Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Eur J Pharm Sci ; 192: 106644, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37981049

ABSTRACT

INTRODUCTION: SY-009 produces a hypoglycemic effect via inhibiting sodium/glucose cotransporter 1 (SGLT1) in type 2 diabetes mellitus (T2DM) patients. This randomized, double-blind, placebo-controlled, and multiple-dose escalation clinical trial aimed to evaluate the pharmacokinetic and pharmacodynamical characteristics as well as the safety and tolerability of SY-009 in T2DM patients. METHOD: Fifty T2DM patients were randomized into experimental and placebo groups, and hospitalized for 9 days managed with a unified diet and rest management. Subjects were given SY-009 or placebo from day 1 to day 7 at different frequencies and dosages. Single dose cohort was defined as the first dose on day 1 and multiple dose cohort included all the dose from day 1 to 7. Blood samples were collected for pharmacokinetic analysis. Mixed meal tolerance tests were performed. Blood samples were collected to determine glucose, C-peptide, insulin, glucagon-like peptide-1 (GLP-1), and gastric inhibitory polypeptide (GIP). RESULTS: PK parameters were not obtained because blood SY-009 concentrations were below the limit of quantitation in all subjects. SY-009 decreased the postprandial glucose. Blood glucose was controlled within 4 hours after taking the drug. Short-term administration of SY-009 (7 days) had no significant effects on fasting glucose but reduced the secretion of C-peptide, insulin, and GIP and increased GLP-1 secretion. The most common adverse event was gastrointestinal disorder manifesting abdominal pain, diarrhea, and bloating. CONCLUSION: Plasma exposure of SY-009 and its metabolites was fairly low in T2DM patients at doses of 1.0-4.0 mg. SY-009 reduced postprandial glucose, C-peptide, and insulin levels, showing relative safety and tolerability in the dose range of 1.0-4.0 mg. TRIALS REGISTRATION: ClinicalTrials.gov Identifier: NCT04345107.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/metabolism , C-Peptide/therapeutic use , Hypoglycemic Agents , Blood Glucose , Insulin/therapeutic use , Glucagon-Like Peptide 1 , Glucose , Gastric Inhibitory Polypeptide/adverse effects , Gastric Inhibitory Polypeptide/metabolism , Double-Blind Method
2.
Lancet Diabetes Endocrinol ; 11(12): 915-925, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37931637

ABSTRACT

BACKGROUND: Metabolic outcomes in type 1 diabetes remain suboptimal. Disease modifying therapy to prevent ß-cell loss presents an alternative treatment framework but the effect on metabolic outcomes is unclear. We, therefore, aimed to define the relationship between insulin C-peptide as a marker of ß-cell function and metabolic outcomes in new-onset type 1 diabetes. METHODS: 21 trials of disease-modifying interventions within 100 days of type 1 diabetes diagnosis comprising 1315 adults (ie, those 18 years and older) and 1396 children (ie, those younger than 18 years) were combined. Endpoints assessed were stimulated area under the curve C-peptide, HbA1c, insulin use, hypoglycaemic events, and composite scores (such as insulin dose adjusted A1c, total daily insulin, U/kg per day, and BETA-2 score). Positive studies were defined as those meeting their primary endpoint. Differences in outcomes between active and control groups were assessed using the Wilcoxon rank test. FINDINGS: 6 months after treatment, a 24·8% greater C-peptide preservation in positive studies was associated with a 0·55% lower HbA1c (p<0·0001), with differences being detectable as early as 3 months. Cross-sectional analysis, combining positive and negative studies, was consistent with this proportionality: a 55% improvement in C-peptide preservation was associated with 0·64% lower HbA1c (p<0·0001). Higher initial C-peptide levels and greater preservation were associated with greater improvement in HbA1c. For HbA1c, IDAAC, and BETA-2 score, sample size predictions indicated that 2-3 times as many participants per group would be required to show a difference at 6 months as compared with C-peptide. Detecting a reduction in hypoglycaemia was affected by reporting methods. INTERPRETATION: Interventions that preserve ß-cell function are effective at improving metabolic outcomes in new-onset type 1 diabetes, confirming their potential as adjuncts to insulin. We have shown that improvements in HbA1c are directly proportional to the degree of C-peptide preservation, quantifying this relationship, and supporting the use of C-peptides as a surrogate endpoint in clinical trials. FUNDING: JDRF and Diabetes UK.


Subject(s)
Diabetes Mellitus, Type 1 , Adult , Child , Humans , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/complications , C-Peptide/therapeutic use , Cross-Sectional Studies , Glycated Hemoglobin , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use
3.
Nat Commun ; 14(1): 7214, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37940642

ABSTRACT

The use of a standardized outcome metric enhances clinical trial interpretation and cross-trial comparison. If a disease course is predictable, comparing modeled predictions with outcome data affords the precision and confidence needed to accelerate precision medicine. We demonstrate this approach in type 1 diabetes (T1D) trials aiming to preserve endogenous insulin secretion measured by C-peptide. C-peptide is predictable given an individual's age and baseline value; quantitative response (QR) adjusts for these variables and represents the difference between the observed and predicted outcome. Validated across 13 trials, the QR metric reduces each trial's variance and increases statistical power. As smaller studies are especially subject to random sampling variability, using QR as the outcome introduces alternative interpretations of previous clinical trial results. QR can provide model-based estimates that quantify whether individuals or groups did better or worse than expected. QR also provides a purer metric to associate with biomarker measurements. Using data from more than 1300 participants, we demonstrate the value of QR in advancing disease-modifying therapy in T1D. QR applies to any disease where outcome is predictable by pre-specified baseline covariates, rendering it useful for defining responders to therapy, comparing therapeutic efficacy, and understanding causal pathways in disease.


Subject(s)
Diabetes Mellitus, Type 1 , Humans , Diabetes Mellitus, Type 1/drug therapy , C-Peptide/therapeutic use , Clinical Trials as Topic , Insulin Secretion , Precision Medicine
4.
Psychosom Med ; 85(6): 551-560, 2023.
Article in English | MEDLINE | ID: mdl-37234022

ABSTRACT

OBJECTIVE: The objective of this study was to investigate whether placebo effect induced by pharmacological conditioning with intranasal insulin can affect glucose, insulin, C-peptide, hunger, and memory in patients with diabetes type 2 and healthy controls. METHODS: Placebo effect was induced by pharmacological conditioning. Thirty-two older patients (mean age = 68.3 years) with diabetes type 2 and age- and sex-matched thirty-two healthy older adults (mean age = 67.8 years) were randomly assigned to a conditioned or a control group. On day 1, conditioned group received six administrations of intranasal insulin with a conditioned stimulus (CS; smell of rosewood oil), whereas the control group received a placebo with the CS. On day 2, both groups received a placebo spray with the CS. Glucose, insulin, and C-peptide were repeatedly measured in blood. Hunger and memory were assessed with validated measures. RESULTS: Intranasal insulin stabilized dropping glucose levels in patients ( B = 0.03, SE = 0.02, p = .027) and healthy men ( B = 0.046, SE = 0.02, p = .021), and decreased C-peptide levels in healthy controls ( B = 0.01, SE = 0.001, p = .008). Conditioning also prevented the drop of glucose levels but only in men (both healthy and patients; B = 0.001, SE = 0.0003, p = .024). Conditioning significantly decreased hunger in healthy participants ( B = 0.31, SE = 0.09, p < .001). No effects were found on other measures. CONCLUSIONS: Placebo effect induced by conditioning with intranasal insulin modifies blood glucose levels and decreases hunger in older adults, but its effects depend on health status and sex. Insulin conditioning might be beneficial for groups suffering from intensive hunger but seems not be particularly suitable for blood glucose reduction. TRIAL REGISTRATION: Netherlands Trial Register, NL7783 ( https://www.trialregister.nl/trial/7783 ).


Subject(s)
Diabetes Mellitus, Type 2 , Insulin , Male , Humans , Aged , Blood Glucose , Placebo Effect , C-Peptide/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Glucose/pharmacology , Glucose/therapeutic use , Health Status , Double-Blind Method , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use
5.
Drug Discov Ther ; 17(3): 151-156, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37245985

ABSTRACT

Traditional medicines are recently being focused on to treat diabetes and its complications because of their lack of toxic and/or side effects. This report describes the effects of 7-O-galloyl-D-sedoheptulose (GS), a polyphenolic compound isolated from Corni Fructus, on type 2 diabetic db/db mice with hepatic and pancreatic damage. We examined several biochemical factors and oxidative stress- and inflammation-related markers. In the serum, levels of glucose, leptin, insulin, C-peptide, resistin, tumor necrosis factor-α, and interleukin-6 were down-regulated, while adiponectin was augmented by GS treatment. In addition, GS suppressed the reactive oxygen species and lipid peroxidation in the serum, liver, and pancreas, but increased the pancreatic insulin and pancreatic C-peptide contents. These results were derived from attenuating the expression of nicotinamide adenine dinucleotide phosphate oxidase subunit proteins, Nox-4 and p22phox. Augmented nuclear factor (NF)-E2-related factor 2 and heme oxygenase-1 were reduced with a decrease in oxidative stress during GS treatment. NF-κB-related pro-inflammatory factors were also alleviated in hepatic tissue. Moreover, GS modulated the protein expressions of pro-inflammatory NF-κB, cyclooxygenase-2, inducible nitric oxide synthase, c-Jun N-terminal kinase (JNK), phosphor-JNK, activator protein-1, transforming growth factor-ß1, and fibronectin. Based on these results, we demonstrated that the anti-diabetic action of GS may be due to its anti-oxidative stress property and anti-inflammatory action.


Subject(s)
Cornus , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Mice , Animals , Cornus/chemistry , Diabetes Mellitus, Type 2/complications , Polyphenols/pharmacology , Polyphenols/metabolism , Polyphenols/therapeutic use , NF-kappa B/metabolism , NF-kappa B/pharmacology , NF-kappa B/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , C-Peptide/metabolism , C-Peptide/pharmacology , C-Peptide/therapeutic use , Liver , Pancreas/metabolism , Pancreas/pathology , Insulin/pharmacology
6.
Theranostics ; 13(8): 2424-2438, 2023.
Article in English | MEDLINE | ID: mdl-37215567

ABSTRACT

Rationale: Neovascularization is a hallmark of the late stages of diabetic retinopathy (DR) leading to blindness. The current anti-DR drugs have clinical disadvantages including short circulation half-lives and the need for frequent intraocular administration. New therapies with long-lasting drug release and minimal side effects are therefore needed. We explored a novel function and mechanism of a proinsulin C-peptide molecule with ultra-long-lasting delivery characteristics for the prevention of retinal neovascularization in proliferative diabetic retinopathy (PDR). Methods: We developed a strategy for ultra-long intraocular delivery of human C-peptide using an intravitreal depot of K9-C-peptide, a human C-peptide conjugated to a thermosensitive biopolymer, and investigated its inhibitory effect on hyperglycemia-induced retinal neovascularization using human retinal endothelial cells (HRECs) and PDR mice. Results: In HRECs, high glucose conditions induced oxidative stress and microvascular permeability, and K9-C-peptide suppressed those effects similarly to unconjugated human C-peptide. A single intravitreal injection of K9-C-peptide in mice resulted in the slow release of human C-peptide that maintained physiological levels of C-peptide in the intraocular space for at least 56 days without inducing retinal cytotoxicity. In PDR mice, intraocular K9-C-peptide attenuated diabetic retinal neovascularization by normalizing hyperglycemia-induced oxidative stress, vascular leakage, and inflammation and restoring blood-retinal barrier function and the balance between pro- and anti-angiogenic factors. Conclusions: K9-C-peptide provides ultra-long-lasting intraocular delivery of human C-peptide as an anti-angiogenic agent to attenuate retinal neovascularization in PDR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Hyperglycemia , Retinal Neovascularization , Humans , Mice , Animals , Retinal Neovascularization/drug therapy , Diabetic Retinopathy/drug therapy , C-Peptide/pharmacology , C-Peptide/therapeutic use , Endothelial Cells , Neovascularization, Pathologic/drug therapy , Hyperglycemia/drug therapy
7.
Signal Transduct Target Ther ; 8(1): 158, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37076476

ABSTRACT

Disease modifying therapies aiming to preserve ß-cell function in patients with adult-onset autoimmune type 1 diabetes are lacking. Here, we conducted a multi-centre, randomized, controlled trial to assess the ß-cell preservation effects of saxagliptin alone and saxagliptin combined with vitamin D as adjunctive therapies in adult-onset autoimmune type 1 diabetes. In this 3-arm trial, 301 participants were randomly assigned to a 24-month course of the conventional therapy (metformin with or without insulin) or adjunctive saxagliptin or adjunctive saxagliptin plus vitamin D to the conventional therapy. The primary endpoint was the change from baseline to 24 months in the fasting C-peptide. The secondary endpoints included the area under the concentration-time curve (AUC) for C-peptide level in a 2-h mixed-meal tolerance test, glycemic control, total daily insulin use and safety, respectively. The primary endpoint was not achieved in saxagliptin plus vitamin D group (P = 0.18) and saxagliptin group (P = 0.26). However, compared with the conventional therapy, 2-h C-peptide AUC from 24 months to baseline decreased less with saxagliptin plus vitamin D (-276 pmol/L vs. -419 pmol/L; P = 0.01), and not to the same degree with saxagliptin alone (-314 pmol/L; P = 0.14). Notably, for participants with higher glutamic acid decarboxylase antibody (GADA) levels, the decline of ß-cell function was much lower in saxagliptin plus vitamin D group than in the conventional therapy group (P = 0.001). Insulin dose was significantly reduced in both active treatment groups than in the conventional therapy group despite all groups having similar glycemic control. In conclusion, the combination of saxagliptin and vitamin D preserves pancreatic ß-cell function in adult-onset autoimmune type 1 diabetes, an effect especially efficacious in individuals with higher GADA levels. Our results provide evidence for a novel adjunct to insulin and metformin as potential initial treatment for adult-onset type 1 diabetes. (ClinicalTrials.gov identifier: NCT02407899).


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Metformin , Humans , Adult , Hypoglycemic Agents/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Vitamin D/therapeutic use , C-Peptide/therapeutic use , Blood Glucose , Drug Therapy, Combination , Metformin/therapeutic use , Insulin
8.
Lancet Diabetes Endocrinol ; 11(6): 391-401, 2023 06.
Article in English | MEDLINE | ID: mdl-37105208

ABSTRACT

BACKGROUND: Allogeneic islet transplantation is a validated therapy in type 1 diabetes; however, there is decline of transplanted islet graft function over time and the mechanisms underlying this decline are unclear. We evaluated the distinct association between primary graft function (PGF) and 5-year islet transplantation outcomes. METHODS: In this retrospective, multicentre, observational cohort study, we enrolled all patients from the Collaborative Islet Transplant Registry who received islet transplantation alone (ITA recipients) or islet-after-kidney transplantation (IAK recipients) between Jan 19, 1999, and July 17, 2020, with a calculable PGF (exposure of interest), measured 28 days after last islet infusion with a validated composite index of islet graft function (BETA-2 score). The primary outcome was cumulative incidence of unsuccessful islet transplantation, defined as an HbA1c of 7·0% (53 mmol/mol) or higher, or severe hypoglycaemia (ie, requiring third-party intervention to correct), or a fasting C-peptide concentration of less than 0·2 ng/mL. Secondary outcomes were graft exhaustion (fasting C-peptide <0·3 ng/mL); inadequate glucose control (HbA1c ≥7·0% [53 mmol/mol] or severe hypoglycaemia); and requirement for exogenous insulin therapy (≥14 consecutive days). Associations between PGF and islet transplantation outcomes were explored with a competing risk analysis adjusted for all covariates suspected or known to affect outcomes. A predictive model based on PGF was built and internally validated by using bootstraps resampling method. FINDINGS: In 39 centres worldwide, we enrolled 1210 patients with a calculable PGF (of those without missing data, mean age 47 years [SD 10], 712 [59·5%] were female, and 865 (97·9%) were White), who received a median of 10·8 thousand islet-equivalents per kg of bodyweight (IQR 7·4-13·5). 986 (82·4%) were ITA recipients and 211 (17·6%) were IAK recipients. Of 1210 patients, 452 (37·4%) received a single islet infusion and 758 (62·6%) received multiple islet infusions. Mean PGF was 14·3 (SD 8·8). The 5-year cumulative incidence of unsuccessful islet transplantation was 70·7% (95% CI 67·2-73·9), and was inversely and linearly related to PGF, with an adjusted subhazard ratio (sHR) of 0·77 (95% CI 0·72-0·82) per 5-unit increase of BETA-2 score (p<0·0001). Secondary endpoints were similarly related to PGF. The model-adjusted median C-statistic values of PGF for predicting 5-year cumulative incidences of unsuccessful islet transplantation, graft exhaustion, inadequate glucose control, and exogenous insulin therapy were 0·70 (range 0·69-0·71), 0·76 (0·74-0·77), 0·65 (0·64-0·66), and 0·72 (0·71-0·73), respectively. INTERPRETATION: This global multicentre study reports a linear and independent association between PGF and 5-year clinical outcomes of islet transplantation. The main study limitations are its retrospective design and the absence of analysis of complications. FUNDING: Public Health Service Research, National Institutes of Health, Juvenile Diabetes Research Foundation International, Agence National de la Recherche, Fondation de l'Avenir, and Fonds de Dotation Line Renaud-Loulou Gasté.


Subject(s)
Diabetes Mellitus, Type 1 , Hypoglycemia , Islets of Langerhans Transplantation , Humans , Female , Middle Aged , Male , Diabetes Mellitus, Type 1/surgery , Diabetes Mellitus, Type 1/complications , Islets of Langerhans Transplantation/methods , Blood Glucose , Retrospective Studies , C-Peptide/therapeutic use , Glycated Hemoglobin , Treatment Outcome , Transplantation, Homologous , Insulin/therapeutic use , Hypoglycemia/complications , Registries
9.
JAMA ; 329(12): 980-989, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36826834

ABSTRACT

Importance: Near normalization of glucose levels instituted immediately after diagnosis of type 1 diabetes has been postulated to preserve pancreatic beta cell function by reducing glucotoxicity. Previous studies have been hampered by an inability to achieve tight glycemic goals. Objective: To determine the effectiveness of intensive diabetes management to achieve near normalization of glucose levels on preservation of pancreatic beta cell function in youth with newly diagnosed type 1 diabetes. Design, Setting, and Participants: This randomized, double-blind, clinical trial was conducted at 6 centers in the US (randomizations from July 20, 2020, to October 13, 2021; follow-up completed September 15, 2022) and included youths with newly diagnosed type 1 diabetes aged 7 to 17 years. Interventions: Random assignment to intensive diabetes management, which included use of an automated insulin delivery system (n = 61), or standard care, which included use of a continuous glucose monitor (n = 52), as part of a factorial design in which participants weighing 30 kg or more also were assigned to receive either oral verapamil or placebo. Main Outcomes and Measures: The primary outcome was mixed-meal tolerance test-stimulated C-peptide area under the curve (a measure of pancreatic beta cell function) 52 weeks from diagnosis. Results: Among 113 participants (mean [SD] age, 11.8 [2.8] years; 49 females [43%]; mean [SD] time from diagnosis to randomization, 24 [5] days), 108 (96%) completed the trial. The mean C-peptide area under the curve decreased from 0.57 pmol/mL at baseline to 0.45 pmol/mL at 52 weeks in the intensive management group, and from 0.60 to 0.50 pmol/mL in the standard care group (treatment group difference, -0.01 [95% CI, -0.11 to 0.10]; P = .89). The mean time in the target range of 70 to 180 mg/dL, measured with continuous glucose monitoring, at 52 weeks was 78% in the intensive management group vs 64% in the standard care group (adjusted difference, 16% [95% CI, 10% to 22%]). One severe hypoglycemia event and 1 diabetic ketoacidosis event occurred in each group. Conclusions and Relevance: In youths with newly diagnosed type 1 diabetes, intensive diabetes management, which included automated insulin delivery, achieved excellent glucose control but did not affect the decline in pancreatic C-peptide secretion at 52 weeks. Trial Registration: ClinicalTrials.gov Identifier: NCT04233034.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Female , Adolescent , Humans , Child , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/administration & dosage , Blood Glucose/drug effects , Insulin-Secreting Cells/drug effects , C-Peptide/pharmacology , C-Peptide/therapeutic use , Double-Blind Method , Glycemic Control , Blood Glucose Self-Monitoring , Glycated Hemoglobin , Insulin/adverse effects , Insulin/administration & dosage
10.
JAMA ; 329(12): 990-999, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36826844

ABSTRACT

Importance: In preclinical studies, thioredoxin-interacting protein overexpression induces pancreatic beta cell apoptosis and is involved in glucotoxicity-induced beta cell death. Calcium channel blockers reduce these effects and may be beneficial to beta cell preservation in type 1 diabetes. Objective: To determine the effect of verapamil on pancreatic beta cell function in children and adolescents with newly diagnosed type 1 diabetes. Design, Setting, and Participants: This double-blind, randomized clinical trial including children and adolescents aged 7 to 17 years with newly diagnosed type 1 diabetes who weighed 30 kg or greater was conducted at 6 centers in the US (randomized participants between July 20, 2020, and October 13, 2021) and follow-up was completed on September 15, 2022. Interventions: Participants were randomly assigned 1:1 to once-daily oral verapamil (n = 47) or placebo (n = 41) as part of a factorial design in which participants also were assigned to receive either intensive diabetes management or standard diabetes care. Main Outcomes and Measures: The primary outcome was area under the curve values for C-peptide level (a measure of pancreatic beta cell function) stimulated by a mixed-meal tolerance test at 52 weeks from diagnosis of type 1 diabetes. Results: Among 88 participants (mean age, 12.7 [SD, 2.4] years; 36 were female [41%]; and the mean time from diagnosis to randomization was 24 [SD, 4] days), 83 (94%) completed the trial. In the verapamil group, the mean C-peptide area under the curve was 0.66 pmol/mL at baseline and 0.65 pmol/mL at 52 weeks compared with 0.60 pmol/mL at baseline and 0.44 pmol/mL at 52 weeks in the placebo group (adjusted between-group difference, 0.14 pmol/mL [95% CI, 0.01 to 0.27 pmol/mL]; P = .04). This equates to a 30% higher C-peptide level at 52 weeks with verapamil. The percentage of participants with a 52-week peak C-peptide level of 0.2 pmol/mL or greater was 95% (41 of 43 participants) in the verapamil group vs 71% (27 of 38 participants) in the placebo group. At 52 weeks, hemoglobin A1c was 6.6% in the verapamil group vs 6.9% in the placebo group (adjusted between-group difference, -0.3% [95% CI, -1.0% to 0.4%]). Eight participants (17%) in the verapamil group and 8 participants (20%) in the placebo group had a nonserious adverse event considered to be related to treatment. Conclusions and Relevance: In children and adolescents with newly diagnosed type 1 diabetes, verapamil partially preserved stimulated C-peptide secretion at 52 weeks from diagnosis compared with placebo. Further studies are needed to determine the longitudinal durability of C-peptide improvement and the optimal length of therapy. Trial Registration: ClinicalTrials.gov Identifier: NCT04233034.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Adolescent , Humans , Child , Female , Male , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/therapeutic use , C-Peptide/metabolism , C-Peptide/pharmacology , C-Peptide/therapeutic use , Double-Blind Method , Verapamil/adverse effects , Insulin-Secreting Cells/drug effects
11.
J Korean Med Sci ; 38(2): e12, 2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36625174

ABSTRACT

During the coronavirus disease 2019 (COVID-19) pandemic, COVID-19 vaccination-induced hyperglycemia and related complications have been reported. However, there have been few reports of type 1 diabetes triggered by COVID-19 vaccines in subjects without diabetes. Here, we report the case of a 56-year-old female patient who developed hyperglycemia after the second dose of COVID-19 mRNA-based vaccination without a prior history of diabetes. She visited our hospital with uncontrolled hyperglycemia despite administration of oral hyperglycemic agents. Her initial glycated hemoglobin level was high (11.0%), and fasting serum C-peptide level was normal. The fasting serum C-peptide level decreased to 0.269 ng/mL 5 days after admission, and the anti-glutamic acid decarboxylase antibody was positive. The patient was discharged in stable condition with insulin treatment. To our knowledge, this is the first case of the development of type 1 diabetes without diabetic ketoacidosis after mRNA-based COVID-19 vaccination, and is the oldest case of type 1 diabetes development under such circumstances.


Subject(s)
COVID-19 Vaccines , COVID-19 , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Hyperglycemia , Adult , Female , Humans , Middle Aged , C-Peptide/therapeutic use , COVID-19/prevention & control , COVID-19/complications , COVID-19 Vaccines/adverse effects , Diabetes Mellitus, Type 2/complications , Glycated Hemoglobin , Hyperglycemia/complications , Insulin/therapeutic use , Vaccination/adverse effects
12.
Indian J Pharmacol ; 54(4): 244-252, 2022.
Article in English | MEDLINE | ID: mdl-36204807

ABSTRACT

OBJECTIVE: Compliance to insulin injections is poor due to difficulty in subcutaneous administration. Hence, there is a need of an oral formulation of insulin. Oral insulin is currently under investigation. The present analysis aimed to evaluate oral insulin versus placebo for patients with diabetes mellitus (type-1 and type-2). MATERIALS AND METHODS: Results from PUBMED and MEDLINE were searched and compiled from January 1, 2000 to January 9, 2020. Postprandial blood glucose excursions (2PPG), glycated hemoglobin (HbA1c), C-peptide levels, and immune antibody (IAA) levels were compared between the arms. In addition, time to diabetes and safety of oral insulin were discussed. RESULTS: Thirteen out of 1778 trials were included to the analysis. Oral insulin was found to induce significant reduction in mean 2PPG excursion (standardized mean difference [SMD]: -1.94, 95% CI: -3.20 to -0.68, I2 = 91.81, P < 0.005) and mean IAA levels (SMD:-0.49, 95% CI: -0.82 to -0.16, I2 = 27.12, P < 0.005) compared with placebo. Mean C-peptide levels were notably lower in the oral insulin arm. However, the difference was not statistically significant. No significant difference was observed in mean HbA1c levels. The rate of development of type-1 diabetes was not significantly influenced by oral insulin. No deaths or treatment-related serious adverse events were reported. CONCLUSION: Oral insulin provided significant benefits for acute maintenance of diabetes mellitus. It elicited lower immune response and was well tolerated. This new formulation has potential to augment the management of diabetes mellitus. More studies are required to assess its long-term effects.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Blood Glucose/analysis , C-Peptide/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/adverse effects , Insulin/adverse effects
13.
Am J Health Syst Pharm ; 79(23): 2099-2117, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36056809

ABSTRACT

PURPOSE: Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of beta cells, resulting in a loss of insulin production. Patients with T1D carry a substantial disease burden as well as substantial short-term and long-term risks associated with inadequate glycemic control. Currently, treatment mainly consists of insulin, which only treats the symptoms of T1D and not the root cause. Thus, disease-modifying agents such as anti-CD3 monoclonal antibodies (mAbs) that target the autoimmune destruction of beta cells in T1D would provide significant relief and health benefits for patients with T1D. This review summarizes the clinical evidence regarding the safety and efficacy of anti-CD3 mAbs in the prevention and treatment of T1D. SUMMARY: A total of 27 studies reporting or evaluating data from clinical trials involving otelixizumab and teplizumab were included in the review. Anti-CD3 mAbs have shown significant benefits in both patients at high risk for T1D and those with recent-onset T1D. In high-risk populations, anti-CD3 mAbs delayed time to diagnosis, preserved C-peptide levels, and improved metabolic parameters. In recent-onset T1D, anti-CD3 mAbs preserved C-peptide levels and reduced insulin needs for extended periods. Anti-CD3 mAb therapy appears to be safe, with primarily transient and self-limiting adverse effects and no negative long-term effects. CONCLUSION: Anti-CD3 mAbs are promising disease-modifying treatments for T1D. Their role in T1D may introduce short-term and long-term benefits with the potential to mitigate the significant disease burden; however, more evidence is required for an accurate assessment.


Subject(s)
Diabetes Mellitus, Type 1 , Humans , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/prevention & control , C-Peptide/metabolism , C-Peptide/therapeutic use , CD3 Complex/therapeutic use , Antibodies, Monoclonal/therapeutic use , Insulin/therapeutic use
14.
Curr Med Res Opin ; 38(11): 1807-1814, 2022 11.
Article in English | MEDLINE | ID: mdl-35943899

ABSTRACT

OBJECTIVE: To identify population with type 2 diabetes mellitus (T2DM) who are more likely to benefit from switching to basal insulin (BI) treatment from premixed insulin. METHODS: This secondary analysis included data from a previously published study (Optimization: NCT00693771) which was a single-arm, multicenter, 16 weeks, phase IV study. The analysis included participants with T2DM inadequately controlled with premixed insulin plus oral hypoglycemic drugs (OADs) who switched to BI plus OADs. RESULTS: Among the 297 participants included for analysis, subjects with fasting C-peptide (FCP)>1.2 nmol/L group showed a trend for greater reduction in HbA1c [Least square mean difference (LSMD), -0.59; 95% confidence interval (CI), -0.98 to -0.21; p = 0.003] and FPG (LSMD, -1.36; 95% CI, -2.20 to -0.53; p = 0.002) than those with FCP ≤ 0.4 nmol/L. The baseline insulin glargine 100 U/mL (Gla-100) dose increased significantly in 0.4 to ≤ 1.2 nmol/L group with LS mean difference (SE) of 0.16 (0.01) U/kg/day (p = 0.008) compared to FCP ≤ 0.4 nmol/L group. When combined with Diabetes Treatment Satisfaction Questionnaire (DTSQ) score, participants with a C-peptide level of 0.4 to ≤1.2 nmol/L (OR, 4.05; 95% CI, 1.08 to 15.22; p = 0.039) had significantly higher odds of achieving HbA1c <7%. The number of participants experiencing documented symptomatic hypoglycaemia (≤3.9 mmol/L) was higher in the FCP ≤0.4 nmol/L group compared to those in 0.4 to ≤1.2 nmol/L FCP group at any time of the day (31.6 vs. 17.1%) and during night (00:00 ∼ 05:59) (17.1 vs. 7.5%). CONCLUSION: The findings from this study proposed that FCP is an important biomarker to identify T2DM participants who experience improved glucose control without compromising on hypoglycemia levels during switch from premixed insulin to BI. Participants especially with FCP levels >1.2 nmol/L may respond better in terms of HbA1c reduction without increased hypoglycemia risk compared to those with lower FCP values.


Subject(s)
Diabetes Mellitus, Type 2 , Hypoglycemia , Humans , Diabetes Mellitus, Type 2/drug therapy , Glycated Hemoglobin/analysis , Insulin, Long-Acting , Hypoglycemic Agents/adverse effects , C-Peptide/therapeutic use , Administration, Oral , Hypoglycemia/epidemiology , Insulin Glargine , Insulin/adverse effects , Blood Glucose/analysis
15.
Rev Med Suisse ; 18(792): 1556-1559, 2022 Aug 24.
Article in French | MEDLINE | ID: mdl-36004656

ABSTRACT

Plasma C-peptide represents a direct measure of endogenous insulin secretion. The development of new assays for measuring C-peptide have made it possible to demonstrate that a low insulin secretion persists in 30 to 80% of subjects with type 1 diabetes (T1D), even among those with long-standing disease. Several studies have established that the persistence of B cell function of the islets of Langerhans is associated with a protection against the development of microvascular complications and resulted in a significant reduction in the prevalence of severe hypoglycaemia in people with T1D. Further studies are needed to clarify the underlying pathophysiological mechanisms and the therapeutic strategies that would maintain B-cell function and thus improve the quality of life of patients with T1D.


Le peptide-C plasmatique constitue une mesure directe de la sécrétion endogène d'insuline. Le développement de nouveaux dosages du peptide-C a permis de démontrer qu'il persiste chez 30 à 80 % des sujets diabétiques de type 1 (DT1) une faible sécrétion d'insuline, même sur le long terme. Plusieurs études ont établi que la persistance de la fonction B des îlots de Langerhans était associée à une protection contre le développement des complications microvasculaires et engendrait une réduction significative de la prévalence des hypoglycémies sévères chez les personnes DT1. Mais des études complémentaires sont encore nécessaires afin de préciser les mécanismes physiopathologiques sous-jacents et les stratégies thérapeutiques qui permettraient de maintenir la fonction de la cellule B et d'améliorer ainsi la qualité de vie des sujets avec un DT1.


Subject(s)
Diabetes Mellitus, Type 1 , C-Peptide/metabolism , C-Peptide/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Disease Progression , Humans , Insulin/therapeutic use , Insulin Secretion , Quality of Life
16.
Comput Methods Programs Biomed ; 223: 106973, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35792365

ABSTRACT

BACKGROUND AND OBJECTIVE: The increasing incidence of diabetes continuously stimulates the research on new antidiabetic drugs. Computer simulation can save time and costs, alleviating the need of animal trials and providing useful information for optimal experiment design and drug dosing. We recently presented a type 2 diabetes (T2D) simulator as tool for in silico testing of new molecules and guiding treatment optimization. Here we present a user-friendly interface aimed to increase the usability of the simulator. METHOD: The simulator, based on a large-scale glucose, insulin, and C-peptide model and equipped with 100 virtual subjects well describing system dynamics in a real T2D population, is extended to incorporate pharmacokinetics/pharmacodynamics (PK/PD) of a drug of interest. A graphical interface is developed on top of the simulator, allowing an easy design of in silico experiments: specifically, it is possible to select the population size to test, design the experiment (crossover or parallel), its duration and the sampling grid, choose glucose and insulin doses, and define treatment PK/PD and dose administered. The simulator also provides the outcome metrics requested by the user, and performs statistical comparisons among treatments and/or placebo. RESULTS: To illustrate the potential of the simulator, we provided a case study using metformin and liraglutide. Literature-based PK/PD models of metformin and liraglutide have been incorporated in the simulator, by modulating key drug-sensitive model parameters. An in silico placebo-controlled trial has been done by simulating a three-arm meal tolerance test with subjects receiving placebo, metformin 850 mg, liraglutide 1.80 mg, respectively. The obtained results are in agreement with the clinical evidences, in terms of main glucose, insulin, and C-peptide outcome metrics. CONCLUSIONS: We developed a user-friendly software interface for the T2D simulator to support the design and test of new antidiabetic drugs and treatments. This increases the simulator usability, making it suitable also for users who have low experience with computer programming.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Blood Glucose , C-Peptide/therapeutic use , Computer Simulation , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Liraglutide/pharmacology , Liraglutide/therapeutic use , Metformin/therapeutic use , Software
17.
Rev Med Chil ; 150(1): 115-119, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35856973

ABSTRACT

Insulin antibodies (IAs) induced by exogenous insulin rarely cause hypoglycemia. However, insulin autoantibodies (IAAs) in insulin autoimmune syndrome (IAS) can cause hypoglycemia. The typical manifestations of IAS are fasting or postprandial hypoglycemia, elevated insulin level, decreased C-peptide levels, and positive IAA. We report a 45-year-old male with type 1 diabetes mellitus (T1DM) treated with insulin analogues suffering from recurrent hypoglycemic coma and diabetic ketoacidosis (DKA). His symptoms were caused by exogenous insulin and were similar to IAS. A possible reason was that exogenous insulin induced IA. IA titers were 61.95% (normal: < 5%), and the concentrations of insulin and C-peptide were > 300 mU/L and < 0.02 nmol/L when hypoglycemia occurred. Based on his clinical symptoms and other examinations, he was diagnosed with hyperinsulinemic hypoglycemia caused by IA. His symptoms improved after changing insulin regimens from insulin lispro plus insulin detemir to recombinant human insulin (Gensulin R) and starting prednisone.


Subject(s)
Autoimmune Diseases , Diabetes Mellitus, Type 1 , Diabetic Ketoacidosis , Hypoglycemia , Autoimmune Diseases/diagnosis , C-Peptide/therapeutic use , Coma , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Diabetic Ketoacidosis/chemically induced , Diabetic Ketoacidosis/complications , Diabetic Ketoacidosis/drug therapy , Humans , Hypoglycemia/chemically induced , Hypoglycemic Agents/adverse effects , Insulin/therapeutic use , Insulin Antibodies/therapeutic use , Male , Middle Aged
18.
Syst Rev ; 11(1): 82, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35501872

ABSTRACT

BACKGROUND: Stem cell transplantation (SCT) has paved the way for treatment of autoimmune diseases. SCT has been investigated in type 1 diabetes mellitus (T1DM) as an autoimmune-based disorder, but previous studies have not presented a comprehensive view of its effect on treatment of T1DM. METHODOLOGY: After registration of the present systematic review and meta-analysis in the PROSPERO, a search was done according to the Cochrane guidelines for evaluation of clinical trials to find eligible clinical trials that investigated the effect of SCT on T1DM (based on ADA® diagnostic criteria) from PubMed, Web of science, Scopus, etc, as well as registries of clinical trials from January 1, 2000, to September 31, 2019. A search strategy was designed using MeSH and EM-tree terms. Primary outcome included the changes in the insulin total daily dose (TDD) (U/kg) level, and secondary outcomes included the changes in the HbA1c, c-peptide, and adjusted HbA1c levels. The Q Cochrane test and I2 statistic were performed to assess the heterogeneity and its severity in primary clinical trials. The Cochrane ROB was used to determine risk of bias, and Cochrane Handbook for Systematic Reviews of Interventions was used in the full text papers. The meta-analysis was accomplished in the STATA software, and the results were shown on their forest plots. Confounders were evaluated by the meta-regression test. RESULTS: A total of 9452 studies were electronically screened, and 35 papers were included for data extraction. The results of this review study showed that 173 (26.5%) diabetic patients experienced insulin-free period (from 1 to 80 months), and 445 (68%) showed reduction in TDD of insulin after the SCT. Combination of hematopoietic stem cell (HSC) with mesenchymal stem cell (MSC) transplantation were significantly associated with improvement of the TDD (SMD: - 0.586, 95% CI: - 1.204/- 0.509, I2: 0%), HbA1c (SMD: - 0.736, 95% CI: - 1.107/- 0.365, I2: 0%), adjusted HbA1c (SMD: - 2.041, 95% CI: - 2.648/- 1.434, I2: 38.4%), and c-peptide (SMD: 1.917, 95% CI: 0.192/3.641, I2: 92.5%) on month 3 of follow-up, while its association had a growing trend from 3 to 12 months after the transplantation. Considering severe adverse events, HSC transplantation accompanied with conditioning could not be suggested as a safe treatment. CONCLUSION: Most of the clinical trials of SCT in T1DM were single arm. Although meta-analysis illustrated the SCT is associated with T1DM improvement, well-designed randomized clinical trials are needed to clarify its efficacy. RECOMMENDATION: Based on the results of this meta-analysis, the MSC and its combination with HSC could be considered as "Safe Cell" for SCT in T1DM. Furthermore, to evaluate the SCT efficacy, calculation of insulin TDD (U/kg/day), AUC of c-peptide, and adjusted HbA1c are highly recommended.


Subject(s)
Diabetes Mellitus, Type 1 , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , C-Peptide/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Glycated Hemoglobin/metabolism , Humans , Insulin/therapeutic use
19.
Rev. méd. Chile ; 150(1): 115-119, ene. 2022. tab
Article in English | LILACS | ID: biblio-1389609

ABSTRACT

ABSTRACT Insulin antibodies (IAs) induced by exogenous insulin rarely cause hypoglycemia. However, insulin autoantibodies (IAAs) in insulin autoimmune syndrome (IAS) can cause hypoglycemia. The typical manifestations of IAS are fasting or postprandial hypoglycemia, elevated insulin level, decreased C-peptide levels, and positive IAA. We report a 45-year-old male with type 1 diabetes mellitus (T1DM) treated with insulin analogues suffering from recurrent hypoglycemic coma and diabetic ketoacidosis (DKA). His symptoms were caused by exogenous insulin and were similar to IAS. A possible reason was that exogenous insulin induced IA. IA titers were 61.95% (normal: 300 mU/L and < 0.02 nmol/L when hypoglycemia occurred. Based on his clinical symptoms and other examinations, he was diagnosed with hyperinsulinemic hypoglycemia caused by IA. His symptoms improved after changing insulin regimens from insulin lispro plus insulin detemir to recombinant human insulin (Gensulin R) and starting prednisone.


Los anticuerpos contra la insulina (AI) inducidos por la insulina exógena raramente causan hipoglucemia. No obstante, los autoanticuerpos contra la insulina (AIA) en el síndrome autoinmune de insulina (SAI) pueden causar hipoglucemia. Las manifestaciones típicas del SAI son la hipoglucemia en ayunas o posprandial, niveles elevados de insulina, la disminución del nivel de péptido C y AIA positivos. Presentamos un paciente hombre de 45 años con diabetes mellitus de tipo 1 (DMT1) tratado con análogos de insulina, que sufría comas hipoglucémicos recurrentes y cetoacidosis diabética (CAD). Sus síntomas fueron causados por la insulina exógena y fueron similares al SAI. La posible razón fue que la insulina exógena indujo AI. El título de AI era del 61,95% (Normal: 300 mU/L y < 0,02 nmol/L cuando se producía la hipoglucemia. Basados en sus síntomas clínicos y otros exámenes, se le diagnosticó hipoglucemia hiperinsulinémica causada por la AI. Sus síntomas mejoraron después de cambiar el régimen de insulina de lispro más insulina detemir a insulina humana recombinante (Gensulin R) y de empezar a tomar prednisona.


Subject(s)
Humans , Male , Middle Aged , Autoimmune Diseases/diagnosis , Diabetic Ketoacidosis/complications , Diabetic Ketoacidosis/chemically induced , Diabetic Ketoacidosis/drug therapy , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemia/chemically induced , C-Peptide/therapeutic use , Coma , Hypoglycemic Agents/adverse effects , Insulin/therapeutic use , Insulin Antibodies/therapeutic use
20.
Calcif Tissue Int ; 107(3): 266-280, 2020 09.
Article in English | MEDLINE | ID: mdl-32607636

ABSTRACT

C-peptide therapy protects against diabetic micro- and macrovascular damages and neuropatic complications. However, to date, the role of C-peptide in preventing diabetes-related bone loss has not been investigated. Our aim was to evaluate if C-peptide infusion improves bone quality in diabetic rats. Twenty-three male Wistar rats were randomly divided into three groups: normal control group; sham diabetic control group; diabetic plus C-peptide group. Diabetes was induced by streptozotocin injection and C-peptide was delivered subcutaneously for 6 weeks. We performed micro-CT and histological testing to assess several trabecular microarchitectural parameters. At the end, diabetic plus C-peptide rats had a higher serum C-peptide (p = 0.02) and calcium (p = 0.04) levels and tibia weight (p = 0.02) than the diabetic control group. The diabetic plus C-peptide group showed a higher trabecular thickness and cross-sectional thickness than the diabetic control group (p = 0.01 and p = 0.03). Both the normal control and diabetic plus C-peptide groups had more Runx-2 and PLIN1 positive cells in comparison with the diabetic control group (p = 0.045 and p = 0.034). Diabetic rats receiving C-peptide had higher quality of trabecular bone than diabetic rats not receiving this treatment. If confirmed, C-peptide could have a role in improving bone quality in diabetes.


Subject(s)
Bone Density , C-Peptide/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Animals , Diabetes Mellitus, Experimental/chemically induced , Male , Rats , Rats, Wistar , Streptozocin
SELECTION OF CITATIONS
SEARCH DETAIL
...