a role, including the proinsulin/c-peptide ratio, cytokines, and microRNA. Studies using thresholds to determine if residual beta cell function is present often differ in values used to define remission. Conclusions: As interventions are approved to preserve beta cell function, it will become increasingly necessary to quantify residual beta cell function in research and clinical contexts. In this report, we have highlighted the strengths and limitations of the current approaches.

1. Background

Type 1 diabetes (T1D) results from the autoimmune T-cell-mediated destruction of pancreatic beta cells leading to insufficient insulin secretion. Four stages of clinical progression have been described as the beta cell reserves deplete. There is normal glucose tolerance at the first stage, with subsequent increasing dysglycemia and an absence of endogenous insulin secretion at the final stage [1].
At the time of diagnosis of T1D, there is residual beta cell function that declines over the subsequent months to years [2,3], with significant variation in trajectory between patients [4,5]. Four years after diagnosis of T1D, approximately 30% of patients no longer have detectable beta cell reserve, compared with 2% at the end of the first year [6]. These residual functional pancreatic beta cells continue to produce endogenous insulin, reducing the requirement for exogenous insulin administration. For some patients, beta cell decline is incomplete, and a low level of insulin secretion is maintained throughout life. In the short-term, residual beta cell function is associated with a reduced risk of hypoglycaemia [7], and in the longer term, reduced rates of T1D complications are seen [8,9,10].
Many interventions to delay disease progression have been studied and teplizumab, a humanised anti-CD3 monoclonal antibody, has recently been approved as a disease-modifying medication that can slow the development of T1D. This is approved in individuals aged 8 years or older with two or more detectable diabetes-related autoantibodies, as well as evidence of preclinical hyperglycaemia [11,12]. Recent studies also suggest some preservation of beta cell function when teplizumab is used in patients with newly diagnosed T1D [13,14]. However, clinical outcomes including glycaemic control and insulin doses were not impacted by teplizumab treatment in this population [13].
Given the availability of beta cell-preserving interventions in T1D, the ability to accurately assess beta cell function is critical to assess treatment efficacy. Various approaches to quantifying beta cell function have been described clinically and in research, including stimulated and single c-peptide measurement, as well as mathematical models including insulin-dose-adjusted glycosylated haemoglobin A1c (HbA1c) estimation. The aim of this review is to summarise the approaches used to assess residual beta cell function in evolving T1D, and to highlight potential future directions.

2. Search Methodology

A PubMed search was performed on 3/1/23. The terms “(type 1 diabetes) and (partial remission)” and “(type 1 diabetes) and (honeymoon)” were included. Studies were limited to those involving individuals aged 0–18 years. References of included articles were reviewed and relevant articles were also included. Therefore, some studies relating to an adult population may have been included if deemed to be relevant to the paediatric literature by the authors. In each study, the methodology of assessment and definition of partial remission were extracted. In studies where new approaches were validated successfully against prior methodologies, both definitions were included. Where novel approaches were not validated, they were summarised separately.

3. Results

A total of 145 studies were identified by our search criteria, and 90 of these were included in our review. Examples of exclusion criteria include if the manuscript did not define partial clinical remission, or if we were unable to access the full paper and the definition of remission was not included in the abstract. The included studies describe 7 different methodologies for defining clinical partial remission. In studies using the same methods to assess for residual function, the threshold used to define remission often varied (Table 1).

4. Approach 1: C-peptide Measurement

Directly measuring circulating insulin levels to estimate beta cell reserve in T1D is not feasible for a number of reasons. There can be assay cross-reactivity between endogenous and exogenous insulin [101], endogenous insulin has a short half-life [102] and up to 80% of secreted insulin is excreted through hepatic first-pass metabolism [101,103,104,105]. Insulin and c-peptide are secreted in an equimolar ratio by the pancreatic beta cells following cleavage of proinsulin. Circulating c-peptide levels subsequently reflect insulin production [103].
C-peptide has physiological properties that make it a more suitable marker of beta cell reserve than insulin. C-peptide has a longer half-life, undergoes negligible hepatic clearance, and is not detected by conventional insulin assays, thereby permitting its use in patients using exogenous insulin [106,107]. C-peptide can be measured in both serum and urine [106,108,109,110]. Although some newer assays use monoclonal antibodies and report improved reproducibility, multiple c-peptide assays are available, and this can affect the comparability of results [106]. Serum samples can be fasting, or stimulated [106], and both approaches have been used to estimate beta cell reserve.

4.1. 1a. Stimulated C-peptide

C-peptide levels measured after a stimulus helps to directly quantify beta cell reserve. Compared with fasting c-peptide measurements, stimulated tests elicit measurable c-peptide responses even in those who have undetectable fasting c-peptide concentrations [111]. Both MMTT and glucagon stimulation tests have demonstrated reproducibility [111]. MMTT is widely used in clinical trials to estimate beta cell stimulation and involves an overnight fast followed by IV cannulation, ingestion of a standardised liquid meal and repeated phlebotomy over a two to four hour period [84,111,112,113,114,115]. Peak response usually occurs at 90 min, and c-peptide levels return to baseline at 120 min. The AUC c-peptide or peak stimulated c-peptide measured during an MMTT are generally considered the preferred methods for measuring beta cell function in interventional trials related to T1D [110,112,116]. In clinical settings, it is not a practical method of regularly measuring beta cell reserve, especially in the paediatric population, where venepuncture can be more technically challenging and associated with distress for the child [110].
Intravenous glucagon is an alternative stimulus for c-peptide secretion to the mixed meal. Following glucagon administration, c-peptide is measured over the following ten minutes. Both the mixed meal and glucagon have reproducible results when performed in the same patient 3 to 10 days apart, but the MMTT is more reproducible. Furthermore, peak c-peptide levels are higher following the MMTT [111]. Compared to the MMTT, patients described more nausea with the glucagon stimulation test, but the shorter duration of the study may be advantageous [111].
Single c-peptide measurement at 90 min following an MMTT (90CP) strongly correlates with AUC c-peptide [110]. From a practical standpoint, this c-peptide measurement at 90 min still requires the same pre-MMTT preparation and a longer test time for the patient compared with a fasting c-peptide sample. However, it requires fewer blood samples, thereby reducing cost and test duration.

4.2. 1b. Fasting C-peptide

Fasting c-peptide levels correlate with other measures of beta cell reserve, including area under the curve (AUC) c-peptide, following an MMTT [110]. Fasting c-peptide, corrected for fasting glucose, has also been shown to correlate strongly with AUC c-peptide. This has been suggested as a practical alternative to AUC c-peptide measured during a MMTT, or to a fasting c-peptide measurement on its own (R2 0.94 vs. 0.88) [112]. However, the sensitivity of fasting c-peptide measurement for residual beta cell function is lower than the MMTT, as many children with undetectable fasting c-peptide levels will have measurable stimulated c-peptide [111].

4.3. 1c. Urinary C-peptide/Creatinine Ratio

The urinary c-peptide-to-creatinine ratio (UCCR) has been used as an alternative to stimulated serum c-peptide measurement to assess residual beta cell function. Urinary measurement 120 min into an MMTT correlates well with the 90 min serum c-peptide concentrations (r = 0.97). Similarly, UCCR measured at home after an evening meal also showed correlation with the 90 min serum c-peptide (r = 0.91) [108]. However, fasting UCCR does not correlate well with AUC c-peptide measured during an MMTT (r = 0.4172) [109]. These results suggest that at-home urinary c-peptide measurements could be used to quantify beta cell function without the time and cost associated with stimulated c-peptide tests, but correlate more closely with stimulated c-peptide levels when measured in a postprandial rather than a fasting state.

5. Approach 2: Clinical Models

The use of stimulation testing to estimate beta cell function is generally confined to clinical research and not routinely used in clinical care. Clinical models that estimate beta cell reserve without the use of stimulated tests have been used as surrogate markers of endogenous insulin secretion. Various components have been suggested as useful contributors to these clinical models.

5.1. 2a Insulin Dose-Adjusted A1C

Exogenous insulin requirements increase as T1D progresses and beta cell reserve diminishes. Total daily dose of insulin (TDD) has been used as a surrogate marker of beta cell function, but TDD needs to account for glycaemic control if this measure is to be used. TDD is often used in conjunction with HbA1c as a means of estimating if a patient is in ‘honeymoon’ or not (Table 1) [22,24,26,117,118]. HbA1c is limited in its ability to estimate beta cell function in the context of evolving deficiency and it does not represent real-time beta cell function, rather an estimate of function over the previous three months [3]. Consequently, variation in HbA1c-adjusted insulin requirement values may not be as reflective of endogenous insulin production as measured through stimulated c-peptide tests [119].
Insulin dose-adjusted A1C (IDAA1C) [5] is the most commonly used clinical method to estimate residual beta cell function at present (Table 1). This model uses current insulin TDD and HbA1c levels to predict if a patient is in remission, as defined by a stimulated c-peptide > 300 pmol/L. While it is suggested that IDAA1C is an alternative to directly measuring c-peptide [5], it underestimates the proportion of patients who have a stimulated c-peptide > 200 pmol/L, especially in a paediatric population [6].

5.2. 2b Model-Estimated Average Plasma C-peptide Concentration

The model-Estimated Average Plasma C-peptide Concentration (CPEST) is a suggested clinical model which uses routinely measured clinical parameters from a single timepoint to estimate beta cell function. The model includes disease duration, body mass index, insulin dose, HbA1c, fasting plasma c-peptide and fasting plasma glucose to estimate the average plasma c-peptide result from an MMTT [120]. Favourable results are reported compared with the previously described IDAA1C (area under ROC 0.89, 95% CI 0.87, 0.92 vs. area under ROC 0.72, 95% CI 0.68, 0.76, respectively). This model has been validated as a potential substitute for simulated c-peptide testing, with a strong correlation between this model and the AUC of meal-stimulated c-peptide (Spearman’s R = 0.911, 95% CI 0.892, 0.926). This is favourable when compared to the correlation between the previously described IDAA1C and the AUC of meal-stimulated c-peptide (Spearman’s R = −0.555, 95% CI −0.619, −0.484) [121]. This model has therefore been suggested as an alternative to stimulated c-peptide testing in future interventional trials [121].

5.3. 2c Model-Estimated Stimulated Peak C-peptide Concentration

This third clinical model to estimate beta cell reserve includes age, body mass index, gender, HbA1c and insulin dose to predict 90 min stimulated c-peptide results [94]. Instead of including c-peptide measurement, which is not routinely measured in children with T1D, this model includes variables which contribute to insulin sensitivity as a surrogate for a real-time c-peptide levels. This acknowledges that c-peptide measurement reflects both the secretory function of beta cells and insulin sensitivity. This model is also reported to have a better predictive value than the IDAA1C model in estimating the 90 min stimulated c-peptide result (adjusted R2 = 0.63, p < 0.0001 vs. R2 = 0.37, p < 0.0001, respectively). When validated in a larger clinical cohort, the model was not as accurate in predicting measured c-peptide levels. When estimated and stimulated 90 min c-peptide levels obtained at 6 months and 12 months post diagnosis were compared, R2 values were 0.36 and 0.37, respectively. When compared to measured c-peptide levels, this model underestimates higher c-peptide levels and overestimates lower levels. The aim of this model is not to replace stimulated c-peptide tests in clinical trials, but to offer a practical approach to estimating beta cell reserve in clinical situations [94].

6. Approach 3: Other Biomarkers

6.1. 3a Proinsulin/C-peptide Ratio

C-peptide and insulin are secreted in equal molar amounts following cleavage of proinsulin. The proinsulin/c-peptide ratio (PI:C) has been used as a biomarker of beta cell stress [122,123]. Fasting and MMTT-stimulated PI:C increased from baseline measurement early in T1D diagnosis to measurements at 12 months; demonstrating increasing beta cell stress. One year after diagnosis, those with an IDAA1C > 9 (i.e., considered not be in remission) had a higher PI:C. Patients who had a higher PI:C at baseline were shown to have a greater decline in their c-peptide over the first year of disease [123]. Although these results may signify beta cell stress, PI:C has not been used to define a partial clinical remission to date.

6.2. 3b Cytokines

Cytokines have been investigated for their potential use as biomarkers of beta cell function. Tumour necrosis factor (TNF)-α, interleukin (IL)-2 and IL-6 have been found to be inversely correlated with stimulated c-peptide levels. TNF-α and IL-10 levels measured early in the disease process may also correlate with stimulated c-peptide levels at 6 months, and have therefore been suggested as possible biomarkers of beta cell reserve, which can be measured in fasting plasma samples [20]. IL-8 has also been suggested as a potential marker of clinical remission in patients with new-onset T1D [59].
Adipokines have also been investigated for their use in quantifying beta cell reserve. Serum leptin and resistin levels positively correlate with fasting c-peptide and MMTT-simulated c-peptide levels [124]. To date, prospective studies have not utilised cytokines as the primary measure of beta cell function.

6.3. 3c MicroRNA

MicroRNA (miR)-204 is another potential biomarker of beta cell function. MiR-204 is be released from apoptosing beta cells and plays a role in insulin production and secretion [125]. Levels are not increased in the serum of patients with T2D or other autoimmune diseases, suggesting that this may be a marker of beta cell decline in this condition. Serum levels of miR-204 inversely correlate with MMTT-stimulated c-peptide AUC [125]. Similar to other potential biomarkers, it has not yet been used in a study to define the honeymoon period.

7. Limitations of Beta Cell Reserve Assessment

7.1. Various Cut off Values

While variable methods to assess beta cell reserve have been described, there are differences noted in their interpretation across each method studied. Mixed meal-stimulated c-peptide thresholds of ≥300 pmol/L at 120 min, >300 pmol/L at 90 min, or AUC ≥ 200 pmol/L have been used to define patients with residual beta cell function. Similarly, some studies using IDAA1C described a value of ≤9 to define the honeymoon period, whereas others used <9, potentially classifying patients differently across studies. Studies using TDD and Hba1c as a marker of remission also use widely variable cut-offs to define partial remission in their population (Table 1).

7.2. Various Immunoassays

In addition to variation in reported thresholds, there are differences in assays used in measuring c-peptide concentrations across studies. These include chemiluminescence immunoassays and fluoroimmunoassays. Despite the use of different assays, some studies have used the same c-peptide threshold to define residual beta cell function (Table 1). This may be a limitation, as differences in reported measurements have been described between assays [126]. Assay standardisation should be considered in future clinical trials describing partial clinical remission.

7.3. Insulin Sensitivity

Insulin resistance in T1D contributes to beta-cell decline [127,128,129] and varies between individuals according to factors including age, sex, puberty and body mass index. Directly measuring c-peptide levels may provide information on both the secretory ability of beta cells and insulin sensitivity of the individual [94], although it is argued that serum c-peptide levels alone do not accurately reflect insulin resistance. If this is the case, defining a partial clinical remission in T1D by measuring stimulated c-peptide alone is insufficient and measures of insulin resistance would be required [128]. In one longitudinal study, the IDAA1C was compared to stimulated c-peptide levels in children with T1D [93]. Partial clinical remission was defined as IDAA1C ≤ 9, and significant beta cell function as stimulated c-peptide > 300 pmol/L. More than a year after T1D diagnosis, almost 55% of the study cohort who had a stimulated c-peptide > 300 pmol/L were not in partial remission as defined by their IDAA1C. This group of patients were reported to have a significantly lower insulin sensitivity (p < 0.001), as measured by an insulin sensitivity score developed in 2011 to estimate insulin sensitivity [130].
A large cohort study found that children diagnosed with T1D at age 5 years or younger were significantly less likely to have a partial remission period compared to children who were older at the time they were diagnosed [70]. They described this finding as biphasic. Although early in the study it appeared that younger children had a more aggressive disease course with lower probability of remission, a greater proportion of this group of younger patients were in remission compared to their older peers at the end of the 6-year observation. Their explanation for this finding was linked to insulin sensitivity. Older children may have had a less aggressive disease course, but fewer of them met the IDAA1C criteria for remission because insulin sensitivity is reduced with puberty. Children diagnosed at a younger age were more likely to still be prepubertal at the end of the 6-year study.

7.4. Patient Population

Some methods of measuring beta cell reserve are not as practical for a paediatric population as in adults. The MMTT can take up to four hours and involves the placement of an intravenous cannula for blood sampling, which can be problematic in the paediatric population [110]. Similarly, nausea has been described as a common adverse effect for paediatric patients undergoing glucagon stimulation tests, compared with older patients [111]. These practical issues may also complicate translation of these research tests to clinical practice.

8. Conclusions

The emergence of approved therapies that may modify the rate of beta cell decline in T1D places a renewed focus on the need to quantify this trajectory. In this review, we have highlighted the various models that have been used to describe beta cell reserve and provided comparative data to guide their use. AUC c-peptide and peak-stimulated c-peptide levels are the most reproducible measures of beta cell function, and limitations of clinical models should be considered when used.
The definition of partial remission varies significantly between studies using the same modality to assess beta cell reserve, limiting comparisons between results across studies. Clinical models that do not necessitate prolonged, expensive testing are more practical in clinical settings where clinicians want to establish if patients are in remission or not, and are used most frequently at present. However, these approaches do not correlate closely with stimulated c-peptide measurement [120,121].
Future directions may include the use of continuous glucose monitor (CGM) data to highlight patterns associated with the remission. The utility of IDAA1C may be limited by the duration of glycaemic control reflected by HbA1c measurement. However, real-time CGM data combined with insulin doses and glycaemic variation may reflect active beta cell reserve more accurately [56,67]. Biomarkers, especially those related to beta cell stress, may also have a role in assessing effects of medical interventions to preserve beta cell function.
This review highlights the wide variation in defining partial remission in T1D. With treatments such as teplizumab now approved to protect beta cells, it is essential that an agreed definition of remission is used. This would ensure the reported efficacy of potential treatments is based on a standardised measurement.

Author Contributions

E.C.K. and C.P.H. designed this study. E.C.K. prepared the first draft and performed the literature review. C.P.H. edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by the INFANT Research Centre at University College Cork and APC Microbiome Ireland at University College Cork.

Institutional Review Board Statement

This study was exempt from ethical review.

Conflicts of Interest

There are no conflicts of interests to disclose. E.C.K. and C.P.H. have no financial relationships relevant to this article to disclose.

Abbreviations

AUCArea Under the Curve
CGMContinuous Glucose Monitor
CPESTModel-Estimated Average Plasma C-peptide Concentration
HbA1cglycosylated haemoglobin A1c
IDAA1CInsulin Dose-adjusted A1C
MMTTMixed Meal Tolerance Test
PI:CProinsulin/C-peptide ratio
TNFTumour Necrosis Factor
ILInterleukin
T1DType 1 Diabetes
TDDTotal Daily Dose
UCCRUrinary C-Peptide/Creatinine Ratio

References

  1. Couper, J.J.; Haller, M.J.; Greenbaum, C.J.; Ziegler, A.G.; Wherrett, D.K.; Knip, M.; Craig, M.E. ISPAD Clinical Practice Consensus Guidelines 2018: Stages of type 1 diabetes in children and adolescents. Pediatr. Diabetes 2018, 19 (Suppl. S27), 20–27. [Google Scholar] [CrossRef]
  2. Fonolleda, M.; Murillo, M.; Vazquez, F.; Bel, J.; Vives-Pi, M. Remission Phase in Paediatric Type 1 Diabetes: New Understanding and Emerging Biomarkers. Horm. Res. Paediatr. 2017, 88, 307–315. [Google Scholar] [CrossRef]
  3. Zhong, T.; Tang, R.; Gong, S.; Li, J.; Li, X.; Zhou, Z. The remission phase in type 1 diabetes: Changing epidemiology, definitions, and emerging immuno-metabolic mechanisms. Diabetes Metab. Res. Rev. 2020, 36, e3207. [Google Scholar] [CrossRef]
  4. Scholin, A.; Berne, C.; Schvarcz, E.; Karlsson, F.A.; Bjork, E. Factors predicting clinical remission in adult patients with type 1 diabetes. J. Intern. Med. 1999, 245, 155–162. [Google Scholar] [CrossRef]
  5. Mortensen, H.B.; Hougaard, P.; Swift, P.; Hansen, L.; Holl, R.W.; Hoey, H.; Bjoerndalen, H.; De Beaufort, C.; Chiarelli, F.; Danne, T.; et al. New definition for the partial remission period in children and adolescents with type 1 diabetes. Diabetes Care 2009, 32, 1384–1390. [Google Scholar] [CrossRef]
  6. Hao, W.; Gitelman, S.; DiMeglio, L.A.; Boulware, D.; Greenbaum, C.J. Fall in C-Peptide During First 4 Years From Diagnosis of Type 1 Diabetes: Variable Relation to Age, HbA1c, and Insulin Dose. Diabetes Care 2016, 39, 1664–1670. [Google Scholar] [CrossRef]
  7. Sørensen, J.S.; Johannesen, J.; Pociot, F.; Kristensen, K.; Thomsen, J.; Hertel, N.T.; Kjaersgaard, P.; Brorsson, C.; Birkebaek, N.H.; Danish Society for Diabetes in Childhood and Adolescence. Residual β-Cell function 3-6 years after onset of type 1 diabetes reduces risk of severe hypoglycemia in children and adolescents. Diabetes Care 2013, 36, 3454–3459. [Google Scholar] [CrossRef]
  8. Keenan, H.A.; Sun, J.K.; Levine, J.; Doria, A.; Aiello, L.P.; Eisenbarth, G.; Bonner-Weir, S.; King, G.L. Residual insulin production and pancreatic ss-cell turnover after 50 years of diabetes: Joslin Medalist Study. Diabetes 2010, 59, 2846–2853. [Google Scholar] [CrossRef] [PubMed]
  9. Panero, F.; Novelli, G.; Zucco, C.; Fornengo, P.; Perotto, M.; Segre, O.; Grassi, G.; Cavallo-Perin, P.; Bruno, G. Fasting plasma C-peptide and micro- and macrovascular complications in a large clinic-based cohort of type 1 diabetic patients. Diabetes Care 2009, 32, 301–305. [Google Scholar] [CrossRef] [PubMed]
  10. Nakanishi, K.; Watanabe, C. Rate of beta-cell destruction in type 1 diabetes influences the development of diabetic retinopathy: Protective effect of residual beta-cell function for more than 10 years. J. Clin. Endocrinol. Metab. 2008, 93, 4759–4766. [Google Scholar] [CrossRef] [PubMed]
  11. Hirsch, J.S. FDA approves teplizumab: A milestone in type 1 diabetes. Lancet Diabetes Endocrinol. 2023, 11, 18. [Google Scholar] [CrossRef] [PubMed]
  12. Herold, K.C.; Bundy, B.N.; Long, S.A.; Bluestone, J.A.; DiMeglio, L.A.; Dufort, M.J.; Gitelman, S.E.; Gottlieb, P.A.; Krischer, J.P.; Linsley, P.S.; et al. An Anti-CD3 Antibody, Teplizumab, in Relatives at Risk for Type 1 Diabetes. N. Engl. J. Med. 2019, 381, 603–613. [Google Scholar] [CrossRef]
  13. Ramos, E.L.; Dayan, C.M.; Chatenoud, L.; Sumnik, Z.; Simmons, K.M.; Szypowska, A.; Gitelman, S.E.; Knecht, L.A.; Niemoeller, E.; Tian, W.; et al. Teplizumab and beta-Cell Function in Newly Diagnosed Type 1 Diabetes. N. Engl. J. Med. 2023, 389, 2151–2161. [Google Scholar] [CrossRef]
  14. Herold, K.C.; Gitelman, S.E.; Gottlieb, P.A.; Knecht, L.A.; Raymond, R.; Ramos, E.L. Teplizumab: A Disease-Modifying Therapy for Type 1 Diabetes That Preserves beta-Cell Function. Diabetes Care 2023, 46, 1848–1856. [Google Scholar] [CrossRef] [PubMed]
  15. Li, X.; Zhong, T.; Tang, R.; Wu, C.; Xie, Y.; Liu, F.; Zhou, Z. PD-1 and PD-L1 Expression in Peripheral CD4/CD8+ T Cells Is Restored in the Partial Remission Phase in Type 1 Diabetes. Journal Clin. Endocrinol. Metab. 2020, 105, 1947–1956. [Google Scholar] [CrossRef]
  16. Zhong, T.; Tang, R.; Xie, Y.; Liu, F.; Li, X.; Zhou, Z. Frequency, clinical characteristics, and determinants of partial remission in type 1 diabetes: Different patterns in children and adults. J. Diabetes 2020, 12, 761–768. [Google Scholar] [CrossRef] [PubMed]
  17. Chen, Y.; Xia, Y.; Xie, Z.; Zhong, T.; Tang, R.; Li, X.; Zhou, Z. The Unfavorable Impact of DR9/DR9 Genotype on the Frequency and Quality of Partial Remission in Type 1 Diabetes. J. Clin. Endocrinol. Metab. 2022, 107, e293–e302. [Google Scholar] [CrossRef]
  18. Andersen, M.L.C.M.; Hougaard, P.; Pörksen, S.; Nielsen, L.B.; Fredheim, S.; Svensson, J.; Thomsen, J.; Vikre-Jørgensen, J.; Hertel, T.; Petersen, J.S.; et al. Partial remission definition: Validation based on the insulin dose-adjusted HbA1c (IDAA1C) in 129 Danish children with new-onset type 1 diabetes. Pediatr. Diabetes 2014, 15, 469–476. [Google Scholar] [CrossRef]
  19. Madsen, J.O.B.; Herskin, C.W.; Zerahn, B.; Jensen, A.K.; Jørgensen, N.R.; Olsen, B.S.; Svensson, J.; Pociot, F.; Johannesen, J. Bone turnover markers during the remission phase in children and adolescents with type 1 diabetes. Pediatr. Diabetes 2020, 21, 366–376. [Google Scholar] [CrossRef]
  20. Overgaard, A.J.; Madsen, J.O.B.; Pociot, F.; Johannesen, J.; Størling, J. Systemic TNFα correlates with residual β-cell function in children and adolescents newly diagnosed with type 1 diabetes. BMC Pediatr. 2020, 20, 446. [Google Scholar] [CrossRef]
  21. Kara, Ö.; Esen, İ.; Tepe, D. Factors Influencing Frequency and Duration of Remission in Children and Adolescents Newly Diagnosed with Type 1 Diabetes. Med. Sci. Monit. 2018, 24, 5996–6001. [Google Scholar] [CrossRef]
  22. Bowden, S.A.; Duck, M.M.; Hoffman, R.P. Young children (<5 yr) and adolescents (>12 yr) with type 1 diabetes mellitus have low rate of partial remission: Diabetic ketoacidosis is an important risk factor. Pediatr. Diabetes 2008, 9 Pt 1, 197–201. [Google Scholar]
  23. Stuart, A.A.V.; Jager, W.; Klein, M.R.; Teklenburg, G.; Nuboer, R.; Hoorweg, J.J.G.; Vroede, M.A.M.J.; Kruijff, I.; Fick, M.; Schroor, E.J.; et al. Recognition of heat shock protein 60 epitopes in children with type 1 diabetes. Diabetes Metab. Res. Rev. 2012, 28, 527–534. [Google Scholar] [CrossRef] [PubMed]
  24. Ortqvist, E.; Falorni, A.; Scheynius, A.; Persson, B.; Lernmark, A. Age governs gender-dependent islet cell autoreactivity and predicts the clinical course in childhood IDDM. Acta Paediatr. 1997, 86, 1166–1171. [Google Scholar] [CrossRef]
  25. Meng, X.; Gong, C.; Cao, B.; Peng, X.; Wu, D.; Gu, Y.; Wei, L.; Liang, X.; Liu, M.; Li, W.; et al. Glucose fluctuations in association with oxidative stress among children with T1DM: Comparison of different phases. J. Clin. Endocrinol. Metab. 2015, 100, 1828–1836. [Google Scholar] [CrossRef] [PubMed]
  26. Nordwall, M.; Ludvigsson, J. Clinical manifestations and beta cell function in Swedish diabetic children have remained unchanged during the last 25 years. Diabetes Metab. Res. Rev. 2008, 24, 472–479. [Google Scholar] [CrossRef]
  27. Araujo, D.B.; Dantas, J.R.; Silva, K.R.; Souto, D.L.; Pereira, M.D.F.C.; Moreira, J.P.; Luiz, R.R.; Claudio-Da-Silva, C.S.; Gabbay, M.A.; Dib, S.A.; et al. Allogenic Adipose Tissue-Derived Stromal/Stem Cells and Vitamin D Supplementation in Patients With Recent-Onset Type 1 Diabetes Mellitus: A 3-Month Follow-Up Pilot Study. Front. Immunol. 2020, 11, 993. [Google Scholar] [CrossRef] [PubMed]
  28. Pyziak, A.; Zmyslowska, A.; Bobeff, K.; Malachowska, B.; Fendler, W.; Wyka, K.; Baranowska-Jazwiecka, A.; Szymanska, M.; Szadkowska, A.; Mlynarski, W. Markers influencing the presence of partial clinical remission in patients with newly diagnosed type 1 diabetes. J. Pediatr. Endocrinol. Metab. 2017, 30, 1147–1153. [Google Scholar] [CrossRef]
  29. Jamiołkowska-Sztabkowska, M.; Grubczak, K.; Starosz, A.; Krętowska-Grunwald, A.; Krętowska, M.; Parfienowicz, Z.; Moniuszko, M.; Bossowski, A.; Głowińska-Olszewska, B. Circulating Hematopoietic (HSC) and Very-Small Embryonic like (VSEL) Stem Cells in Newly Diagnosed Childhood Diabetes type 1—Novel Parameters of Beta Cell Destruction/Regeneration Balance and Possible Prognostic Factors of Future Disease Course. Stem Cell Rev. Rep. 2022, 18, 1657–1667. [Google Scholar] [CrossRef]
  30. Jamiolkowska-Sztabkowska, M.; Glowinska-Olszewska, B.; Luczynski, W.; Konstantynowicz, J.; Bossowski, A. Regular physical activity as a physiological factor contributing to extend partial remission time in children with new onset diabetes mellitus-Two years observation. Pediatr. Diabetes 2020, 21, 800–807. [Google Scholar] [CrossRef]
  31. Jamiołkowska-Sztabkowska, M.; Głowińska-Olszewska, B.; Bossowski, A. C-peptide and residual β-cell function in pediatric diabetes—State of the art. Pediatr. Endocrinol. Diabetes Metab. 2021, 27, 123–133. [Google Scholar] [CrossRef] [PubMed]
  32. de Souza, L.C.V.F.; Kraemer, G.d.C.; Koliski, A.; Carreiro, J.E.; Cat, M.N.L.; De Lacerda, L.; França, S.N. Diabetic Ketoacidosis as the Initial Presentation of Type 1 Diabetes in Children and Adolescents: Epidemiological Study in Southern Brazil. Rev. Paul. Pediatr. 2020, 38, e2018204. [Google Scholar] [CrossRef] [PubMed]
  33. Villalba, A.; Fonolleda, M.; Murillo, M.; Rodriguez-Fernandez, S.; Ampudia, R.-M.; Perna-Barrull, D.; Raina, M.B.; Quirant-Sanchez, B.; Planas, R.; Teniente-Serra, A.; et al. Partial remission and early stages of pediatric type 1 diabetes display immunoregulatory changes. A pilot study. Transl. Res. 2019, 210, 8–25. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, Y.; Gong, C.; Cao, B.; Meng, X.; Wei, L.; Wu, D.; Liang, X.; Li, W.; Liu, M.; Gu, Y.; et al. Influence of initial insulin dosage on blood glucose dynamics of children and adolescents with newly diagnosed type 1 diabetes mellitus. Pediatr. Diabetes 2017, 18, 196–203. [Google Scholar] [CrossRef]
  35. The pediatric diabetes consortium: Improving care of children with type 1 diabetes through collaborative research. Diabetes Technol. Ther. 2010, 12, 685–688. [CrossRef] [PubMed]
  36. Dost, A.; Herbst, A.; Kintzel, K.; Haberland, H.; Roth, C.L.; Gortner, L.; Holl, R.W. Shorter remission period in young versus older children with diabetes mellitus type 1. Exp. Clin. Endocrinol. Diabetes 2007, 115, 33–37. [Google Scholar] [CrossRef] [PubMed]
  37. Chobot, A.; Stompór, J.; Szyda, K.; Sokołowska, M.; Deja, G.; Polańska, J.; Jarosz-Chobot, P. Remission phase in children diagnosed with type 1 diabetes in years 2012 to 2013 in Silesia, Poland: An observational study. Pediatr. Diabetes 2019, 20, 286–292. [Google Scholar] [CrossRef]
  38. Abdul-Rasoul, M.; Habib, H.; Al-Khouly, M. ‘The honeymoon phase’ in children with type 1 diabetes mellitus: Frequency, duration, and influential factors. Pediatr. Diabetes 2006, 7, 101–107. [Google Scholar] [CrossRef]
  39. Humphreys, A.; Bravis, V.; Kaur, A.; Walkey, H.C.; Godsland, I.F.; Misra, S.; Johnston, D.G.; Oliver, N.S. Individual and diabetes presentation characteristics associated with partial remission status in children and adults evaluated up to 12 months following diagnosis of type 1 diabetes: An ADDRESS-2 (After Diagnosis Diabetes Research Support System-2) study analysis. Diabetes Res. Clin. Pract. 2019, 155, 107789. [Google Scholar]
  40. Schloot, N.C.; Hanifi-Moghaddam, P.; Aabenhus-Andersen, N.; Alizadeh, B.Z.; Saha, M.T.; Knip, M.; Devendra, D.; Wilkin, T.; Bonifacio, E.; Roep, B.O.; et al. Association of immune mediators at diagnosis of Type 1 diabetes with later clinical remission. Diabet. Med. 2007, 24, 512–520. [Google Scholar] [CrossRef] [PubMed]
  41. Barone, R.; Procaccini, E.; Chianelli, M.; Annovazzi, A.; Fiore, V.; Hawa, M.; Nardi, G.; Ronga, G.; Pozzilli, P.; Signore, A.; et al. Prognostic relevance of pancreatic uptake of technetium-99m labelled human polyclonal immunoglobulins in patients with type 1 diabetes. Eur. J. Nucl. Med. 1998, 25, 503–508. [Google Scholar] [CrossRef]
  42. Kamado, K.; Fujita, S.; Izumi, K.; Hoshi, M. Remission phase in childhood diabetes--an investigation of summer campers in Japan. Tohoku J. Exp. Med. 1983, 141, 191–198. [Google Scholar] [CrossRef]
  43. Bonfanti, R.; Bognetti, E.; Meschi, F.; Brunelli, A.; Riva, M.C.; Pastore, M.R.; Calori, G.; Chiumello, G. Residual beta-cell function and spontaneous clinical remission in type 1 diabetes mellitus: The role of puberty. Acta Diabetol. 1998, 35, 91–95. [Google Scholar] [CrossRef]
  44. Cook, J.J.; Hudson, I.; Harrison, L.C.; Dean, B.; Colman, P.G.; Werther, G.; Warne, G.L.; Court, J.M. Double-blind controlled trial of azathioprine in children with newly diagnosed type I diabetes. Diabetes 1989, 38, 779–783. [Google Scholar] [CrossRef]
  45. Bober, E.; Dundar, B.; Buyukgebiz, A. Partial remission phase and metabolic control in type 1 diabetes mellitus in children and adolescents. J. Pediatr. Endocrinol. Metab. 2001, 14, 435–441. [Google Scholar] [CrossRef]
  46. Sanda, S.; Roep, B.O.; von Herrath, M. Islet antigen specific IL-10+ immune responses but not CD4+CD25+FoxP3+ cells at diagnosis predict glycemic control in type 1 diabetes. Clin. Immunol. 2008, 127, 138–143. [Google Scholar] [CrossRef]
  47. Kordonouri, O.; Danne, T.; Enders, I.; Weber, B. Does the long-term clinical course of type I diabetes mellitus differ in patients with prepubertal and pubertal onset? Results of the Berlin Retinopathy Study. Eur. J. Pediatr. 1998, 157, 202–207. [Google Scholar] [CrossRef]
  48. Glisic-Milosavljevic, S.; Wang, T.; Koppen, M.; Kramer, J.; Ehlenbach, S.; Waukau, J.; Jailwala, P.; Jana, S.; Alemzadeh, R.; Ghosh, S. Dynamic changes in CD4+ CD25+(high) T cell apoptosis after the diagnosis of type 1 diabetes. Clin. Exp. Immunol. 2007, 150, 75–82. [Google Scholar] [CrossRef] [PubMed]
  49. Muhammad, B.J.; Swift, P.G.; Raymond, N.T.; Botha, J.L. Partial remission phase of diabetes in children younger than age 10 years. Arch. Dis. Child. 1999, 80, 367–369. [Google Scholar] [CrossRef] [PubMed]
  50. Cadario, F.; Savastio, S.; Rizzo, A.M.; Carrera, D.; Bona, G.; Ricordi, C. Can Type 1 diabetes progression be halted? Possible role of high dose vitamin D and omega 3 fatty acids. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1604–1609. [Google Scholar] [PubMed]
  51. Lundberg, R.L.; Marino, K.R.; Jasrotia, A.; Maranda, L.S.; Barton, B.A.; Alonso, L.C.; Nwosu, B.U. Partial clinical remission in type 1 diabetes: A comparison of the accuracy of total daily dose of insulin of <0.3 units/kg/day to the gold standard insulin-dose adjusted hemoglobin A1c of ≤9 for the detection of partial clinical remission. J. Pediatr. Endocrinol. Metab. 2017, 30, 823–830. [Google Scholar]
  52. Nwosu, B.U.; Zhang, B.; Ayyoub, S.S.; Choi, S.; Villalobos-Ortiz, T.R.; Alonso, L.C.; Barton, B.A. Children with type 1 diabetes who experienced a honeymoon phase had significantly lower LDL cholesterol 5 years after diagnosis. PLoS ONE 2018, 13, e0196912. [Google Scholar] [CrossRef]
  53. Feutren, G.; Assan, R.; Karsenty, G.; Du Rostu, H.; Sirmai, J.; Papoz, L.; Vialettes, B.; Vexiau, P.; Rodier, M.; Lallemand, A.; et al. Cyclosporin increases the rate and length of remissions in insulin-dependent diabetes of recent onset. Results of a multicentre double-blind trial. Lancet 1986, 2, 119–124. [Google Scholar] [CrossRef] [PubMed]
  54. Agner, T.; Damm, P.; Binder, C. Remission in IDDM: Prospective study of basal C-peptide and insulin dose in 268 consecutive patients. Diabetes Care 1987, 10, 164–169. [Google Scholar] [CrossRef] [PubMed]
  55. Blair, J.C.; McKay, A.; Ridyard, C.; Thornborough, K.; Bedson, E.; Peak, M.; Didi, M.; Annan, F.; Gregory, J.W.; A Hughes, D.; et al. Continuous subcutaneous insulin infusion versus multiple daily injection regimens in children and young people at diagnosis of type 1 diabetes: Pragmatic randomised controlled trial and economic evaluation. BMJ 2019, 365, l1226. [Google Scholar] [CrossRef] [PubMed]
  56. Pollé, O.G.; Delfosse, A.; Martin, M.; Louis, J.; Gies, I.; Brinker, M.D.; Seret, N.; Lebrethon, M.-C.; Mouraux, T.; Gatto, L.; et al. Glycemic Variability Patterns Strongly Correlate With Partial Remission Status in Children With Newly Diagnosed Type 1 Diabetes. Diabetes Care 2022, 45, 2360–2368. [Google Scholar] [CrossRef] [PubMed]
  57. Addala, A.; Zaharieva, D.P.; Gu, A.J.; Prahalad, P.; Scheinker, D.; Buckingham, B.; Hood, K.K.; Maahs, D.M. Clinically Serious Hypoglycemia Is Rare and Not Associated With Time-in-range in Youth With New-onset Type 1 Diabetes. J. Clin. Endocrinol. Metab. 2021, 106, 3239–3247. [Google Scholar] [CrossRef]
  58. Kingery, S.E.; Wu, Y.L.; Zhou, B.; Hoffman, R.P.; Yu, C.Y. Gene CNVs and protein levels of complement C4A and C4B as novel biomarkers for partial disease remissions in new-onset type 1 diabetes patients. Pediatr. Diabetes 2012, 13, 408–418. [Google Scholar] [CrossRef]
  59. Pyziak-Skupien, A.; Bobeff, K.; Wyka, K.; Banach, K.; Malachowska, B.; Fendler, W.; Szadkowska, A.; Mlynarski, W.; Zmyslowska, A. Fetuin-A and Interleukine-8 in Children with the Clinical Remission of Type 1 Diabetes. Iran J. Immunol. 2020, 17, 144–153. [Google Scholar]
  60. Cadario, F.; Pozzi, E.; Rizzollo, S.; Stracuzzi, M.; Beux, S.; Giorgis, A.; Carrera, D.; Fullin, F.; Riso, S.; Rizzo, A.M.; et al. Vitamin D and ω-3 Supplementations in Mediterranean Diet During the 1st Year of Overt Type 1 Diabetes: A Cohort Study. Nutrients 2019, 11, 2158. [Google Scholar] [CrossRef]
  61. Casas, R.; Dietrich, F.; Barcenilla, H.; Tavira, B.; Wahlberg, J.; Achenbach, P.; Ludvigsson, J. Glutamic Acid Decarboxylase Injection Into Lymph Nodes: Beta Cell Function and Immune Responses in Recent Onset Type 1 Diabetes Patients. Front. Immunol. 2020, 11, 564921. [Google Scholar] [CrossRef]
  62. Gomez-Muñoz, L.; Perna-Barrull, D.; Caroz-Armayones, J.M.; Murillo, M.; Rodriguez-Fernandez, S.; Valls, A.; Vazquez, F.; Perez, J.; Corripio, R.; Castaño, L.; et al. Candidate Biomarkers for the Prediction and Monitoring of Partial Remission in Pediatric Type 1 Diabetes. Front. Immunol. 2022, 13, 825426. [Google Scholar] [CrossRef]
  63. Gomez-Muñoz, L.; Perna-Barrull, D.; Villalba, A.; Rodriguez-Fernandez, S.; Ampudia, R.-M.; Teniente-Serra, A.; Vazquez, F.; Murillo, M.; Perez, J.; Corripio, R.; et al. NK Cell Subsets Changes in Partial Remission and Early Stages of Pediatric Type 1 Diabetes. Front. Immunol. 2020, 11, 611522. [Google Scholar] [CrossRef] [PubMed]
  64. Nwosu, B.U.; Parajuli, S.; Khatri, K.; Jasmin, G.; Al-Halbouni, L.; Lee, A.F. Partial Clinical Remission Reduces Lipid-Based Cardiovascular Risk in Adult Patients With Type 1 Diabetes. Front. Endocrinol. 2021, 12, 705565. [Google Scholar] [CrossRef] [PubMed]
  65. Nwosu, B.U.; Villalobos-Ortiz, T.R.; Jasmin, G.A.; Parajuli, S.; Zitek-Morrison, E.; Barton, B.A. Mechanisms and early patterns of dyslipidemia in pediatric type 1 and type 2 diabetes. J. Pediatr. Endocrinol. Metab. 2020, 33, 1399–1408. [Google Scholar] [CrossRef]
  66. Nwosu, B.U. The Theory of Hyperlipidemic Memory of Type 1 Diabetes. Front. Endocrinol. 2022, 13, 819544. [Google Scholar] [CrossRef] [PubMed]
  67. Yeşiltepe-Mutlu, G.; Çapacı, M.; Can, E.; Gökçe, T.; Bayrakçı, G.; Muradoğlu, S.; İncir, S.; Çakır, E.P.; Hatun, Ş. A comparison of glycemic parameters and their relationship with C-peptide and Proinsulin levels during partial remission and non-remission periods in children with type 1 diabetes mellitus—A cross-sectional study. BMC Endocr. Disord. 2021, 21, 18. [Google Scholar]
  68. Chiavaroli, V.; Derraik, J.G.B.; Jalaludin, M.Y.; Albert, B.B.; Ramkumar, S.; Cutfield, W.S.; Hofman, P.L.; Jefferies, C.A. Partial remission in type 1 diabetes and associated factors: Analysis based on the insulin dose-adjusted hemoglobin A1c in children and adolescents from a regional diabetes center, Auckland, New Zealand. Pediatr. Diabetes 2019, 20, 892–900. [Google Scholar] [CrossRef]
  69. Pecheur, A.; Barrea, T.; Vandooren, V.; Beauloye, V.; Robert, A.; Lysy, P.A. Characteristics and determinants of partial remission in children with type 1 diabetes using the insulin-dose-adjusted A1C definition. J. Diabetes Res. 2014, 2014, 851378. [Google Scholar] [CrossRef] [PubMed]
  70. Nagl, K.; Hermann, J.M.; Plamper, M.; Schröder, C.; Dost, A.; Kordonouri, O.; Rami-Merhar, B.; Holl, R.W. Factors contributing to partial remission in type 1 diabetes: Analysis based on the insulin dose-adjusted HbA1c in 3657 children and adolescents from Germany and Austria. Pediatr. Diabetes 2017, 18, 428–434. [Google Scholar] [CrossRef]
  71. Moya, R.; Robertson, H.K.; Payne, D.; Narsale, A.; Koziol, J.; Davies, J.D. A pilot study showing associations between frequency of CD4(+) memory cell subsets at diagnosis and duration of partial remission in type 1 diabetes. Clin. Immunol. 2016, 166–167, 72–80. [Google Scholar] [CrossRef] [PubMed]
  72. Narsale, A.; Lam, B.; Moya, R.; Lu, T.; Mandelli, A.; Gotuzzo, I.; Pessina, B.; Giamporcaro, G.; Geoffrey, R.; Buchanan, K.; et al. CD4+CD25+CD127hi cell frequency predicts disease progression in type 1 diabetes. JCI Insight 2021, 6, e136114. [Google Scholar] [CrossRef] [PubMed]
  73. Kaas, A.; Max Andersen, M.L.; Fredheim, S.; Hougaard, P.; Buschard, K.; Petersen, J.S.; De Beaufort, C.; Robertson, K.J.; Hansen, L.; Mortensen, H.B.; et al. Proinsulin, GLP-1, and glucagon are associated with partial remission in children and adolescents with newly diagnosed type 1 diabetes. Pediatr. Diabetes 2012, 13, 51–58. [Google Scholar] [CrossRef] [PubMed]
  74. Genzano, C.B.; Bezzecchi, E.; Carnovale, D.; Mandelli, A.; Morotti, E.; Castorani, V.; Favalli, V.; Stabilini, A.; Insalaco, V.; Ragogna, F.; et al. Combined unsupervised and semi-automated supervised analysis of flow cytometry data reveals cellular fingerprint associated with newly diagnosed pediatric type 1 diabetes. Front. Immunol. 2022, 13, 1026416. [Google Scholar] [CrossRef] [PubMed]
  75. Lundgren, M.; Jonsdottir, B.; Elding Larsson, H. Effect of screening for type 1 diabetes on early metabolic control: The DiPiS study. Diabetologia 2019, 62, 53–57. [Google Scholar] [CrossRef] [PubMed]
  76. Klocperk, A.; Petruzelkova, L.; Pavlikova, M.; Rataj, M.; Kayserova, J.; Pruhova, S.; Kolouskova, S.; Sklenarova, J.; Parackova, Z.; Sediva, A.; et al. Changes in innate and adaptive immunity over the first year after the onset of type 1 diabetes. Acta Diabetol. 2020, 57, 297–307. [Google Scholar] [CrossRef]
  77. Lawes, T.; Franklin, V.; Farmer, G. HbA1c tracking and bio-psychosocial determinants of glycaemic control in children and adolescents with type 1 diabetes: Retrospective cohort study and multilevel analysis. Pediatr. Diabetes 2014, 15, 372–383. [Google Scholar] [CrossRef]
  78. Pinckney, A.; Rigby, M.R.; Keyes-Elstein, L.; Soppe, C.L.; Nepom, G.T.; Ehlers, M.R. Correlation Among Hypoglycemia, Glycemic Variability, and C-Peptide Preservation After Alefacept Therapy in Patients with Type 1 Diabetes Mellitus: Analysis of Data from the Immune Tolerance Network T1DAL Trial. Clin. Ther. 2016, 38, 1327–1339. [Google Scholar] [CrossRef]
  79. Cengiz, E.; Cheng, P.; Ruedy, K.J.; Kollman, C.; Tamborlane, W.V.; Klingensmith, G.J.; Gal, R.L.; Silverstein, J.; Lee, J.; Redondo, M.J.; et al. Clinical outcomes in youth beyond the first year of type 1 diabetes: Results of the Pediatric Diabetes Consortium (PDC) type 1 diabetes new onset (NeOn) study. Pediatr. Diabetes 2017, 18, 566–573. [Google Scholar] [CrossRef]
  80. Neylon, O.M.; White, M.; O’Connell, M.A.; Cameron, F.J. Insulin-dose-adjusted HbA1c-defined partial remission phase in a paediatric population--when is the honeymoon over? Diabet. Med. 2013, 30, 627–628. [Google Scholar] [CrossRef]
  81. Moosavi, M.; Seguin, J.; Polychronakos, C. Effect of autoimmunity risk loci on the honeymoon phase in type 1 diabetes. Pediatr. Diabetes 2017, 18, 459–462. [Google Scholar] [CrossRef]
  82. McGill, D.E.; Volkening, L.K.; Pober, D.M.; Muir, A.B.; Young-Hyman, D.L.; Laffel, L.M. Depressive Symptoms at Critical Times in Youth With Type 1 Diabetes: Following Type 1 Diabetes Diagnosis and Insulin Pump Initiation. J. Adolesc. Health 2018, 62, 219–225. [Google Scholar] [CrossRef]
  83. Neuman, V.; Pruhova, S.; Kulich, M.; Kolouskova, S.; Vosahlo, J.; Romanova, M.; Petruzelkova, L.; Obermannova, B.; Funda, D.P.; Cinek, O.; et al. Gluten-free diet in children with recent-onset type 1 diabetes: A 12-month intervention trial. Diabetes Obes. Metab. 2020, 22, 866–872. [Google Scholar] [CrossRef]
  84. Quattrin, T.; Haller, M.J.; Steck, A.K.; Felner, E.I.; Li, Y.; Xia, Y.; Leu, J.H.; Zoka, R.; Hedrick, J.A.; Rigby, M.R.; et al. Golimumab and Beta-Cell Function in Youth with New-Onset Type 1 Diabetes. N. Engl. J. Med. 2020, 383, 2007–2017. [Google Scholar] [CrossRef]
  85. Cabrera, S.M.; Engle, S.; Kaldunski, M.; Jia, S.; Geoffrey, R.; Simpson, P.; Szabo, A.; Speake, C.; Greenbaum, C.J.; Chen, Y.-G.; et al. Innate immune activity as a predictor of persistent insulin secretion and association with responsiveness to CTLA4-Ig treatment in recent-onset type 1 diabetes. Diabetologia 2018, 61, 2356–2370. [Google Scholar] [CrossRef] [PubMed]
  86. Passanisi, S.; Salzano, G.; Gasbarro, A.; Brancati, V.U.; Mondio, M.; Pajno, G.B.; Alibrandi, A.; Lombardo, F. Influence of Age on Partial Clinical Remission among Children with Newly Diagnosed Type 1 Diabetes. Int. J. Environ. Res. Public Health 2020, 17, 4801. [Google Scholar] [CrossRef] [PubMed]
  87. Marino, K.R.; Lundberg, R.L.; Jasrotia, A.; Maranda, L.S.; Thompson, M.J.; Barton, B.A.; Alonso, L.C.; Nwosu, B.U. A predictive model for lack of partial clinical remission in new-onset pediatric type 1 diabetes. PLoS ONE 2017, 12, e0176860. [Google Scholar] [CrossRef] [PubMed]
  88. Camilo, D.S.; Pradella, F.; Paulino, M.F.; Baracat, E.C.E.; Marini, S.H.; Guerra, G.; Pavin, E.J.; Parisi, C.; Longhini, A.L.F.; Marques, S.B.; et al. Partial remission in Brazilian children and adolescents with type 1 diabetes. Association with a haplotype of class II human leukocyte antigen and synthesis of autoantibodies. Pediatr. Diabetes 2020, 21, 606–614. [Google Scholar] [CrossRef] [PubMed]
  89. Nielens, N.; Polle, O.; Robert, A.; Lysy, P.A. Integration of Routine Parameters of Glycemic Variability in a Simple Screening Method for Partial Remission in Children with Type 1 Diabetes. J. Diabetes Res. 2018, 2018, 5936360. [Google Scholar] [CrossRef] [PubMed]
  90. Redondo, M.J.; Libman, I.; Cheng, P.; Kollman, C.; Tosur, M.; Gal, R.L.; Bacha, F.; Klingensmith, G.J.; Clements, M. Racial/Ethnic Minority Youth With Recent-Onset Type 1 Diabetes Have Poor Prognostic Factors. Diabetes Care 2018, 41, 1017–1024. [Google Scholar] [CrossRef] [PubMed]
  91. Franceschi, R.; Cauvin, V.; Stefani, L.; Berchielli, F.; Soffiati, M.; Maines, E. Early Initiation of Intermittently Scanned Continuous Glucose Monitoring in a Pediatric Population With Type 1 Diabetes: A Real World Study. Front. Endocrinol. 2022, 13, 907517. [Google Scholar] [CrossRef]
  92. Cimbek, E.A.; Bozkır, A.; Usta, D.; Beyhun, N.E.; Ökten, A.; Karagüzel, G. Partial remission in children and adolescents with type 1 diabetes: An analysis based on the insulin dose-adjusted hemoglobin A1c. J. Pediatr. Endocrinol. Metab. 2021, 34, 1311–1317. [Google Scholar] [CrossRef]
  93. Mørk, F.C.B.; Madsen, J.O.B.; Jensen, A.K.; Hall, G.V.; Pilgaard, K.A.; Pociot, F.; Johannesen, J. Differences in insulin sensitivity in the partial remission phase of childhood type 1 diabetes; a longitudinal cohort study. Diabet. Med. 2022, 39, e14702. [Google Scholar] [CrossRef]
  94. Buchanan, K.; Mehdi, A.M.; Hughes, I.; Cotterill, A.; Le Cao, K.-A.; Thomas, R.; Harris, M. An improved clinical model to predict stimulated C-peptide in children with recent-onset type 1 diabetes. Pediatr. Diabetes 2019, 20, 166–171. [Google Scholar] [CrossRef]
  95. Hocking, M.D.; Rayner, P.W.; Nattrass, M. Residual insulin secretion in adolescent diabetics after remission. Arch. Dis. Child. 1987, 62, 1144–1147. [Google Scholar] [CrossRef] [PubMed]
  96. Pilacinski, S.; Adler, A.I.; Zozulinska-Ziolkiewicz, D.A.; Gawrecki, A.; Wierusz-Wysocka, B. Smoking and other factors associated with short-term partial remission of Type 1 diabetes in adults. Diabet. Med. 2012, 29, 464–469. [Google Scholar] [CrossRef] [PubMed]
  97. Vetter, U.; Heinze, E.; Thon, A.; Beischer, W.; Teller, W. The effect of glucose, tolbutamide, and arginine on C-peptide release during remission in type I diabetes mellitus. Eur. J. Pediatr. 1983, 140, 305–310. [Google Scholar] [CrossRef] [PubMed]
  98. Al Rashed, A.M. Pattern of presentation in type 1 diabetic patients at the diabetes center of a university hospital. Ann. Saudi Med. 2011, 31, 243–249. [Google Scholar] [CrossRef] [PubMed]
  99. Ludvigsson, J.; Heding, L.; Lieden, G.; Marner, B.; Lernmark, A. Plasmapheresis in the initial treatment of insulin-dependent diabetes mellitus in children. Br. Med. J. 1983, 286, 176–178. [Google Scholar] [CrossRef] [PubMed]
  100. Ludvigsson, J.; Lindblom, B. Human lymphocyte antigen DR types in relation to early clinical manifestations in diabetic children. Pediatr. Res. 1984, 18, 1239–1241. [Google Scholar] [CrossRef] [PubMed]
  101. Leighton, E.; Sainsbury, C.A.; Jones, G.C. A Practical Review of C-Peptide Testing in Diabetes. Diabetes Ther. 2017, 8, 475–487. [Google Scholar] [CrossRef]
  102. Pociot, F. Capturing residual beta cell function in type 1 diabetes. Diabetologia 2019, 62, 28–32. [Google Scholar] [CrossRef]
  103. Polonsky, K.S.; Licinio-Paixao, J.; Given, B.D.; Pugh, W.; Rue, P.; Galloway, J.; Karrison, T.; Frank, B. Use of biosynthetic human C-peptide in the measurement of insulin secretion rates in normal volunteers and type I diabetic patients. J. Clin. Invest. 1986, 77, 98–105. [Google Scholar] [CrossRef]
  104. Najjar, S.M.; Perdomo, G. Hepatic Insulin Clearance: Mechanism and Physiology. Physiology 2019, 34, 198–215. [Google Scholar] [CrossRef] [PubMed]
  105. Duckworth, W.C.; Bennett, R.G.; Hamel, F.G. Insulin degradation: Progress and potential. Endocr. Rev. 1998, 19, 608–624. [Google Scholar] [PubMed]
  106. Jones, A.G.; Hattersley, A.T. The clinical utility of C-peptide measurement in the care of patients with diabetes. Diabet. Med. 2013, 30, 803–817. [Google Scholar] [CrossRef] [PubMed]
  107. Clark, P.M. Assays for insulin, proinsulin(s) and C-peptide. Ann. Clin. Biochem. 1999, 36 Pt 5, 541–564. [Google Scholar] [CrossRef]
  108. Besser, R.E.; Ludvigsson, J.; Jones, A.G.; McDonald, T.J.; Shields, B.M.; Knight, B.A.; Hattersley, A.T. Urine C-peptide creatinine ratio is a noninvasive alternative to the mixed-meal tolerance test in children and adults with type 1 diabetes. Diabetes Care 2011, 34, 607–609. [Google Scholar] [CrossRef] [PubMed]
  109. Ambery, P.; Donaldson, J.; Parkin, J.; Austin, D.J. Urinary C-peptide analysis in an intervention study: Experience from the DEFEND-2 otelixizumab trial. Diabet. Med. 2016, 33, 1559–1563. [Google Scholar] [CrossRef] [PubMed]
  110. Besser, R.E.; Shields, B.M.; Casas, R.; Hattersley, A.T.; Ludvigsson, J. Lessons from the mixed-meal tolerance test: Use of 90-minute and fasting C-peptide in pediatric diabetes. Diabetes Care 2013, 36, 195–201. [Google Scholar] [CrossRef] [PubMed]
  111. Greenbaum, C.J.; Mandrup-Poulsen, T.; McGee, P.F.; Battelino, T.; Haastert, B.; Ludvigsson, J.; Pozzilli, P.; Lachin, J.M.; Kolb, H.; Type 1 Diabetes Trial Net Research Group; et al. Mixed-meal tolerance test versus glucagon stimulation test for the assessment of beta-cell function in therapeutic trials in type 1 diabetes. Diabetes Care 2008, 31, 1966–1971. [Google Scholar] [CrossRef] [PubMed]
  112. Ruan, Y.; Willemsen, R.H.; Wilinska, M.E.; Tauschmann, M.; Dunger, D.B.; Hovorka, R. Mixed-meal tolerance test to assess residual beta-cell secretion: Beyond the area-under-curve of plasma C-peptide concentration. Pediatr. Diabetes 2019, 20, 282–285. [Google Scholar] [CrossRef] [PubMed]
  113. Haller, M.J.; Schatz, D.A.; Skyler, J.S.; Krischer, J.P.; Bundy, B.N.; Miller, J.L.; Atkinson, M.A.; Becker, D.J.; Baidal, D.; DiMeglio, L.A.; et al. Low-Dose Anti-Thymocyte Globulin (ATG) Preserves β-Cell Function and Improves HbA(1c) in New-Onset Type 1 Diabetes. Diabetes Care 2018, 41, 1917–1925. [Google Scholar] [CrossRef] [PubMed]
  114. Orban, T.; Bundy, B.; Becker, D.J.; A DiMeglio, L.; E Gitelman, S.; Goland, R.; A Gottlieb, P.; Greenbaum, C.J.; Marks, J.B.; Monzavi, R.; et al. Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: A randomised, double-blind, placebo-controlled trial. Lancet 2011, 378, 412–419. [Google Scholar] [CrossRef] [PubMed]
  115. Boughton, C.K.; Allen, J.M.; Ware, J.; Wilinska, M.E.; Hartnell, S.; Thankamony, A.; Randell, T.; Ghatak, A.; Besser, R.E.; Elleri, D.; et al. Closed-Loop Therapy and Preservation of C-Peptide Secretion in Type 1 Diabetes. N. Engl. J. Med. 2022, 387, 882–893. [Google Scholar] [CrossRef] [PubMed]
  116. Palmer, J.P.; Fleming, G.A.; Greenbaum, C.J.; Herold, K.C.; Jansa, L.D.; Kolb, H.; Lachin, J.M.; Polonsky, K.S.; Pozzilli, P.; Skyler, J.S.; et al. C-peptide is the appropriate outcome measure for type 1 diabetes clinical trials to preserve beta-cell function: Report of an ADA workshop, 21-22 October 2001. Diabetes 2004, 53, 250–264. [Google Scholar] [CrossRef] [PubMed]
  117. Lombardo, F.; Valenzise, M.; Wasniewska, M.G.; Messina, M.F.; Ruggeri, C.; Arrigo, T.; De Luca, F. Two-year prospective evaluation of the factors affecting honeymoon frequency and duration in children with insulin dependent diabetes mellitus: The key-role of age at diagnosis. Diabetes Nutr. Metab. 2002, 15, 246–251. [Google Scholar]
  118. Sokolowska, M.; Chobot, A.; Jarosz-Chobot, P. The honeymoon phase—What we know today about the factors that can modulate the remission period in type 1 diabetes. Pediatr. Endocrinol. Diabetes Metab. 2016, 22, 66–70. [Google Scholar] [CrossRef]
  119. Haller, M.J.; Long, S.A.; Blanchfield, J.L.; Schatz, D.A.; Skyler, J.S.; Krischer, J.P.; Bundy, B.N.; Geyer, S.M.; Warnock, M.V.; Miller, J.L.; et al. Low-Dose Anti-Thymocyte Globulin Preserves C-Peptide, Reduces HbA(1c), and Increases Regulatory to Conventional T-Cell Ratios in New-Onset Type 1 Diabetes: Two-Year Clinical Trial Data. Diabetes 2019, 68, 1267–1276. [Google Scholar] [CrossRef]
  120. Wentworth, J.M.; Bediaga, N.G.; Giles, L.C.; Ehlers, M.; Gitelman, S.E.; Geyer, S.; Evans-Molina, C.; Harrison, L.C.; The Type 1 Diabetes TrialNet Study Group; The Immune Tolerance Network Study Group. Beta cell function in type 1 diabetes determined from clinical and fasting biochemical variables. Diabetologia 2019, 62, 33–40. [Google Scholar] [CrossRef]
  121. Wentworth, J.M.; Bediaga, N.G.; Gitelman, S.E.; Evans-Molina, C.; Gottlieb, P.A.; Colman, P.G.; Haller, M.J.; Harrison, L.C. Clinical trial data validate the C-peptide estimate model in type 1 diabetes. Diabetologia 2020, 63, 885–886. [Google Scholar] [CrossRef]
  122. Watkins, R.A.; Evans-Molina, C.; Terrell, J.K.; Day, K.H.; Guindon, L.; Restrepo, I.A.; Mirmira, R.G.; Blum, J.S.; DiMeglio, L.A. Proinsulin and heat shock protein 90 as biomarkers of beta-cell stress in the early period after onset of type 1 diabetes. Transl. Res. 2016, 168, 96–106.e1. [Google Scholar] [CrossRef]
  123. Freese, J.; Al-Rawi, R.; Choat, H.; Martin, A.; Lunsford, A.; Tse, H.; Mick, G.; McCormick, K. Proinsulin to C-Peptide Ratio in the First Year After Diagnosis of Type 1 Diabetes. J. Clin. Endocrinol. Metab. 2021, 106, e4318–e4326. [Google Scholar] [CrossRef]
  124. Pham, M.N.; Kolb, H.; Mandrup-Poulsen, T.; Battelino, T.; Ludvigsson, J.; Pozzilli, P.; Roden, M.; Schloot, N.C.; European C-Peptide Trial. Serum adipokines as biomarkers of beta-cell function in patients with type 1 diabetes: Positive association with leptin and resistin and negative association with adiponectin. Diabetes Metab. Res. Rev. 2013, 29, 166–170. [Google Scholar] [CrossRef]
  125. Xu, G.; Thielen, L.A.; Chen, J.; Grayson, T.B.; Grimes, T.; Bridges, S.L., Jr.; Tse, H.M.; Smith, B.; Patel, R.; Li, P.; et al. Serum miR-204 is an early biomarker of type 1 diabetes-associated pancreatic beta-cell loss. Am. J. Physiol. Endocrinol. Metab. 2019, 317, E723–E730. [Google Scholar] [CrossRef]
  126. Horber, S.; Orth, M.; Fritsche, A.; Peter, A. Comparability of c-peptide measurements—Current status and clinical relevance. Exp. Clin. Endocrinol. Diabetes 2023, 131, 173–178. [Google Scholar] [CrossRef] [PubMed]
  127. Fourlanos, S.; Narendran, P.; Byrnes, G.B.; Colman, P.G.; Harrison, L.C. Insulin resistance is a risk factor for progression to type 1 diabetes. Diabetologia 2004, 47, 1661–1667. [Google Scholar] [CrossRef] [PubMed]
  128. Nwosu, B.U. Partial Clinical Remission of Type 1 Diabetes: The Need for an Integrated Functional Definition Based on Insulin-Dose Adjusted A1c and Insulin Sensitivity Score. Front. Endocrinol. 2022, 13, 884219. [Google Scholar] [CrossRef] [PubMed]
  129. Nadeau, K.J.; Regensteiner, J.G.; Bauer, T.A.; Brown, M.S.; Dorosz, J.L.; Hull, A.; Zeitler, P.; Draznin, B.; Reusch, J.E.B. Insulin resistance in adolescents with type 1 diabetes and its relationship to cardiovascular function. J. Clin. Endocrinol. Metab. 2010, 95, 513–521. [Google Scholar] [CrossRef] [PubMed]
  130. Dabelea, D.; D’agostino, R.B.; Mason, C.C.; West, N.; Hamman, R.F.; Mayer-Davis, E.J.; Maahs, D.; Klingensmith, G.; Knowler, W.C.; Nadeau, K. Development, validation and use of an insulin sensitivity score in youths with diabetes: The SEARCH for Diabetes in Youth study. Diabetologia 2011, 54, 78–86. [Google Scholar] [CrossRef] [PubMed]
Table 1. Methodologies used in assessing beta cell reserve, and thresholds used to define residual beta cell function in included studies. HbA1c = haemoglobin A1c; IDAA1c = insulin dose-adjusted HbA1c; MMTT = mixed meal tolerance test; TDD = total daily dose. * Denotes the use of chemiluminescence immunoassay. ^ Denotes the use of fluoroimmunoassay.
Table 1. Methodologies used in assessing beta cell reserve, and thresholds used to define residual beta cell function in included studies. HbA1c = haemoglobin A1c; IDAA1c = insulin dose-adjusted HbA1c; MMTT = mixed meal tolerance test; TDD = total daily dose. * Denotes the use of chemiluminescence immunoassay. ^ Denotes the use of fluoroimmunoassay.
MethodThreshold to Define Residual Beta Cell FunctionAuthor
MMTT-Stimulated C-Peptide≥300 pmol/L at 120 minLi X * [15], Zhong T * [16], Chen Y * [17]
>300 pmol/L at 90 minMortensen HB ^ [5]
Max Anderson ML ^ [18], Madsen JOB * [19], Overgaard AJ * [20]
Insulin TDD and HbA1c<0.5 U/kg/d, HbA1c < 8%Kara O [21]
Bowden SA [22]
≤0.5 U/kg/d, HbA1c ≤ 7.5%Verrijn SAA [23]
Ortqvist E [24], Meng X [25]
<0.5 U/kg/d, HbA1c < 7.5%Nordwall M [26]
≤0.5 U/kg/d, HbA1c < 7.5%Araujo DB [27]
<0.5 U/kg/d, HbA1c < 7%Pyziak A [28]
Jamiolkowska-Sztabkowska [29,30,31], Souza L [32], Villalba A [33]
≤0.5 U/kg/d, HbA1c < 7%Wang Y [34]
Pediatric Diabetes Consortium [35]
Dost A [36]
<0.5 U/kg/d, HbA1c ≤ 7%Chobot A [37]
<0.5 U/kg/d, HbA1c ≤ 6%Abdul-Rasoul M [38]
≤0.4 U/kg/d, HbA1c < 7%Humphreys A [39]
≤0.38 U/kg/d, HbA1c < 7.5%Schloot NC [40]
<0.1 U/kg/d, “normal HbA1c” for >3 weeksBarone R [41]
<10 units/day, “normal metabolic state”Kamado K [42]
<0.3 U/kg/d, “normal” HbA1c for at least 10 daysBonfanti R [43]
<0.5 U/kg/d, HbA1c ≤ 7.9%, preprandial blood glucose ≤ 8mMCook JJ [44]
Insulin TDD<0.5 U/kg/dBober E [45], Sanda S [46]
Kordonouri O [47]
Glisic-Milosavljevic S [48]
Muhammad BJ [49], Cadario F [50]
≤0.49 U/kg/dMeng X [25]
<0.3 U/kg/dLundberg RL [51]
Jamiolkowska-Sztabkowska [31]
Nwosu BU [52]
<0.25 U/kg/dFeutren G [53]
≤50% of dose at time of dischargeGlisic-Milosavljevic S [48]
Agner T [54]
Clinical Model-IDAA1C<9Blair JC [55], Polle OG [56], Addala A [57], Kingery SE [58], Villalba A [33]
Pyziak-Skupien A [59]
Cadario F [60]
Casas R [61]
Fonolleda M [2]
≤9Mortensen HB [5], Gomez-Munoz L [62,63], Nwosu BU [52,64,65,66], Yesiltepe-Mutlu [67], Chiavaroli V [68], Pecheur A [69], Nagl K [70], Moya R [71], Narsale A [72], Kaas A [73], Bechi Genzano C [74], Lundgren M [75], Klocperk A [76], Lawes T [77], Pinckney A [78], Cengiz E [79], Neylon OM [80], Moosavi M [81], McGill DE [82], Max Andersen ML [18], Madsen JOB [19], Zhong T [16], Li X [15], Neuman V [83], Quattrin T [84], Cabrera SM [85]
Pyziak A [28], Lundberg RL [51]
Passanisi S [86], Marino KR [87]
Camilo DS [88], Nielens N [89]
Redondo MJ [90]
Franceschi R [91]
Cimbek EA [92], Mork FCB [93]
Clinical Model-Glycemic Target-Adjusted HbA1c≤4.5Nielens N [89]
Clinical Model-Estimated C-Peptide ModelClinical Model to estimate 90 min stimulated c peptideBuchanan K [94]
OtherInsulin TDD < 0.5 U/kg/d and minimal/no glycosuriaHocking MD [95]
Insulin TDD < 0.3 U/kg/d and “proper glycaemic control” and c-peptide > 0.5 ng/mlJamiolkowska-Sztabkowska [31]
Insulin TDD ≤ 0.3 U/kg/d and HbA1c < 7% and a random serum c-peptide > 0.5 ng/mlPilacinski S [96]
Insulin TDD < 0.5 U/kg/d and no glycosuria and “detectable c-peptide”Vetter U [97]
Insulin TDD < 0.5 U/kg/d and absent/minimal glycosuria for >4 weeks and period of no clinical symptomsAl Rashed AM [98]
Insulin TDD < 0.5 U/kg/d and minimal/no glycosuria for ≥1 monthLudvigsson J [99,100]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kennedy, E.C.; Hawkes, C.P. Approaches to Measuring Beta Cell Reserve and Defining Partial Clinical Remission in Paediatric Type 1 Diabetes. Children 2024, 11, 186. https://doi.org/10.3390/children11020186

AMA Style

Kennedy EC, Hawkes CP. Approaches to Measuring Beta Cell Reserve and Defining Partial Clinical Remission in Paediatric Type 1 Diabetes. Children. 2024; 11(2):186. https://doi.org/10.3390/children11020186

Chicago/Turabian Style

Kennedy, Elaine C., and Colin P. Hawkes. 2024. "Approaches to Measuring Beta Cell Reserve and Defining Partial Clinical Remission in Paediatric Type 1 Diabetes" Children 11, no. 2: 186. https://doi.org/10.3390/children11020186

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop