1
|
George M, Zilbermint M, Sokolinsky S, Batty K, Motevalli M, Stanback C, Gonzales E, Miller C, Sequeira L, Demidowich AP. Effects of preprandial versus postprandial nutritional insulin administration in the inpatient setting. Diabetes Res Clin Pract 2024; 214:111785. [PMID: 39019331 DOI: 10.1016/j.diabres.2024.111785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/07/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
AIMS Hospitalized patients can have inconsistent nutritional intake due to acute illness, changing diet, or unpredictable meal delivery. The aim of this study was to evaluate whether implementation of a hospital-wide policy shifting nutritional insulin administration from pre-meal to post-meal was associated with changes in glycemic control or length of stay (LOS). METHODS This retrospective study performed at a community hospital evaluated adult inpatients receiving nutritional insulin across three time periods. pre-intervention, immediate post-intervention, and distant post-intervention. Outcomes included rates of hypoglycemia (glucose ≤ 70 mg/dL), moderate hypoglycemia (< 54 mg/dL), severe hypoglycemia (≤ 40 mg/dL), severe hyperglycemia (≥ 300 mg/dL), daily mean glucose level, and LOS. RESULTS The number of patient-days analyzed across the cohorts were 1948, 1751, and 3244, respectively. After multivariate adjustment, risk of developing any hypoglycemia and severe hypoglycemia significantly decreased over time (p = 0.001 and p = 0.009, respectively). Daily mean glucose increased over time (194.6 ± 62.5 vs 196.8 ± 65.5 vs 199.3 ± 61.5 mg/dL; p = 0.003), but there were no significant differences among rates of severe hyperglycemia (p = 0.10) or LOS (p = 0.74). CONCLUSIONS Implementing a hospital-wide shift to postprandial nutritional insulin administration significantly reduced hypoglycemia rates without increasing severe hyperglycemia. This suggests a promising strategy for improving patient safety, but further prospective randomized controlled trials are warranted to confirm these findings.
Collapse
Affiliation(s)
- Merit George
- Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, 1830 East Monument Street, Suite 333, Baltimore, MD 21287, USA
| | - Mihail Zilbermint
- Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, 1830 East Monument Street, Suite 333, Baltimore, MD 21287, USA; Division of Hospital Medicine, Johns Hopkins Community Physicians, Johns Hopkins Medicine, 6225 Smith Ave., Suite B-300, Baltimore, MD 21209, USA; Suburban Hospital, Johns Hopkins Medicine, 8600 Old Georgetown Road, Bethesda, MD 20814, USA
| | - Sam Sokolinsky
- JHHS Quality and Clinical Analytics, Johns Hopkins Hospital, Johns Hopkins Medicine, Baltimore, MD 21209, USA
| | - Kristine Batty
- Division of Hospital Medicine, Johns Hopkins Community Physicians, Johns Hopkins Medicine, 6225 Smith Ave., Suite B-300, Baltimore, MD 21209, USA; Suburban Hospital, Johns Hopkins Medicine, 8600 Old Georgetown Road, Bethesda, MD 20814, USA
| | - Mahsa Motevalli
- Division of Hospital Medicine, Johns Hopkins Community Physicians, Johns Hopkins Medicine, 6225 Smith Ave., Suite B-300, Baltimore, MD 21209, USA; Suburban Hospital, Johns Hopkins Medicine, 8600 Old Georgetown Road, Bethesda, MD 20814, USA
| | - Camille Stanback
- Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, 1830 East Monument Street, Suite 333, Baltimore, MD 21287, USA; Division of Hospital Medicine, Johns Hopkins Community Physicians, Johns Hopkins Medicine, 6225 Smith Ave., Suite B-300, Baltimore, MD 21209, USA; Sibley Memorial Hospital, Johns Hopkins Medicine, 5255 Loughboro Rd NW, Washington, DC 20016, USA
| | - Eva Gonzales
- Johns Hopkins Howard County Medical Center, Johns Hopkins Medicine, 5755 Cedar Ln, Columbia, MD 20794, USA
| | - Catherine Miller
- Johns Hopkins Howard County Medical Center, Johns Hopkins Medicine, 5755 Cedar Ln, Columbia, MD 20794, USA
| | - Lynette Sequeira
- Department of Internal Medicine, Johns Hopkins University School of Medicine, 2024 E Monument St, Baltimore, MD 21287
| | - Andrew P Demidowich
- Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, 1830 East Monument Street, Suite 333, Baltimore, MD 21287, USA; Division of Hospital Medicine, Johns Hopkins Community Physicians, Johns Hopkins Medicine, 6225 Smith Ave., Suite B-300, Baltimore, MD 21209, USA; Johns Hopkins Howard County Medical Center, Johns Hopkins Medicine, 5755 Cedar Ln, Columbia, MD 20794, USA.
| |
Collapse
|
2
|
Villasenor M, Selzer AR. Preoperative Patient Evaluation: Newer Hypoglycemic Agents. Anesthesiol Clin 2024; 42:41-52. [PMID: 38278591 DOI: 10.1016/j.anclin.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
New medications in the treatment of diabetes are an active area of research and drug development. Although many hypoglycemic therapies have been in use for decades, new evidence continues to emerge highlighting benefits of these medications for other indications. In this article, the authors review the classes of newer hypoglycemic agents and summarize medications currently in phase 2 and 3 clinical trials. The literature to support specific recommendations for perioperative management is scant, however, where it exists, we have included it. In other instances, the authors have noted a reasonable approach based on pharmacokinetics and principles of perioperative medication management.
Collapse
Affiliation(s)
- Mario Villasenor
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Angela Roberts Selzer
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
3
|
Gorai B, Vashisth H. Structural models of viral insulin-like peptides and their analogs. Proteins 2023; 91:62-73. [PMID: 35962629 PMCID: PMC9772067 DOI: 10.1002/prot.26410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/21/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022]
Abstract
The insulin receptor (IR), the insulin-like growth factor-1 receptor (IGF1R), and the insulin/IGF1 hybrid receptors (hybR) are homologous transmembrane receptors. The peptide ligands, insulin and IGF1, exhibit significant structural homology and can bind to each receptor via site-1 and site-2 residues with distinct affinities. The variants of the Iridoviridae virus family show capability in expressing single-chain insulin/IGF1 like proteins, termed viral insulin-like peptides (VILPs), which can stimulate receptors from the insulin family. The sequences of VILPs lacking the central C-domain (dcVILPs) are known, but their structures in unbound and receptor-bound states have not been resolved to date. We report all-atom structural models of three dcVILPs (dcGIV, dcSGIV, and dcLCDV1) and their complexes with the receptors (μIR, μIGF1R, and μhybR), and probed the peptide/receptor interactions in each system using all-atom molecular dynamics (MD) simulations. Based on the nonbonded interaction energies computed between each residue of peptides (insulin and dcVILPs) and the receptors, we provide details on residues establishing significant interactions. The observed site-1 insulin/μIR interactions are consistent with previous experimental studies, and a residue-level comparison of interactions of peptides (insulin and dcVILPs) with the receptors revealed that, due to sequence differences, dcVILPs also establish some interactions distinct from those between insulin and IR. We also designed insulin analogs and report enhanced interactions between some analogs and the receptors.
Collapse
Affiliation(s)
- Biswajit Gorai
- Department of Chemical Engineering, University of New Hampshire, Durham, NH 03824, USA
| | - Harish Vashisth
- Department of Chemical Engineering, University of New Hampshire, Durham, NH 03824, USA
| |
Collapse
|
4
|
DeMarsilis A, Reddy N, Boutari C, Filippaios A, Sternthal E, Katsiki N, Mantzoros C. Pharmacotherapy of type 2 diabetes: An update and future directions. Metabolism 2022; 137:155332. [PMID: 36240884 DOI: 10.1016/j.metabol.2022.155332] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Type 2 diabetes (T2D) is a widely prevalent disease with substantial economic and social impact for which multiple conventional and novel pharmacotherapies are currently available; however, the landscape of T2D treatment is constantly changing as new therapies emerge and the understanding of currently available agents deepens. This review aims to provide an updated summary of the pharmacotherapeutic approach to T2D. Each class of agents is presented by mechanism of action, details of administration, side effect profile, cost, and use in certain populations including heart failure, non-alcoholic fatty liver disease, obesity, chronic kidney disease, and older individuals. We also review targets of novel therapeutic T2D agent development. Finally, we outline an up-to-date treatment approach that starts with identification of an individualized goal for glycemic control then selection, initiation, and further intensification of a personalized therapeutic plan for T2D.
Collapse
Affiliation(s)
- Antea DeMarsilis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Niyoti Reddy
- Department of Medicine, School of Medicine, Boston University, Boston, USA
| | - Chrysoula Boutari
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Andreas Filippaios
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Elliot Sternthal
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02115, USA
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Sindos, Greece; School of Medicine, European University Cyprus, Nicosia, Cyprus.
| | - Christos Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
De Block CEM, Van Cauwenberghe J, Bochanen N, Dirinck E. Rapid-acting insulin analogues: Theory and best clinical practice in type 1 and type 2 diabetes. Diabetes Obes Metab 2022; 24 Suppl 1:63-74. [PMID: 35403348 DOI: 10.1111/dom.14713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 12/01/2022]
Abstract
Since the discovery of insulin 100 years ago, insulin preparations have improved significantly. Starting from purified animal insulins, evolving to human insulins produced by genetically modified organisms, and ultimately to insulin analogues, all in an attempt to mimic physiological insulin action profiles seen in individuals without diabetes. Achieving strict glucose control without hypoglycaemia and preventing chronic complications of diabetes while preserving quality of life remains a challenging goal, but the advent of newer ultra-rapid-acting insulin analogues may enable intensive insulin therapy without being too disruptive to daily life. Ultra-rapid-acting insulin analogues can be administered shortly before meals and give better coverage of mealtime-induced glucose excursions than conventional insulin preparations. They also increase convenience with timing of bolus dosing. In this review, we focus on the progress that has been made in rapid-acting insulins. We summarize pharmacokinetic and pharmacodynamic data, clinical trial data supporting the use of these new formulations as part of a basal-bolus regimen and continuous subcutaneous insulin infusion, and provide a clinical perspective to help guide healthcare professionals when and for whom to use ultra-fast-acting insulins.
Collapse
Affiliation(s)
- Christophe E M De Block
- Department of Endocrinology, Diabetology & Metabolism, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- University of Antwerp, Faculty of Medicine & Health Sciences, Laboratory of Experimental Medicine and Paediatrics (LEMP), Wilrijk, Belgium
| | - Jolijn Van Cauwenberghe
- Department of Endocrinology, Diabetology & Metabolism, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- University of Antwerp, Faculty of Medicine & Health Sciences, Laboratory of Experimental Medicine and Paediatrics (LEMP), Wilrijk, Belgium
| | - Niels Bochanen
- Department of Endocrinology, Diabetology & Metabolism, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- University of Antwerp, Faculty of Medicine & Health Sciences, Laboratory of Experimental Medicine and Paediatrics (LEMP), Wilrijk, Belgium
| | - Eveline Dirinck
- Department of Endocrinology, Diabetology & Metabolism, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- University of Antwerp, Faculty of Medicine & Health Sciences, Laboratory of Experimental Medicine and Paediatrics (LEMP), Wilrijk, Belgium
| |
Collapse
|
6
|
Gan L, Huang S, Hu Y, Zhang J, Wang X. Heat treatment reduced the expression of miR-7-5p to facilitate insulin-stimulated lactate secretion by targeting IRS2 in boar Sertoli cells. Theriogenology 2021; 180:161-170. [PMID: 34973648 DOI: 10.1016/j.theriogenology.2021.12.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 12/06/2021] [Accepted: 12/26/2021] [Indexed: 12/26/2022]
Abstract
Insulin dysfunction of diabetes mellitus (DM) disorders the glucose metabolism in Sertoli cells (SCs), resulting in the impairment of spermatogenesis.Insulin signaling system in Sertoli cells (SCs) plays an important role in regulating lactate secretion. Heat treatment could increase the lactate secretion of boar SCs, but whether heat treatment participates in lactate secretion by improving the sensitivity of insulin is unknown. In the current study, the primary SCs from 21-day-old boar were employed to treat with 100 nM insulin for 24 h or heat treatment (43 °C, 30 min). Heat treatment strengthened the effect of insulin on the effect of lactate secretion. In addition, heat treatment increased the expression of insulin-induced insulin receptor substrate 2 (IRS2), but reduced the expression of miR-7-5p. Using dual luciferase reporter assay and Western blot, the study found that IRS2 is a potential target gene of miR-7-5p. Heat treatment also enhanced the Phosphorylation of insulin-stimulated PI3K/Akt, and increased lactate secretion by promoting the expression of Glucose Transporter 3 (GLUT3), Lactate Dehydrogenase-A (LDHA) and monocarboxylate transporter 1 (MCT1). Furthermore, miR-7-5p inhibitor could partly mimic the effects of heat treatment on lactate production of SCs, indicating that heat treatment improves insulin sensitivity by regulating the expression of miR-7-5p/IRS2/PI3K/Akt. These results reveal a novel miRNA-mediated mechanism of heat treatment on the regulation of lactate metabolism production, and suggest that targeting miR-7-5p is a probably therapeutic method to insulin dysfunction-induced metabolic diseases.
Collapse
Affiliation(s)
- Lu Gan
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicnie, Southwest University, Beibei, Chongqing, 400715, PR China
| | - Sha Huang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicnie, Southwest University, Beibei, Chongqing, 400715, PR China
| | - Yu Hu
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicnie, Southwest University, Beibei, Chongqing, 400715, PR China
| | - JiaoJiao Zhang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicnie, Southwest University, Beibei, Chongqing, 400715, PR China
| | - XianZhong Wang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicnie, Southwest University, Beibei, Chongqing, 400715, PR China.
| |
Collapse
|
7
|
Lane W, Faurby M, Husemoen LLN, Markovich DL, Rathor N, De Block C. Glycaemic Control in People with Diabetes Starting Treatment with Fast-Acting Insulin Aspart: a US Database Study. Diabetes Ther 2021; 12:3067-3077. [PMID: 34637112 PMCID: PMC8586136 DOI: 10.1007/s13300-021-01165-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/27/2021] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION This study investigated glycaemic control in individuals with type 1 (T1D) or type 2 diabetes (T2D) 6 months after initiating fast-acting insulin aspart (faster aspart) in a real-world setting. METHODS This was a single-arm, observational study using extracted patient data from the IBM® Explorys® database (USA) for individuals with T1D or T2D initiating faster aspart (at least one prescription of faster aspart) in the study period 1 January 2018 to 27 October 2020. Clinical characteristics, including age, body mass index, and baseline HbA1c, were extracted, as well as recorded events of hypoglycaemia. The primary endpoint was the change in HbA1c from baseline to 6 months. RESULTS A total of 787 individuals were included; 36.6% of these individuals had T1D and 63.4% had T2D (of whom 46.9% were new users of rapid-acting insulin when initiating faster aspart [T2D new users] and 53.1% were switching from another rapid-acting insulin to faster aspart [T2D switchers]). For individuals with T1D, T2D new users, or T2D switchers, estimated mean change in HbA1c from baseline to 6 months was - 0.20% (95% CI - 0.53, 0.14; p = 0.252), - 1.00% (95% CI - 1.34, - 0.67; p < 0.0001), and - 0.70% (95% CI - 1.06, - 0.35; p = 0.0001), respectively. In the baseline HbA1c > 8.5% subgroup, there was a significant estimated decrease in HbA1c from baseline to 6 months in individuals with T1D (- 1.2% [95% CI - 1.80, - 0.60]; p = 0.0001) or T2D (- 0.6% [95% CI - 0.92, - 0.35]; p < 0.0001). Event rates of hypoglycaemia after 12 months were 0.68, 0.38, and 0.59 events/year for individuals with T1D, T2D new users, and T2D switchers, respectively. CONCLUSION US IBM® Explorys® data demonstrated a clinically relevant reduction in HbA1c 6 months after initiating faster aspart treatment for individuals with T2D, but not T1D overall, although patients with baseline HbA1c > 8.5% had significant HbA1c reductions regardless of diabetes type.
Collapse
Affiliation(s)
- Wendy Lane
- Mountain Diabetes and Endocrine Center, Asheville, NC, USA
| | | | | | | | | | - Christophe De Block
- Department of Endocrinology-Diabetology-Metabolism, Antwerp University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium.
| |
Collapse
|
8
|
Cui T, Li Y, Wei Z, Zhang X, Li W, Zhou W, Lu J, Li J, Yi X, Zeng Y, Liu C, Yan F. Pharmacokinetics, tissue distribution and excretion of a novel long-acting human insulin analogue - recombinant insulin LysArg in rats. Xenobiotica 2020; 51:307-315. [PMID: 33151101 DOI: 10.1080/00498254.2020.1847361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
As a novel long-acting recombinant human insulin analogue, it is necessary to carry out the preclinical research for insulin LysArg. The purpose of this study was to characterise the pharmacokinetic, tissue distribution and excretion of insulin LysArg and provide a reference for its development. Three methods were used to measure the content of insulin LysArg in biological samples after a single subcutaneous administration in rats, including radioassay, radioassay after precipitation with TCA and separation by HPLC. After Subcutaneous administration of recombinant insulin LysArg 1, 2, 4 U/kg in rats, it showed both Cmax and AUC0-t were positively correlated with the dose. In the meanwhile, after a single subcutaneous administration of recombinant insulin LysArg at 2 U/kg in rats, the amount of radioactivity in most organs was highest at 1.5 h and then decreased gradually, no accumulation was found. The highest level of insulin LysArg was observed in the kidney. Like other macromolecules, insulin LysArg was mainly excreted from urine. The study fully illustrated the pharmacokinetic pattern of insulin LysArg, provided valuable informations to support its further development about safety and toxicology.
Collapse
Affiliation(s)
- Tao Cui
- Tianjin Institute of Pharmaceutical Research, State Key Laboratory of Drug Delivery Technologies and Pharmacokinetics , Tianjin, China
| | - Yazhuo Li
- Tianjin Institute of Pharmaceutical Research, State Key Laboratory of Drug Delivery Technologies and Pharmacokinetics , Tianjin, China
| | - Zihong Wei
- Tianjin Institute of Pharmaceutical Research, State Key Laboratory of Drug Delivery Technologies and Pharmacokinetics , Tianjin, China
| | - Xingyan Zhang
- Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Wei Li
- Tianjin Institute of Pharmaceutical Research, State Key Laboratory of Drug Delivery Technologies and Pharmacokinetics , Tianjin, China
| | - Wei Zhou
- Hefei Tianmai Biotechnology Development Co. Ltd , Hefei, China
| | - Jiangjie Lu
- Hefei Tianmai Biotechnology Development Co. Ltd , Hefei, China
| | - Jing Li
- Hefei Tianmai Biotechnology Development Co. Ltd , Hefei, China
| | - Xiulin Yi
- Tianjin Institute of Pharmaceutical Research, State Key Laboratory of Drug Delivery Technologies and Pharmacokinetics , Tianjin, China
| | - Yong Zeng
- Tianjin Institute of Pharmaceutical Research, State Key Laboratory of Drug Delivery Technologies and Pharmacokinetics , Tianjin, China
| | - Changxiao Liu
- Tianjin Institute of Pharmaceutical Research, State Key Laboratory of Drug Delivery Technologies and Pharmacokinetics , Tianjin, China
| | - Fengying Yan
- Tianjin Institute of Pharmaceutical Research, State Key Laboratory of Drug Delivery Technologies and Pharmacokinetics , Tianjin, China.,Research Unit for Drug Metabolism, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|