Clinical Dilemmas in Diabetes. Группа авторов

Чтение книги онлайн.

Читать онлайн книгу Clinical Dilemmas in Diabetes - Группа авторов страница 25

Clinical Dilemmas in Diabetes - Группа авторов

Скачать книгу

TILT Tregs + IL‐2 Safety and Treg proportion at 3 years NCT02804165 Gene‐virus Interactions Implicated in Type 1 Diabetes Enterovirus vaccination T1D diagnosis NCT03243058 A Randomized, Double Blind, Phase I/II Trial of Low‐Dose Interlekin‐2 Immunotherapy in Established Type 1 Diabetes Proleukin (IL‐2) MMTT C‐peptide at 12 months NCT02081326 Repeat BCG Vaccinations for the Treatment of Established Type 1 Diabetes BCG HbA1C at 1,2,3,4 and 5 years EudraCT: 2014‐004319‐35 TregVac2.0 Tregs + anti‐CD20 antibody (Phase II Trial assessing expanded polytTregs in patients with recent onset Type 1 diabetes mellitus) EudraCT 2014‐004760‐37 Incretin‐based therapy in non‐symptomatic, early diagnosed Type 1 Diabetics Liraglutide MMTT C‐peptide at 3, 6, 9, and 12 months

      Pro‐inflammatory cytokine‐based treatments have proven to be safe and effective for treatment of various autoimmune diseases. Thus, inhibition of expression of those molecules can induce important changes in pancreatic β‐cells induce important changes in pancreatic β‐cells [51].

      The aim of using the Anti‐Interleukin‐1 in newly diagnosed T1D subjects is to test the feasibility, safety/tolerability and potential efficacy of anti‐IL‐1 therapy in maintaining or enhancing β‐cell function in people with new onset T1D. Anti‐IL‐1 administration for rheumatoid arthritis has been proven to be well tolerated in patients [52, 53]. IL‐1 is also involved in T1D progression by activating T‐helper cells and improving the number of circulating memory T‐cells [54]. The active substance is interleukin‐1 receptor antagonist, a blocker of an immune‐signal molecule named interleukin‐1. Two randomized placebo‐controlled trials aimed to assess whether canakinumab, a human monoclonal anti‐interleukin‐1 antibody, or anakinra, a human interleukin‐1 receptor antagonist, improved β‐cell function in recent‐onset T1D, but their effectiveness was not demonstrated [54, 55].

      More recently, the ongoing clinical trial EXTEND (Clinical trial NCT02293837; www.clinicaltrials.gov) is currently examining whether the blockade of IL‐6 signaling through tocilizumab, an anti‐IL‐6 receptor antibody, can induce a protection of β‐cell function in T1D patients (ages 6 to 17 years) (Table 2.3).

      Interleukin‐8 appears to be another important mediator in the progression of T1D. Circulating levels of IL‐8 are elevated in children with T1D compared to non‐diabetic controls. Furthermore, levels of IL‐8 correlate with glycemic control, higher level being associated to poorer glucose control. As a result, the modulation or inhibition of IL8 activity may be a valid target for the development of novel treatments aimed to control the progression of T1D.

      A multicenter, randomized, double‐blind, placebo‐controlled phase 2 trial of CXCR1/2 IL‐8 inhibitor (Ladarixin) has just presented its results at the American Diabetes Association's (ADA) 80th Scientific Sessions (Clinical trial NCT02814838; www.clinicaltrials.gov). The trial involved 76 patients with new‐onset T1D, randomly (2:1) assigned to receive either Ladarixin treatment (400 mg b.i.d. for 3 cycles of 14 days on/14 days off – treatment group) or placebo (control group). Although results indicated no statistically significant differences in stimulated C‐peptide at weeks 13 and 26, investigators noted 76.6% of patients receiving Ladarixin had an HbA1c below 7% and a daily insulin requirement of less than 0.50 IU/kg compared to just 45.8% of patients receiving placebo. Furthermore, in a prespecified subgroup analysis of patients with fasting C‐peptide below the median value of the trial population at baseline, MMTT AUC of C‐peptide trended at week 13 and reached statistical significance at week 26.

       Incretin‐based Therapies

      Recent knowledge regarding the heterogeneity in the extent of the β‐cell impairment and pancreatic lesions as well as the differences in circulating T‐cell and autoantibody immune signatures underscore the potential applications for incretin treatments, which improve capacity for insulin production by residual β‐cells and suppress glucagon secretion, as well as the need for therapeutics to reduce β‐cell stress co‐administered with immunomodulatory therapy to reverse autoimmunity in symptomatic T1D. Hence, therapies once considered only applicable to those with T2D may be of potential benefit for those with T1D. In this regard, ongoing T1D immunotherapy trials are investigating the potential benefits on β‐cell function in C‐peptide positive early diagnosed T1D patients.

      Other studies on preservation of β‐cell function using incretin‐based therapies are currently active but not recruiting participants (NCT02443155; NCT02127047). Results from these studies are warranted to prove that incretin‐based therapy might preserve C‐peptide secretion (Table 2.3).

      Today, one of the therapeutic goals in T1D is the preservation of the residual C‐peptide secretion that is detected in a significant percentage of patients at diagnosis and which potentially may influence the clinical course of the disease.

      Several studies have been demonstrated that residual C‐peptide secretion, after T1D diagnosis, depends on genetic factors, the patient's age at the diabetes diagnosis, the number of anti‐islet antibodies, and the residual C‐peptide secretion. In the same way, intensive

Скачать книгу