231 results on '"Greenbaum CJ"'
Search Results
2. B-lymphocyte depletion with rituximab and β-cell function: Two-year results
- Author
-
Gitelman, Stephen, Pescovitz, MD, Greenbaum, CJ, Bundy, B, Becker, DJ, Gitelman, SE, Goland, R, Gottlieb, PA, Marks, JB, Moran, A, and Raskin, P
- Abstract
OBJECTIVE We previously reported that selective depletion of β-lymphocytes with rituximab, an anti-CD20 monoclonal antibody, slowed decline of b-cell function in recentonset type 1 diabetesmellitus (T1DM) at 1 year. Subjects were followed further to determ
- Published
- 2014
3. Costimulation modulation with abatacept in patients with recent-onset type 1 diabetes: Follow-up 1 year aftercessation of treatment
- Author
-
Gitelman, Stephen, Orban, T, Bundy, B, Becker, DJ, DiMeglio, LA, Gitelman, SE, Goland, R, Gottlieb, PA, Greenbaum, CJ, Marks, JB, and Monzavi, R
- Abstract
OBJECTIVE: We previously reported that 2 years of costimulation modulation with abatacept slowed decline of β-cell function in recent-onset type 1 diabetes (T1D). Subsequently, abatacept was discontinued and subjects were followed to determine whether ther
- Published
- 2014
4. Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs β-cell function
- Author
-
Long, SA, Rieck, M, Sanda, S, Bollyky, JB, Samuels, PL, Goland, R, Ahmann, A, Rabinovitch, A, Aggarwal, S, Phippard, D, Turka, LA, Ehlers, MR, Bianchine, PJ, Boyle, KD, Adah, SA, Bluestone, JA, Buckner, JH, and Greenbaum, CJ
- Subjects
Endocrinology & Metabolism ,Clinical Sciences ,Medical Physiology - Published
- 2013
5. Fall in C-peptide during first 2 years from diagnosis: Evidence of at least two distinct phases from composite type 1 diabetes trialnet data
- Author
-
Gitelman, Stephen, Greenbaum, CJ, Beam, CA, Boulware, D, Gitelman, SE, Gottlieb, PA, Herold, KC, Lachin, JM, McGee, P, Palmer, JP, and Pescovitz, MD
- Abstract
Interpretation of clinical trials to alter the decline in β-cell function after diagnosis of type 1 diabetes depends on a robust understanding of the natural history of disease. Combining data from the Type 1 Diabetes TrialNet studies, we describe the natu
- Published
- 2012
6. Intra-islet insulin permits glucose to directly suppress pancreatic A cell function.
- Author
-
Greenbaum, CJ, Havel, PJ, Taborsky, GJ, and Klaff, LJ
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Health Sciences ,Digestive Diseases ,Diabetes ,Autoimmune Disease ,Metabolic and endocrine ,Alloxan ,Animals ,Arginine ,Dogs ,Glucagon ,Glucose ,Hyperglycemia ,Insulin ,Insulin Secretion ,Islets of Langerhans ,Somatostatin ,INSULIN ,GLUCAGON ,HYPERGLYCEMIA ,Medical and Health Sciences ,Immunology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Inhibition of pancreatic glucagon secretion during hyperglycemia could be mediated by (a) glucose, (b) insulin, (c) somatostatin, or (d) glucose in conjunction with insulin. To determine the role of these factors in the mediation of glucagon suppression, we injected alloxan while clamping the arterial supply of the pancreatic splenic lobe of dogs, thus inducing insulin deficiency localized to the ventral lobe and avoiding hyperglycemia. Ventral lobe insulin, glucagon, and somatostatin outputs were then measured in response to a stepped IV glucose infusion. In control dogs glucagon suppression occurred at a glucose level of 150 mg/dl and somatostatin output increased at glucose greater than 250 mg/dl. In alloxan-treated dogs glucagon output was not suppressed nor did somatostatin output increase. We concluded that insulin was required in the mediation of glucagon suppression and somatostatin stimulation. Subsequently, we infused insulin at high rates directly into the artery that supplied the beta cell-deficient lobe in six alloxan-treated dogs. Insulin infusion alone did not cause suppression of glucagon or stimulation of somatostatin; however, insulin repletion during glucose infusions did restore the ability of hyperglycemia to suppress glucagon and stimulate somatostatin. We conclude that intra-islet insulin permits glucose to suppress glucagon secretion and stimulate somatostatin during hyperglycemia.
- Published
- 1991
7. Effect of oral insulin on prevention of diabetes in relatives of patients with T1DM: a randomized clinical trial
- Author
-
Krischer Jp, Skyler Js, undy B, Desmond A. Schatz, and Greenbaum Cj
- Subjects
medicine.medical_specialty ,Randomized controlled trial ,business.industry ,law ,Internal medicine ,Diabetes mellitus ,Insulin ,medicine.medical_treatment ,medicine ,medicine.disease ,business ,law.invention - Published
- 2018
- Full Text
- View/download PDF
8. First Genome-Wide Association Study of Latent Autoimmune Diabetes in Adults Reveals Novel Insights Linking Immune and Metabolic Diabetes
- Author
-
Cousminer, DL, Ahlqvist, E, Mishra, R, Andersen, MK, Chesi, A, Hawa, MI, Davis, A, Hodge, KM, Bradfield, JP, Zhou, K, Guy, VC, Akerlund, M, Wod, M, Fritsche, LG, Vestergaard, H, Snyder, J, Hojlund, K, Linneberg, A, Karajamaki, A, Brandslund, I, Kim, CE, Witte, D, Sorgjerd, EP, Brillon, DJ, Pedersen, O, Beck-Nielsen, H, Grarup, N, Pratley, RE, Rickels, MR, Vella, A, Ovalle, F, Melander, O, Harris, RI, Varvel, S, Grill, VER, Hakonarson, H, Froguel, P, Lonsdale, JT, Mauricio, D, Schloot, NC, Khunti, K, Greenbaum, CJ, Asvold, BO, Yderstraede, KB, Pearson, ER, Schwartz, S, Voight, BF, Hansen, T, Tuomi, T, Boehm, BO, Groop, L, Leslie, RD, Grant, SFA, Cousminer, DL, Ahlqvist, E, Mishra, R, Andersen, MK, Chesi, A, Hawa, MI, Davis, A, Hodge, KM, Bradfield, JP, Zhou, K, Guy, VC, Akerlund, M, Wod, M, Fritsche, LG, Vestergaard, H, Snyder, J, Hojlund, K, Linneberg, A, Karajamaki, A, Brandslund, I, Kim, CE, Witte, D, Sorgjerd, EP, Brillon, DJ, Pedersen, O, Beck-Nielsen, H, Grarup, N, Pratley, RE, Rickels, MR, Vella, A, Ovalle, F, Melander, O, Harris, RI, Varvel, S, Grill, VER, Hakonarson, H, Froguel, P, Lonsdale, JT, Mauricio, D, Schloot, NC, Khunti, K, Greenbaum, CJ, Asvold, BO, Yderstraede, KB, Pearson, ER, Schwartz, S, Voight, BF, Hansen, T, Tuomi, T, Boehm, BO, Groop, L, Leslie, RD, and Grant, SFA
- Abstract
OBJECTIVE: Latent autoimmune diabetes in adults (LADA) shares clinical features with both type 1 and type 2 diabetes; however, there is ongoing debate regarding the precise definition of LADA. Understanding its genetic basis is one potential strategy to gain insight into appropriate classification of this diabetes subtype. RESEARCH DESIGN AND METHODS: We performed the first genome-wide association study of LADA in case subjects of European ancestry versus population control subjects (n = 2,634 vs. 5,947) and compared against both case subjects with type 1 diabetes (n = 2,454 vs. 968) and type 2 diabetes (n = 2,779 vs. 10,396). RESULTS: The leading genetic signals were principally shared with type 1 diabetes, although we observed positive genetic correlations genome-wide with both type 1 and type 2 diabetes. Additionally, we observed a novel independent signal at the known type 1 diabetes locus harboring PFKFB3, encoding a regulator of glycolysis and insulin signaling in type 2 diabetes and inflammation and autophagy in autoimmune disease, as well as an attenuation of key type 1-associated HLA haplotype frequencies in LADA, suggesting that these are factors that distinguish childhood-onset type 1 diabetes from adult autoimmune diabetes. CONCLUSIONS: Our results support the need for further investigations of the genetic factors that distinguish forms of autoimmune diabetes as well as more precise classification strategies.
- Published
- 2018
9. Stability of human rapamycin-expanded CD4+CD25+ T regulatory cells
- Author
-
Orban T, Bundy B, Becker DJ, DiMeglio LA, Gitelman SE, Goland R, Gottlieb PA, Greenbaum CJ, Marks JB, Monzavi R, Moran A, Raskin P, Tresoldi E, Dell'Albani I, Stabilini A, Jofra T, Valle A, Gagliani N, BONDANZA , ATTILIO, RONCAROLO , MARIA GRAZIA, BATTAGLIA, MARCO MARIA, Orban, T, Bundy, B, Becker, Dj, Dimeglio, La, Gitelman, Se, Goland, R, Gottlieb, Pa, Greenbaum, Cj, Marks, Jb, Monzavi, R, Moran, A, Raskin, P, Tresoldi, E, Dell'Albani, I, Stabilini, A, Jofra, T, Valle, A, Gagliani, N, Bondanza, Attilio, Roncarolo, MARIA GRAZIA, and Battaglia, MARCO MARIA
- Subjects
Cell ,Mice, SCID ,C-C chemokine receptor type 6 ,Biology ,T-Lymphocytes, Regulatory ,Immunophenotyping ,Cell therapy ,Mice ,Mice, Inbred NOD ,In vivo ,medicine ,Animals ,Humans ,Cells, Cultured ,Cell Proliferation ,Sirolimus ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,Forkhead Transcription Factors ,Hematology ,Phenotype ,Molecular biology ,In vitro ,Cell biology ,medicine.anatomical_structure ,Online-Only Articles ,CD4 Antigens ,Leukocytes, Mononuclear ,Cytokines ,Th17 Cells ,Female ,Ex vivo - Abstract
"\"\\\"\\\\\\\"Background The clinical use of ex vivo-expanded T-regulatory cells for the treatment of T-cell-mediated diseases has gained increasing momentum. However, the recent demonstration that FOXP3(+) T-regulatory cells may contain interleukin-17-producing cells and that they can convert into effector cells once transferred in vivo raises significant doubts about their safety. We previously showed that rapamycin permits the ex vivo expansion of FOXP3(+) T-regulatory cells while impairing the proliferation of non-T-regulatory cells. Here we investigated the Th17-cell content and the in vivo stability of rapamycin-expanded T-regulatory cells as pertinent aspects of cell-based therapy. Design and Methods T-regulatory-enriched cells were isolated from healthy volunteers and were expanded ex vivo with rapamycin with a pre-clinical applicable protocol. T-regulatory cells cultured with and without rapamycin were compared for their regulatory activity, content of pro-inflammatory cells and stability. Results We found that CD4(+)CCR6(+)CD161(+) T cells (i.e., precursor\\\\\\\\\\\\\\\/committed Th17 cells) contaminate the T-regulatory cells cultured ex vivo in the absence of rapamycin. In addition, Th17 cells do not expand when rapamycin-treated T-regulatory cells are exposed to a \\\\\\\"Th17-favorable\\\\\\\" environment. Rapamycin-expanded T-regulatory cells maintain their in vitro regulatory phenotype even after in vivo transfer into immunodeficient NOD-SCID mice despite being exposed to the irradiation-induced pro-inflammatory environment. Importantly, no additional rapamycin treatment, either in vitro or in vivo, is required to keep their phenotype fixed. Conclusions These data demonstrate that rapamycin secures ex vivo-expanded human T-regulatory cells and provide additional justification for their clinical use in future cell therapy-based trials.\\\\\\\"\\\"\""
- Published
- 2011
- Full Text
- View/download PDF
10. Understanding and preventing type 1 diabetes through the unique working model of TrialNet
- Author
-
Battaglia, M, Anderson, MS, Buckner, JH, Geyer, SM, Gottlieb, PA, Kay, TWH, Lernmark, A, Muller, S, Pugliese, A, Roep, BO, Greenbaum, CJ, Peakman, M, Battaglia, M, Anderson, MS, Buckner, JH, Geyer, SM, Gottlieb, PA, Kay, TWH, Lernmark, A, Muller, S, Pugliese, A, Roep, BO, Greenbaum, CJ, and Peakman, M
- Abstract
Type 1 diabetes is an autoimmune disease arising from the destruction of pancreatic insulin-producing beta cells. The disease represents a continuum, progressing sequentially at variable rates through identifiable stages prior to the onset of symptoms, through diagnosis and into the critical periods that follow, culminating in a variable depth of beta cell depletion. The ability to identify the very earliest of these presymptomatic stages has provided a setting in which prevention strategies can be trialled, as well as furnishing an unprecedented opportunity to study disease evolution, including intrinsic and extrinsic initiators and drivers. This niche opportunity is occupied by Type 1 Diabetes TrialNet, an international consortium of clinical trial centres that leads the field in intervention and prevention studies, accompanied by deep longitudinal bio-sampling. In this review, we focus on discoveries arising from this unique bioresource, comprising more than 70,000 samples, and outline the processes and science that have led to new biomarkers and mechanistic insights, as well as identifying new challenges and opportunities. We conclude that via integration of clinical trials and mechanistic studies, drawing in clinicians and scientists and developing partnership with industry, TrialNet embodies an enviable and unique working model for understanding a disease that to date has no cure and for designing new therapeutic approaches.
- Published
- 2017
11. Effects of Insulin in Relatives of Patients with Type 1 Diabetes Mellitus
- Author
-
W. E. Winter, N. Vega, R. A. Jackson, J. P. Palmer, K. Herold, D. Conboy, F. R. Kaufman, A. Schiffrin, Greenbaum Cj, P. R. Robertson, S. Harris, Desmond A. Schatz, J. Fradkin, J. Wolfsdorf, John M. Lachin, C. Cowie, George S. Eisenbarth, Della Matheson, J. P. Krischer, J. Valenzuela, T. Smith, Jennifer B. Marks, M. McCulloch-Olsen, H. Dickler, Rebecca J. Cook, Jay S. Skyler, J. I. Malone, B. Zinman, A. Zeidler, L. M. Rafkin-Mervis, R. C. Eastman, Darrell M. Wilson, E. Collier, A. T. Ricker, M. Steffes, M. A. Dennis, D. DeMets, N. K. Maclaren, G. Grave, A. Rossini, Lisa Rafkin-Mervis, D. Brown, L. Finney, Anastasios A. Tsiatis, H. P. Chase, D. Cuthbertson, O. B. Crofford, and B. Aneju
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Injections, Subcutaneous ,medicine.medical_treatment ,Islets of Langerhans ,Risk Factors ,Internal medicine ,medicine ,Humans ,Hypoglycemic Agents ,Insulin ,Family ,Treatment Failure ,Child ,Type 1 diabetes ,business.industry ,General Medicine ,Glucose Tolerance Test ,Middle Aged ,medicine.disease ,Hypoglycemia ,Diabetes Mellitus, Type 1 ,Child, Preschool ,Female ,business - Abstract
It is unknown whether insulin therapy can delay or prevent diabetes in nondiabetic relatives of patients with diabetes.In a randomized, controlled, nonblinded clinical trial, we screened 84,228 first-degree and second-degree relatives of patients with diabetes for islet-cell antibodies; 3152 tested positive; 2103 of the 3152 underwent genetic, immunologic, and metabolic staging to quantify their risk; 372 of the 2103 had a projected five-year risk of more than 50 percent; 339 of the 372 (median age, 11.2 years) were randomly assigned to undergo either close observation or an intervention that consisted of low-dose subcutaneous ultralente insulin, administered twice daily for a total dose of 0.25 unit per kilogram of body weight per day, plus annual four-day continuous intravenous infusions of insulin. Oral glucose-tolerance tests were performed every six months. Median follow-up was 3.7 years. The primary end point was a diagnosis of diabetes.Diabetes was diagnosed in 69 subjects in the intervention group and 70 subjects in the observation group. The annualized rate of progression to diabetes was 15.1 percent in the intervention group and 14.6 percent in the observation group. The cumulative incidence of diabetes was similar in the two groups (relative risk in the intervention group as compared with the observation group, 0.96). Most subjects in whom diabetes developed were asymptomatic. Progression to diabetes occurred at a faster rate among subjects with abnormal base-line glucose tolerance (22 percent per year) than among those with normal base-line glucose tolerance (10 percent per year, P0.001). There were no episodes of severe hypoglycemia. The incidence of chemical hypoglycemia, assessed without ascertainment bias, was similar in the two groups.In persons at high risk for diabetes, insulin at the dosage used in this study does not delay or prevent type 1 diabetes.
- Published
- 2002
- Full Text
- View/download PDF
12. Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs β-cell function.
- Author
-
Long SA, Rieck M, Sanda S, Bollyky JB, Samuels PL, Goland R, Ahmann A, Rabinovitch A, Aggarwal S, Phippard D, Turka LA, Ehlers MR, Bianchine PJ, Boyle KD, Adah SA, Bluestone JA, Buckner JH, Greenbaum CJ, for Diabetes TrialNet and the Immune Tolerance Network, and Long, S Alice
- Abstract
Rapamycin/interleukin-2 (IL-2) combination treatment of NOD mice effectively treats autoimmune diabetes. We performed a phase 1 clinical trial to test the safety and immunologic effects of rapamycin/IL-2 combination therapy in type 1 diabetic (T1D) patients. Nine T1D subjects were treated with 2-4 mg/day rapamycin orally for 3 months and 4.5 × 10(6) IU IL-2 s.c. three times per week for 1 month. β-Cell function was monitored by measuring C-peptide. Immunologic changes were monitored using flow cytometry and serum analyses. Regulatory T cells (Tregs) increased within the first month of therapy, yet clinical and metabolic data demonstrated a transient worsening in all subjects. The increase in Tregs was transient, paralleling IL-2 treatment, whereas the response of Tregs to IL-2, as measured by STAT5 phosphorylation, increased and persisted after treatment. No differences were observed in effector T-cell subset frequencies, but an increase in natural killer cells and eosinophils occurred with IL-2 therapy. Rapamycin/IL-2 therapy, as given in this phase 1 study, resulted in transient β-cell dysfunction despite an increase in Tregs. Such results highlight the difficulties in translating therapies to the clinic and emphasize the importance of broadly interrogating the immune system to evaluate the effects of therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
13. Fall in C-peptide during first 2 years from diagnosis: evidence of at least two distinct phases from composite Type 1 Diabetes TrialNet data.
- Author
-
Greenbaum CJ, Beam CA, Boulware D, Gitelman SE, Gottlieb PA, Herold KC, Lachin JM, McGee P, Palmer JP, Pescovitz MD, Krause-Steinrauf H, Skyler JS, Sosenko JM, Type 1 Diabetes TrialNet Study Group, Greenbaum, Carla J, Beam, Craig A, Boulware, David, Gitelman, Stephen E, Gottlieb, Peter A, and Herold, Kevan C
- Abstract
Interpretation of clinical trials to alter the decline in β-cell function after diagnosis of type 1 diabetes depends on a robust understanding of the natural history of disease. Combining data from the Type 1 Diabetes TrialNet studies, we describe the natural history of β-cell function from shortly after diagnosis through 2 years post study randomization, assess the degree of variability between patients, and investigate factors that may be related to C-peptide preservation or loss. We found that 93% of individuals have detectable C-peptide 2 years from diagnosis. In 11% of subjects, there was no significant fall from baseline by 2 years. There was a biphasic decline in C-peptide; the C-peptide slope was -0.0245 pmol/mL/month (95% CI -0.0271 to -0.0215) through the first 12 months and -0.0079 (-0.0113 to -0.0050) from 12 to 24 months (P < 0.001). This pattern of fall in C-peptide over time has implications for understanding trial results in which effects of therapy are most pronounced early and raises the possibility that there are time-dependent differences in pathophysiology. The robust data on the C-peptide obtained under clinical trial conditions should be used in planning and interpretation of clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
14. The application of the diabetes prevention trial-type 1 risk score for identifying a preclinical state of type 1 diabetes.
- Author
-
Sosenko JM, Skyler JS, Mahon J, Krischer JP, Beam CA, Boulware DC, Greenbaum CJ, Rafkin LE, Cowie C, Cuthbertson D, Palmer JP, Type 1 Diabetes TrialNet Study Group, Sosenko, Jay M, Skyler, Jay S, Mahon, Jeffrey, Krischer, Jeffrey P, Beam, Craig A, Boulware, David C, Greenbaum, Carla J, and Rafkin, Lisa E
- Abstract
Objective: We assessed the utility of the Diabetes Prevention Trial-Type 1 Risk Score (DPTRS) for identifying individuals who are highly likely to progress to type 1 diabetes (T1D) within 2 years.Research Design and Methods: The DPTRS was previously developed from Diabetes Prevention Trial-Type 1 (DPT-1) data and was subsequently validated in the TrialNet Natural History Study (TNNHS). DPTRS components included C-peptide and glucose indexes from oral glucose tolerance testing, along with age and BMI. The cumulative incidence of T1D was determined after DPTRS thresholds were first exceeded and after the first occurrences of glucose abnormalities.Results: The 2-year risks after the 9.00 DPTRS threshold was exceeded were 0.88 and 0.77 in DPT-1 (n = 90) and the TNNHS (n = 69), respectively. In DPT-1, the 2-year risks were much lower after dysglycemia first occurred (0.37; n = 306) and after a 2-h glucose value between 190 and 199 mg/dL was first reached (0.64; n = 59). Among those who developed T1D in DPT-1, the 9.00 threshold was exceeded 0.81 ± 0.53 years prior to the conventional diagnosis. Postchallenge C-peptide levels were substantially higher (P = 0.001 for 30 min; P < 0.001 for other time points) when the 9.00 threshold was first exceeded compared with the levels at diagnosis.Conclusions: A DPTRS threshold of 9.00 identifies individuals who are very highly likely to progress to the conventional diagnosis of T1D within 2 years and, thus, are essentially in a preclinical diabetic state. The 9.00 threshold is exceeded well before diagnosis, when stimulated C-peptide levels are substantially higher. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
15. Through the fog: recent clinical trials to preserve β-cell function in type 1 diabetes.
- Author
-
Greenbaum CJ, Schatz DA, Haller MJ, Sanda S, Greenbaum, Carla J, Schatz, Desmond A, Haller, Michael J, and Sanda, Srinath
- Published
- 2012
- Full Text
- View/download PDF
16. Failure to preserve beta-cell function with mycophenolate mofetil and daclizumab combined therapy in patients with new- onset type 1 diabetes.
- Author
-
Gottlieb PA, Quinlan S, Krause-Steinrauf H, Greenbaum CJ, Wilson DM, Rodriguez H, Schatz DA, Moran AM, Lachin JM, Skyler JS, Type 1 Diabetes TrialNet MMF/DZB Study Group, Gottlieb, Peter A, Quinlan, Scott, Krause-Steinrauf, Heidi, Greenbaum, Carla J, Wilson, Darrell M, Rodriguez, Henry, Schatz, Desmond A, Moran, Antoinette M, and Lachin, John M
- Abstract
Objective: This trial tested whether mycophenolate mofetil (MMF) alone or with daclizumab (DZB) could arrest the loss of insulin-producing beta-cells in subjects with new-onset type 1 diabetes.Research Design and Methods: A multi-center, randomized, placebo-controlled, double-masked trial was initiated by Type 1 Diabetes TrialNet at 13 sites in North America and Europe. Subjects diagnosed with type 1 diabetes and with sufficient C-peptide within 3 months of diagnosis were randomized to either MMF alone, MMF plus DZB, or placebo, and then followed for 2 years. The primary outcome was the geometric mean area under the curve (AUC) C-peptide from the 2-h mixed meal tolerance test.Results: One hundred and twenty-six subjects were randomized and treated during the trial. The geometric mean C-peptide AUC at 2 years was unaffected by MMF alone or MMF plus DZB versus placebo. Adverse events were more frequent in the active therapy groups relative to the control group, but not significantly.Conclusions: Neither MMF alone nor MMF in combination with DZB had an effect on the loss of C-peptide in subjects with new-onset type 1 diabetes. Higher doses or more targeted immunotherapies may be needed to affect the autoimmune process. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
17. Preservation of beta-cell function in autoantibody-positive youth with diabetes.
- Author
-
Greenbaum CJ, Anderson AM, Dolan LM, Mayer-Davis EJ, Dabelea D, Imperatore G, Marcovina S, Pihoker C, SEARCH Study Group, Greenbaum, Carla J, Anderson, Andrea M, Dolan, Lawrence M, Mayer-Davis, Elizabeth J, Dabelea, Dana, Imperatore, Giuseppina, Marcovina, Santica, and Pihoker, Catherine
- Abstract
Objective: To determine the extent of beta-cell function in youth with diabetes and GAD65 and/or IA2 autoantibodies.Research Design and Methods: Fasting C-peptide levels from 2,789 GAD65- and/or IA2 autoantibody-positive youth aged 1-23 years from the SEARCH for Diabetes in Youth study were used. Preserved beta-cell function was defined on the basis of cut points derived from the Diabetes Control and Complications Trial (DCCT) (fasting C-peptide > or =0.23 ng/ml) and from the U.S. adolescent population of the National Health and Nutrition Examination Survey (NHANES) 5th percentile for fasting C-peptide (> or =1.0 ng/ml). We compared the clinical characteristics between those with and without preserved beta-cell function.Results: Within the first year of diagnosis, 82.9% of youth had a fasting C-peptide > or =0.23 ng/ml and 31.2% had values > or =1.0 ng/ml. Among those with > or =5 years of diabetes duration, 10.7% had preserved beta-cell function based on the DCCT cutoff and 1.0% were above the 5th percentile of the NHANES population.Conclusions: Within the 1st year of diagnosis, four of five youth with autoantibody-positive diabetes have clinically significant amounts of residual beta-cell function and about one-third have fasting C-peptide levels above the 5th percentile of a healthy adolescent population. Even 5 years after diagnosis, 1 of 10 has fasting C-peptide above a clinically significant threshold. These findings have implications for clinical classification of youth with diabetes as well as clinical trials aimed to preserve beta-cell function after diabetes onset. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
18. Mixed-meal tolerance test versus glucagon stimulation test for the assessment of beta-cell function in therapeutic trials in type 1 diabetes.
- Author
-
Greenbaum CJ, Mandrup-Poulsen T, McGee PF, Battelino T, Haastert B, Ludvigsson J, Pozzilli P, Lachin JM, Kolb H, Greenbaum, Carla J, Mandrup-Poulsen, Thomas, McGee, Paula Friedenberg, Battelino, Tadej, Haastert, Burkhard, Ludvigsson, Johnny, Pozzilli, Paolo, Lachin, John M, Kolb, Hubert, Type 1 Diabetes Trial Net Research Group, and European C-Peptide Trial Study Group
- Abstract
Objective: Beta-cell function in type 1 diabetes clinical trials is commonly measured by C-peptide response to a secretagogue in either a mixed-meal tolerance test (MMTT) or a glucagon stimulation test (GST). The Type 1 Diabetes TrialNet Research Group and the European C-peptide Trial (ECPT) Study Group conducted parallel randomized studies to compare the sensitivity, reproducibility, and tolerability of these procedures.Research Design and Methods: In randomized sequences, 148 TrialNet subjects completed 549 tests with up to 2 MMTT and 2 GST tests on separate days, and 118 ECPT subjects completed 348 tests (up to 3 each) with either two MMTTs or two GSTs.Results: Among individuals with up to 4 years' duration of type 1 diabetes, >85% had measurable stimulated C-peptide values. The MMTT stimulus produced significantly higher concentrations of C-peptide than the GST. Whereas both tests were highly reproducible, the MMTT was significantly more so (R(2) = 0.96 for peak C-peptide response). Overall, the majority of subjects preferred the MMTT, and there were few adverse events. Some older subjects preferred the shorter duration of the GST. Nausea was reported in the majority of GST studies, particularly in the young age-group.Conclusions: The MMTT is preferred for the assessment of beta-cell function in therapeutic trials in type 1 diabetes. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
19. Increasing the accuracy of oral glucose tolerance testing and extending its application to individuals with normal glucose tolerance for the prediction of type 1 diabetes: the Diabetes Prevention Trial-Type 1.
- Author
-
Sosenko JM, Palmer JP, Greenbaum CJ, Mahon J, Cowie C, Krischer JP, Chase HP, White NH, Buckingham B, Herold KC, Cuthbertson D, Skyler JS, Sosenko, Jay M, Palmer, Jerry P, Greenbaum, Carla J, Mahon, Jeffrey, Cowie, Catherine, Krischer, Jeffrey P, Chase, H Peter, and White, Neil H
- Abstract
Objective: We assessed the extent to which both standard and alternative indexes from 2-h oral glucose tolerance testing predict type 1 diabetes and whether oral glucose tolerance tests (OGTTs) predict type 1 diabetes in individuals with normal glucose tolerance.Research Design and Methods: The prediction of type 1 diabetes from baseline OGTTs was studied in 704 Diabetes Prevention Trial-Type 1 participants (islet-cell autoantibody [ICA]-positive relatives of type 1 diabetic patients). The maximum follow-up was 7.4 years. Analyses utilized receiver-operator curves (ROCs), proportional hazards models, and survival curves.Results: ROC areas under the curve (ROCAUCs) for both the AUC glucose (0.73 +/- 0.02) and an OGTT prediction index (0.78 +/- 0.02) were higher (P < 0.001) than those for the fasting (0.53 +/- 0.02) and 2-h glucose (0.66 +/- 0.02). ROCAUCs for the 60- and 90-min glucose (0.71 +/- 0.02 and 0.72 +/- 0.02, respectively) were also higher (P < 0.01) than those for the fasting and 2-h glucose. Among individuals with normal glucose tolerance, OGTTs were highly predictive, with 4th versus 1st quartile hazard ratios for the 2-h glucose, AUC glucose, and OGTT prediction index ranging from 3.77 to 5.30 (P < 0.001 for all).Conclusions: Certain alternative OGTT indexes appear to better predict type 1 diabetes than standard OGTT indexes in ICA-positive relatives of type 1 diabetic patients. Moreover, even among those with normal glucose tolerance, OGTTs are strongly predictive. This suggests that subtle metabolic abnormalities are present several years before the diagnosis of type 1 diabetes. [ABSTRACT FROM AUTHOR]- Published
- 2007
- Full Text
- View/download PDF
20. Analysis of T-cell assays to measure autoimmune responses in subjects with type 1 diabetes: results of a blinded controlled study.
- Author
-
Seyfert-Margolis V, Gisler TD, Asare AL, Wang RS, Dosch HM, Brooks-Worrell B, Eisenbarth GS, Palmer JP, Greenbaum CJ, Gitelman SE, Nepom GT, Bluestone JA, and Herold KC
- Abstract
Type 1 diabetes is a chronic autoimmune disease mediated by autoreactive T-cells. Several experimental therapies targeting T-cells are in clinical trials. To understand how these therapies affect T-cell responses in vivo, assays that directly measure human T-cell function are needed. In a blinded, multicenter, case-controlled study conducted by the Immune Tolerance Network, we tested responses in an immunoblot and T-cell proliferative assay to distinguish type 1 diabetic patients from healthy control subjects. Peripheral blood cells from 39 healthy control subjects selected for DR4 and 23 subjects with recently diagnosed type 1 diabetes were studied. Autoantibody responses were measured in serum samples. Positive responses in both assays were more common in peripheral blood mononuclear cells from new-onset type 1 diabetic patients compared with control subjects. The proliferative, immunoblot, and autoantibody assays had sensitivities of 58, 91, and 78% with specificities of 94, 83, and 85%, respectively. When cellular assays were combined with autoantibody measurements, the sensitivity of the measurements was 75% with 100% specificity. We conclude that cellular assays performed on peripheral blood have a high degree of accuracy in discriminating responses in subjects with type 1 diabetes from healthy control subjects. They may be useful for assessment of cellular autoimmune responses involved in type 1 diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
21. Patterns of metabolic progression to type 1 diabetes in the Diabetes Prevention Trial-Type 1.
- Author
-
Sosenko JM, Palmer JP, Greenbaum CJ, Mahon J, Cowie C, Krischer JP, Chase HP, White NH, Buckingham B, Herold KC, Cuthbertson D, Skyler JS, Sosenko, Jay M, Palmer, Jerry P, Greenbaum, Carla J, Mahon, Jeffrey, Cowie, Catherine, Krischer, Jeffrey P, Chase, H Peter, and White, Neil H
- Abstract
Objective: There is little information regarding the pattern of metabolic deterioration before the onset of type 1 diabetes. The goal of this study was to utilize data from the Diabetes Prevention Trial-Type 1 (DPT-1) to obtain a picture of the metabolic progression to type 1 diabetes over a period of approximately 2.5 years before its diagnosis.Research Design and Methods: Fifty-four DPT-1 participants (22 in the parenteral trial and 32 in the oral trial) were studied. All had oral glucose tolerance tests (OGTTs) at 6-month intervals from approximately 30 to 6 months before diagnosis. The vast majority also had OGTTs at diagnosis. Changes in OGTT glucose and C-peptide indexes from 30 to 6 months before diagnosis were examined by calculating slopes of the indexes for each individual over that time period. Changes from 6 months before diagnosis to diagnosis were examined by paired comparisons of the OGTT metabolic indexes between the time points.Results: Glucose levels increased gradually from 30 to 6 months before diagnosis in both the parenteral and oral groups (P < 0.001 for all indexes). Area under the curve (AUC) C-peptide (P < 0.05) and AUC C-peptide-to-AUC glucose ratio (P < 0.001) values decreased in the oral group; peak C-peptide-to-2-h glucose ratio values decreased in both groups (P < 0.001). In participants who also had OGTTs at diagnosis, AUC C-peptide (parenteral group, P < 0.05) and peak C-peptide (oral group, P < 0.05) values decreased from the last 6 months before diagnosis; stimulated C-peptide-to-glucose ratio values decreased in both groups (P < 0.001). Conversely, fasting C-peptide levels increased in both groups (oral group, P < 0.01). Fasting C-peptide-to-fasting glucose ratio values remained constant throughout the 30-month follow-up.Conclusions: These data indicate that over a period of at least 2 years, glucose tolerance gradually deteriorates as stimulated C-peptide levels slowly decline in a substantial number of individuals who develop type 1 diabetes. However, fasting C-peptide levels are maintained, even at diagnosis. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
22. Type I diabetes manifested solely by 2-h oral glucose tolerance test criteria.
- Author
-
Greenbaum CJ, Cuthbertson D, Krischer JP, Diabetes Prevention Trial of Type 1 Diabetes Study Group, Greenbaum, C J, Cuthbertson, D, Krischer, J P, and Disease Prevention Trial of Type I Diabetes Study Group
- Abstract
The clinical presentation of type 1 diabetes usually involves symptoms such as polyuria and polydipsia. However, investigators in the Diabetes Prevention Trial of Type 1 Diabetes (DPT-1) have detected a group of subjects with type 1 diabetes who have a different phenotype. These subjects are asymptomatic, have normal (<6.1 mmol/l) (group A) or impaired (6.1- <7.0 mmol/l) (group B) fasting glucose, but have 2-h glucose values >11.1 mmol/l on their oral glucose tolerance tests (OGTT). Of the 585 OGTTs performed on islet cell antibody (ICA)-positive relatives with insulin autoantibodies (IAA) or low first-phase insulin response (FPIR), normal glucose tolerance (NGT) was found in 427 subjects; impaired glucose tolerance (IGT) was found in 87 subjects, and diabetes was found by 2-h OGTT criteria alone in 61 subjects. Despite marked differences in 2-h glucose values (NGT 5.8 +/- 1.1 mmol/l, IGT 8.9 +/- 0.9 mmol/l, and group A 13.5 +/- 2.5 mmol/l), there were no significant differences in fasting glucose values among NGT (4.8 +/- 0.5 mmol/l), IGT (5.03 +/- 0.5 mmol/l), and group A (4.99 +/- 0.7 mmol/l) categories. Mean FPIR was higher in subjects with NGT compared with subjects with IGT and subjects diagnosed by 2-h OGTT criteria alone. However, the correlation between FPIR and 2-h glucose value was low (r2 = 0.114). Multivariate analysis demonstrated that additional independent variables provide smaller contributions to the 2-h glucose value. In conclusion, there are asymptomatic type 1 diabetic subjects whose diabetes was diagnosed by the 2-h criteria on OGTT alone. Despite the importance of beta-cell dysfunction in the pathogenesis of type I diabetes, factors other than impaired FPIR must also contribute to postprandial glucose tolerance in these subjects. [ABSTRACT FROM AUTHOR]
- Published
- 2001
- Full Text
- View/download PDF
23. Dead or alive?
- Author
-
Greenbaum CJ and Greenbaum, Carla J
- Published
- 2012
- Full Text
- View/download PDF
24. Comment on: Atkinson. It's time to consider changing the rules: the rationale for rethinking control groups in clinical trials aimed at reversing type 1 diabetes. Diabetes 2011;60:361-363.
- Author
-
Sanda S, Greenbaum CJ, Sanda, Srinath, and Greenbaum, Carla J
- Published
- 2011
- Full Text
- View/download PDF
25. Beyond Stages: Predicting Individual Time Dependent Risk for Type 1 Diabetes.
- Author
-
Pribitzer S, O'Rourke C, Ylescupidez A, Smithmyer M, Bender C, Speake C, Lord S, and Greenbaum CJ
- Subjects
- Humans, Female, Male, Adult, Adolescent, Glucose Tolerance Test, Young Adult, Child, Risk Assessment methods, Risk Factors, Glycated Hemoglobin analysis, Time Factors, Middle Aged, Diabetes Mellitus, Type 1, Disease Progression, Blood Glucose analysis, Blood Glucose metabolism, Autoantibodies blood, Autoantibodies immunology
- Abstract
Background: Essentially all individuals with multiple autoantibodies will develop clinical type 1 diabetes. Multiple autoantibodies (AABs) and normal glucose tolerance define stage 1 diabetes; abnormal glucose tolerance defines stage 2. However, the rate of progression within these stages is heterogeneous, necessitating personalized risk calculators to improve clinical implementation., Methods: We developed 3 models using TrialNet's Pathway to Prevention data to accommodate the reality that not all risk variables are clinically available. The small model included AAB status, fasting glucose, hemoglobin A1c, and age, while the medium and large models added predictors of disease progression measured via oral glucose tolerance testing., Findings: All models markedly improved granularity regarding personalized risk missing from current categories of stages of type 1 diabetes. Model-derived risk calculations are consistent with the expected reduction of risk with increasing age and increase in risk with higher glucose and lower insulin secretion, illustrating the suitability of the models. Adding glucose and insulin secretion data altered model predicted probabilities within stages. In those with high 2-hour glucose, a high C-peptide markedly decreased predicted risk; a lower C-peptide obviated the age-dependent risk of 2-hour glucose alone, providing a more nuanced estimate of the rate of disease progression within stage 2., Conclusion: While essentially all those with multiple AABs will develop type 1 diabetes, the rate of progression is heterogeneous and not explained by any individual single risk variable. The model-based probabilities developed here provide an adaptable personalized risk calculator to better inform decisions about how and when to monitor disease progression in clinical practice., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
26. Erratum. Consensus Guidance for Monitoring Individuals With Islet Autoantibody-Positive Pre-Stage 3 Type 1 Diabetes. Diabetes Care 2024;47:1276-1298.
- Author
-
Phillip M, Achenbach P, Addala A, Albanese-O'Neill A, Battelino T, Bell KJ, Besser REJ, Bonifacio E, Colhoun HM, Couper JJ, Craig ME, Danne T, Beaufort C, Dovc K, Driscoll KA, Dutta S, Ebekozien O, Larsson HE, Feiten DJ, Frohnert BI, Gabbay RA, Gallagher MP, Greenbaum CJ, Griffin KJ, Hagopian W, Haller MJ, Hendrieckx C, Hendriks E, Holt RIG, Hughes L, Ismail HM, Jacobsen LM, Johnson SB, Kolb LE, Kordonouri O, Lange K, Lash RW, Lernmark Å, Libman I, Lundgren M, Maahs DM, Marcovecchio ML, Mathieu C, Miller KM, O'Donnell HK, Oron T, Patil SP, Pop-Busui R, Rewers MJ, Rich SS, Schatz DA, Schulman-Rosenbaum R, Simmons KM, Sims EK, Skyler JS, Smith LB, Speake C, Steck AK, Thomas NPB, Tonyushkina KN, Veijola R, Wentworth JM, Wherrett DK, Wood JR, Ziegler AG, and DiMeglio LA
- Published
- 2024
- Full Text
- View/download PDF
27. Evolving Concepts in Pathophysiology, Screening, and Prevention of Type 1 Diabetes: Report of Diabetes Mellitus Interagency Coordinating Committee Workshop.
- Author
-
Greenbaum CJ, Nepom GT, Wood-Heickman LK, Wherrett DK, DiMeglio LA, Herold KC, and Krischer JP
- Subjects
- Humans, United States epidemiology, Mass Screening, Antibodies, Monoclonal, Humanized therapeutic use, Diabetes Mellitus, Type 1 immunology, Diabetes Mellitus, Type 1 physiopathology
- Abstract
The approval of teplizumab to delay the onset of type 1 diabetes is an important inflection point in the decades-long pursuit to treat the cause of the disease rather than its symptoms. The National Institute of Diabetes and Digestive and Kidney Diseases convened a workshop of the Diabetes Mellitus Interagency Coordinating Committee titled "Evolving Concepts in Pathophysiology, Screening, and Prevention of Type 1 Diabetes" to review this accomplishment and identify future goals. Speakers representing Type 1 Diabetes TrialNet (TrialNet) and the Immune Tolerance Network emphasized that the ability to robustly identify individuals destined to develop type 1 diabetes was essential for clinical trials. The presenter from the U.S. Food and Drug Administration described how regulatory approval relied on data from the single clinical trial of TrialNet with testing of teplizumab for delay of clinical diagnosis, along with confirmatory evidence from studies in patients after diagnosis. The workshop reviewed the etiology of type 1 diabetes as a disease involving multiple immune pathways, highlighting the current understanding of prognostic markers and proposing potential strategies to improve the therapeutic response of disease-modifying therapies based on the mechanism of action. While celebrating these achievements funded by the congressionally appropriated Special Diabetes Program, panelists from professional organizations, nonprofit advocacy/funding groups, and industry also identified significant hurdles in translating this research into clinical care., (© 2024 by the American Diabetes Association.)
- Published
- 2024
- Full Text
- View/download PDF
28. Correction to: Consensus guidance for monitoring individuals with islet autoantibody‑positive pre‑stage 3 type 1 diabetes.
- Author
-
Phillip M, Achenbach P, Addala A, Albanese-O'Neill A, Battelino T, Bell KJ, Besser REJ, Bonifacio E, Colhoun HM, Couper JJ, Craig ME, Danne T, de Beaufort C, Dovc K, Driscoll KA, Dutta S, Ebekozien O, Larsson HE, Feiten DJ, Frohnert BI, Gabbay RA, Gallagher MP, Greenbaum CJ, Griffin KJ, Hagopian W, Haller MJ, Hendrieckx C, Hendriks E, Holt RIG, Hughes L, Ismail HM, Jacobsen LM, Johnson SB, Kolb LE, Kordonouri O, Lange K, Lash RW, Lernmark Å, Libman I, Lundgren M, Maahs DM, Marcovecchio ML, Mathieu C, Miller KM, O'Donnell HK, Oron T, Patil SP, Pop-Busui R, Rewers MJ, Rich SS, Schatz DA, Schulman-Rosenbaum R, Simmons KM, Sims EK, Skyler JS, Smith LB, Speake C, Steck AK, Thomas NPB, Tonyushkina KN, Veijola R, Wentworth JM, Wherrett DK, Wood JR, Ziegler AG, and DiMeglio LA
- Published
- 2024
- Full Text
- View/download PDF
29. Consensus guidance for monitoring individuals with islet autoantibody-positive pre-stage 3 type 1 diabetes.
- Author
-
Phillip M, Achenbach P, Addala A, Albanese-O'Neill A, Battelino T, Bell KJ, Besser REJ, Bonifacio E, Colhoun HM, Couper JJ, Craig ME, Danne T, de Beaufort C, Dovc K, Driscoll KA, Dutta S, Ebekozien O, Larsson HE, Feiten DJ, Frohnert BI, Gabbay RA, Gallagher MP, Greenbaum CJ, Griffin KJ, Hagopian W, Haller MJ, Hendrieckx C, Hendriks E, Holt RIG, Hughes L, Ismail HM, Jacobsen LM, Johnson SB, Kolb LE, Kordonouri O, Lange K, Lash RW, Lernmark Å, Libman I, Lundgren M, Maahs DM, Marcovecchio ML, Mathieu C, Miller KM, O'Donnell HK, Oron T, Patil SP, Pop-Busui R, Rewers MJ, Rich SS, Schatz DA, Schulman-Rosenbaum R, Simmons KM, Sims EK, Skyler JS, Smith LB, Speake C, Steck AK, Thomas NPB, Tonyushkina KN, Veijola R, Wentworth JM, Wherrett DK, Wood JR, Ziegler AG, and DiMeglio LA
- Subjects
- Humans, Consensus, Islets of Langerhans immunology, Disease Progression, Diabetic Ketoacidosis diagnosis, Diabetic Ketoacidosis immunology, Diabetes Mellitus, Type 1 immunology, Diabetes Mellitus, Type 1 diagnosis, Autoantibodies immunology, Autoantibodies blood
- Abstract
Given the proven benefits of screening to reduce diabetic ketoacidosis (DKA) likelihood at the time of stage 3 type 1 diabetes diagnosis, and emerging availability of therapy to delay disease progression, type 1 diabetes screening programmes are being increasingly emphasised. Once broadly implemented, screening initiatives will identify significant numbers of islet autoantibody-positive (IAb
+ ) children and adults who are at risk of (confirmed single IAb+ ) or living with (multiple IAb+ ) early-stage (stage 1 and stage 2) type 1 diabetes. These individuals will need monitoring for disease progression; much of this care will happen in non-specialised settings. To inform this monitoring, JDRF in conjunction with international experts and societies developed consensus guidance. Broad advice from this guidance includes the following: (1) partnerships should be fostered between endocrinologists and primary-care providers to care for people who are IAb+ ; (2) when people who are IAb+ are initially identified there is a need for confirmation using a second sample; (3) single IAb+ individuals are at lower risk of progression than multiple IAb+ individuals; (4) individuals with early-stage type 1 diabetes should have periodic medical monitoring, including regular assessments of glucose levels, regular education about symptoms of diabetes and DKA, and psychosocial support; (5) interested people with stage 2 type 1 diabetes should be offered trial participation or approved therapies; and (6) all health professionals involved in monitoring and care of individuals with type 1 diabetes have a responsibility to provide education. The guidance also emphasises significant unmet needs for further research on early-stage type 1 diabetes to increase the rigour of future recommendations and inform clinical care., (© 2024. American Diabetes Association and European Association for the Study of Diabetes.)- Published
- 2024
- Full Text
- View/download PDF
30. Evidence for C-Peptide as a Validated Surrogate to Predict Clinical Benefits in Trials of Disease-Modifying Therapies for Type 1 Diabetes.
- Author
-
Latres E, Greenbaum CJ, Oyaski ML, Dayan CM, Colhoun HM, Lachin JM, Skyler JS, Rickels MR, Ahmed ST, Dutta S, Herold KC, and Marinac M
- Subjects
- Humans, Clinical Trials as Topic, Diabetes Mellitus, Type 1 drug therapy, C-Peptide metabolism, C-Peptide blood, Biomarkers blood, Biomarkers metabolism, Insulin-Secreting Cells metabolism, Insulin-Secreting Cells drug effects
- Abstract
Type 1 diabetes is a chronic autoimmune disease in which destruction of pancreatic β-cells causes life-threatening metabolic dysregulation. Numerous approaches are envisioned for new therapies, but limitations of current clinical outcome measures are significant disincentives to development efforts. C-peptide, a direct byproduct of proinsulin processing, is a quantitative biomarker of β-cell function that is not cleared by the liver and can be measured in the peripheral blood. Studies of quantitative measures of β-cell function have established a predictive relationship between stimulated C-peptide as a measure of β-cell function and clinical benefits. C-peptide levels at diagnosis are often high enough to afford glycemic control benefits associated with protection from end-organ complications of diabetes, and even lower levels offer protection from severe hypoglycemia in type 1 diabetes, as observed in large prospective cohort studies and interventional trials of islet transplantation. These observations support consideration of C-peptide not just as a biomarker of β-cell function but also as a specific, sensitive, feasible, and clinically meaningful outcome defining β-cell preservation or restoration for clinical trials of disease-modifying therapies. Regulatory acceptance of C-peptide as a validated surrogate for demonstration of efficacy would greatly facilitate development of disease-modifying therapies for type 1 diabetes., (© 2024 by the American Diabetes Association.)
- Published
- 2024
- Full Text
- View/download PDF
31. OGTT Metrics Surpass Continuous Glucose Monitoring Data for T1D Prediction in Multiple-Autoantibody-Positive Individuals.
- Author
-
Ylescupidez A, Speake C, Pietropaolo SL, Wilson DM, Steck AK, Sherr JL, Gaglia JL, Bender C, Lord S, and Greenbaum CJ
- Subjects
- Humans, Glucose Tolerance Test, Blood Glucose metabolism, Autoantibodies, Blood Glucose Self-Monitoring, Continuous Glucose Monitoring, Diabetes Mellitus, Type 1 diagnosis
- Abstract
Context: The value of continuous glucose monitoring (CGM) for monitoring autoantibody (AAB)-positive individuals in clinical trials for progression of type 1 diabetes (T1D) is unknown., Objective: Compare CGM with oral glucose tolerance test (OGTT)-based metrics in prediction of T1D., Methods: At academic centers, OGTT and CGM data from multiple-AAB relatives were evaluated for associations with T1D diagnosis. Participants were multiple-AAB-positive individuals in a TrialNet Pathway to Prevention (TN01) CGM ancillary study (n = 93). The intervention was CGM for 1 week at baseline, 6 months, and 12 months. Receiver operating characteristic (ROC) curves of CGM and OGTT metrics for prediction of T1D were analyzed., Results: Five of 7 OGTT metrics and 29/48 CGM metrics but not HbA1c differed between those who subsequently did or did not develop T1D. ROC area under the curve (AUC) of individual CGM values ranged from 50% to 69% and increased when adjusted for age and AABs. However, the highest-ranking metrics were derived from OGTT: 4/7 with AUC ∼80%. Compared with adjusted multivariable models using CGM data, OGTT-derived variables, Index60 and DPTRS (Diabetes Prevention Trial-Type 1 Risk Score), had higher discriminative ability (higher ROC AUC and positive predictive value with similar negative predictive value)., Conclusion: Every 6-month CGM measures in multiple-AAB-positive individuals are predictive of subsequent T1D, but less so than OGTT-derived variables. CGM may have feasibility advantages and be useful in some settings. However, our data suggest there is insufficient evidence to replace OGTT measures with CGM in the context of clinical trials., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
32. Testing a new platform to screen disease-modifying therapy in type 1 diabetes.
- Author
-
Lord SM, Bahnson HT, Greenbaum CJ, Liljenquist DR, Virostko J, and Speake C
- Subjects
- Humans, C-Peptide, Liraglutide, Pilot Projects, Pandemics, Insulin therapeutic use, Verapamil, Diabetes Mellitus, Type 1 drug therapy
- Abstract
Studies of new therapies to preserve insulin secretion in early type 1 diabetes require several years to recruit eligible subjects and to see a treatment effect; thus, there is interest in alternative study designs to speed this process. Most people with longstanding type 1 diabetes no longer secrete insulin. However, studies from pancreata of those with longstanding T1D show that beta cells staining for insulin can persist for decades after diagnosis, and this is paralleled in work showing proinsulin secretion in individuals with longstanding disease; collectively this suggests that there is a reserve of alive but "sleeping" beta cells. Here, we designed a novel clinical trial platform to test whether a short course of therapy with an agent known to have effects in type 1 diabetes with residual endogenous insulin could transiently induce insulin secretion in those who no longer produce insulin. A therapy that transiently "wakes up" sleeping beta cells might be tested next in a fully powered trial in those with endogenous insulin secretion. In this three-arm non-randomized pilot study, we tested three therapies known to impact disease: two beta-cell supportive agents, liraglutide and verapamil, and an immunomodulatory agent, golimumab. The golimumab treated arm was not fully enrolled due to uncertainties about immunotherapy during the COVID-19 pandemic. Participants had mixed-meal tolerance test (MMTT)-stimulated C-peptide below the quantitation limit (<0.02 ng/mL) at enrollment and received 8 to 12 weeks of therapy. At the completion of therapy, none of the individuals achieved the primary outcome of MMTT-stimulated C-peptide ≥ 0.02 ng/mL. An exploratory outcome of the verapamil arm was MRI-assessed pancreas size, diffusion, and longitudinal relaxation time, which showed repeatability of these measures but no treatment effect. The liraglutide and golimumab arms were registered on clinicaltrials.gov under accession number NCT03632759 and the verapamil arm under accession number NCT05847413. Trail registration: Protocols are registered in ClinicalTrials.gov under accession numbers NCT03632759 and NCT05847413., Competing Interests: We have read the journal’s policy and the authors of this manuscript have the following competing interests: CS participates on an advisory board for Vertex Pharmaceuticals. This does not alter our adherence to PLOS ONE policies on sharing data and materials., (Copyright: © 2023 Lord et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
33. A standardized metric to enhance clinical trial design and outcome interpretation in type 1 diabetes.
- Author
-
Ylescupidez A, Bahnson HT, O'Rourke C, Lord S, Speake C, and Greenbaum CJ
- Subjects
- Humans, C-Peptide therapeutic use, Clinical Trials as Topic, Insulin Secretion, Precision Medicine, Diabetes Mellitus, Type 1 drug therapy
- 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., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
34. Islet-autoreactive CD4+ T cells are linked with response to alefacept in type 1 diabetes.
- Author
-
Balmas E, Chen J, Hu AK, DeBerg HA, Rosasco MG, Gersuk VH, Serti E, Speake C, Greenbaum CJ, Nepom GT, Linsley PS, and Cerosaletti K
- Subjects
- Humans, CD4-Positive T-Lymphocytes metabolism, Alefacept therapeutic use, C-Peptide, Leukocytes, Mononuclear metabolism, Biomarkers, Receptors, Antigen, T-Cell therapeutic use, Diabetes Mellitus, Type 1 drug therapy, Diabetes Mellitus, Type 1 metabolism
- Abstract
Variation in the preservation of β cell function in clinical trials in type 1 diabetes (T1D) has emphasized the need to define biomarkers to predict treatment response. The T1DAL trial targeted T cells with alefacept (LFA-3-Ig) and demonstrated C-peptide preservation in approximately 30% of new-onset T1D individuals. We analyzed islet antigen-reactive (IAR) CD4+ T cells in PBMC samples collected prior to treatment from alefacept- and placebo-treated individuals using flow cytometry and single-cell RNA sequencing. IAR CD4+ T cells at baseline had heterogeneous phenotypes. Transcript profiles formed phenotypic clusters of cells along a trajectory based on increasing maturation and activation, and T cell receptor (TCR) chains showed clonal expansion. Notably, the frequency of IAR CD4+ T cells with a memory phenotype and a unique transcript profile (cluster 3) were inversely correlated with C-peptide preservation in alefacept-treated, but not placebo-treated, individuals. Cluster 3 cells had a proinflammatory phenotype characterized by expression of the transcription factor BHLHE40 and the cytokines GM-CSF and TNF-α, and shared TCR chains with effector memory-like clusters. Our results suggest IAR CD4+ T cells as a potential baseline biomarker of response to therapies targeting the CD2 pathway and warrant investigation for other T cell-related therapies.
- Published
- 2023
- Full Text
- View/download PDF
35. Shifts in isoform usage underlie transcriptional differences in regulatory T cells in type 1 diabetes.
- Author
-
Newman JRB, Long SA, Speake C, Greenbaum CJ, Cerosaletti K, Rich SS, Onengut-Gumuscu S, McIntyre LM, Buckner JH, and Concannon P
- Subjects
- Humans, Genome-Wide Association Study, Protein Isoforms genetics, Alternative Splicing, T-Lymphocytes, Regulatory, Diabetes Mellitus, Type 1 genetics
- Abstract
Genome-wide association studies have identified numerous loci with allelic associations to Type 1 Diabetes (T1D) risk. Most disease-associated variants are enriched in regulatory sequences active in lymphoid cell types, suggesting that lymphocyte gene expression is altered in T1D. Here we assay gene expression between T1D cases and healthy controls in two autoimmunity-relevant lymphocyte cell types, memory CD4
+ /CD25+ regulatory T cells (Treg) and memory CD4+ /CD25- T cells, using a splicing event-based approach to characterize tissue-specific transcriptomes. Limited differences in isoform usage between T1D cases and controls are observed in memory CD4+ /CD25- T-cells. In Tregs, 402 genes demonstrate differences in isoform usage between cases and controls, particularly RNA recognition and splicing factor genes. Many of these genes are regulated by the variable inclusion of exons that can trigger nonsense mediated decay. Our results suggest that dysregulation of gene expression, through shifts in alternative splicing in Tregs, contributes to T1D pathophysiology., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
36. Approval of teplizumab: implications for patients.
- Author
-
Speake C and Greenbaum CJ
- Subjects
- Humans, Hypoglycemic Agents, Antibodies, Monoclonal, Humanized therapeutic use, Diabetes Mellitus, Type 1
- Published
- 2023
- Full Text
- View/download PDF
37. Abatacept for Delay of Type 1 Diabetes Progression in Stage 1 Relatives at Risk: A Randomized, Double-Masked, Controlled Trial.
- Author
-
Russell WE, Bundy BN, Anderson MS, Cooney LA, Gitelman SE, Goland RS, Gottlieb PA, Greenbaum CJ, Haller MJ, Krischer JP, Libman IM, Linsley PS, Long SA, Lord SM, Moore DJ, Moore WV, Moran AM, Muir AB, Raskin P, Skyler JS, Wentworth JM, Wherrett DK, Wilson DM, Ziegler AG, and Herold KC
- Subjects
- Humans, Abatacept therapeutic use, Abatacept pharmacology, Immunosuppressive Agents, T-Lymphocytes, Regulatory, Glucose therapeutic use, Diabetes Mellitus, Type 1 drug therapy
- Abstract
Objective: Previous studies showed that inhibiting lymphocyte costimulation reduces declining β-cell function in individuals newly diagnosed with type 1 diabetes. We tested whether abatacept would delay or prevent progression of type 1 diabetes from normal glucose tolerance (NGT) to abnormal glucose tolerance (AGT) or to diabetes and the effects of treatment on immune and metabolic responses., Research Design and Methods: We conducted a phase 2, randomized, placebo-controlled, double-masked trial of abatacept in antibody-positive participants with NGT who received monthly abatacept/placebo infusions for 12 months. The end point was AGT or diabetes, assessed by oral glucose tolerance tests., Results: A total of 101 participants received abatacept and 111 placebo. Of these, 81 (35 abatacept and 46 placebo) met the end point of AGT or type 1 diabetes diagnosis (hazard ratio 0.702; 95% CI 0.452, 1.09; P = 0.11) The C-peptide responses to oral glucose tolerance tests were higher in the abatacept arm (P < 0.03). Abatacept reduced the frequency of inducible T-cell costimulatory (ICOS)+ PD1+ T-follicular helper (Tfh) cells during treatment (P < 0.0001), increased naive CD4+ T cells, and also reduced the frequency of CD4+ regulatory T cells (Tregs) from the baseline (P = 0.0067). Twelve months after treatment, the frequency of ICOS+ Tfh, naive CD4+ T cells, and Tregs returned to baseline., Conclusions: Although abatacept treatment for 1 year did not significantly delay progression to glucose intolerance in at-risk individuals, it impacted immune cell subsets and preserved insulin secretion, suggesting that costimulation blockade may modify progression of type 1 diabetes., (© 2023 by the American Diabetes Association.)
- Published
- 2023
- Full Text
- View/download PDF
38. CGM Metrics Identify Dysglycemic States in Participants From the TrialNet Pathway to Prevention Study.
- Author
-
Wilson DM, Pietropaolo SL, Acevedo-Calado M, Huang S, Anyaiwe D, Scheinker D, Steck AK, Vasudevan MM, McKay SV, Sherr JL, Herold KC, Dunne JL, Greenbaum CJ, Lord SM, Haller MJ, Schatz DA, Atkinson MA, Nelson PW, and Pietropaolo M
- Subjects
- Humans, Female, Adolescent, Male, Blood Glucose metabolism, Blood Glucose Self-Monitoring, Glucose therapeutic use, Autoantibodies, Diabetes Mellitus, Type 1 drug therapy
- Abstract
Objective: Continuous glucose monitoring (CGM) parameters may identify individuals at risk for progression to overt type 1 diabetes. We aimed to determine whether CGM metrics provide additional insights into progression to clinical stage 3 type 1 diabetes., Research Design and Methods: One hundred five relatives of individuals in type 1 diabetes probands (median age 16.8 years; 89% non-Hispanic White; 43.8% female) from the TrialNet Pathway to Prevention study underwent 7-day CGM assessments and oral glucose tolerance tests (OGTTs) at 6-month intervals. The baseline data are reported here. Three groups were evaluated: individuals with 1) stage 2 type 1 diabetes (n = 42) with two or more diabetes-related autoantibodies and abnormal OGTT; 2) stage 1 type 1 diabetes (n = 53) with two or more diabetes-related autoantibodies and normal OGTT; and 3) negative test for all diabetes-related autoantibodies and normal OGTT (n = 10)., Results: Multiple CGM metrics were associated with progression to stage 3 type 1 diabetes. Specifically, spending ≥5% time with glucose levels ≥140 mg/dL (P = 0.01), ≥8% time with glucose levels ≥140 mg/dL (P = 0.02), ≥5% time with glucose levels ≥160 mg/dL (P = 0.0001), and ≥8% time with glucose levels ≥160 mg/dL (P = 0.02) were all associated with progression to stage 3 disease. Stage 2 participants and those who progressed to stage 3 also exhibited higher mean daytime glucose values; spent more time with glucose values over 120, 140, and 160 mg/dL; and had greater variability., Conclusions: CGM could aid in the identification of individuals, including those with a normal OGTT, who are likely to rapidly progress to stage 3 type 1 diabetes., (© 2023 by the American Diabetes Association.)
- Published
- 2023
- Full Text
- View/download PDF
39. Risk Modeling to Reduce Monitoring of an Autoantibody-Positive Population to Prevent DKA at Type 1 Diabetes Diagnosis.
- Author
-
O'Rourke C, Ylescupidez A, Bahnson HT, Bender C, Speake C, Lord S, and Greenbaum CJ
- Subjects
- Adult, Child, Humans, Autoantibodies, Cross-Sectional Studies, Longitudinal Studies, Diabetes Mellitus, Type 1 diagnosis, Diabetes Mellitus, Type 1 epidemiology, Diabetes Mellitus, Type 1 complications, Diabetic Ketoacidosis diagnosis, Diabetic Ketoacidosis epidemiology, Diabetic Ketoacidosis etiology
- Abstract
Context: The presence of islet autoimmunity identifies individuals likely to progress to clinical type 1 diabetes (T1D). In clinical research studies, autoantibody screening followed by regular metabolic monitoring every 6 months reduces incidence of diabetic ketoacidosis (DKA) at diagnosis., Objective: We hypothesized that DKA reduction can be achieved on a population basis with a reduced frequency of metabolic monitoring visits. We reasoned that prolonged time between the development of T1D and the time of clinical diagnosis ("undiagnosed time") would more commonly result in DKA and thus that limiting undiagnosed time would decrease DKA., Methods: An analysis was conducted of data from TrialNet's Pathway to Prevention (PTP), a cross-sectional longitudinal study that identifies and follows at-risk relatives of people with T1D. PTP is a population-based study enrolling across multiple countries. A total of 6193 autoantibody (AAB)-positive individuals participated in PTP from March 2004 to April 2019. We developed models of progression to clinical diagnosis for pediatric and adult populations with single or multiple AAB, and summarized results using estimated hazard rate. An optimal monitoring visit schedule was determined for each model to achieve a minimum average level of undiagnosed time for each population., Results: Halving the number of monitoring visits usually conducted in research studies is likely to substantially lower the population incidence of DKA at diagnosis of T1D., Conclusion: Our study has clinical implications for the metabolic monitoring of at-risk individuals. Fewer monitoring visits would reduce the clinical burden, suggesting a path toward transitioning monitoring beyond the research setting., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
40. Characterizing T cell responses to enzymatically modified beta cell neo-epitopes.
- Author
-
Nguyen H, Arribas-Layton D, Chow IT, Speake C, Kwok WW, Hessner MJ, Greenbaum CJ, and James EA
- Subjects
- Humans, Autoantibodies, Epitopes, Interferon-gamma, Interleukin-4, Peptides, Diabetes Mellitus, Type 1, T-Lymphocytes immunology
- Abstract
Introduction: Previous studies verify the formation of enzymatically post-translationally modified (PTM) self-peptides and their preferred recognition by T cells in subjects with type 1 diabetes (T1D). However, questions remain about the relative prevalence of T cells that recognize PTM self-peptides derived from different antigens, their functional phenotypes, and whether their presence correlates with a specific disease endotype., Methods: To address this question, we identified a cohort of subjects with T1D who had diverse levels of residual beta cell function. Using previously developed HLA class II tetramer reagents, we enumerated T cells that recognize PTM GAD epitopes in the context of DRB1*04:01 or PTM IA2 epitopes in the context of DQB1*03:02 (DQ8)., Results: Consistent with prior studies, we observed higher overall frequencies and a greater proportion of memory T cells in subjects with T1D than in HLA matched controls. There were significantly higher numbers of GAD specific T cells than IA2 specific T cells in subjects with T1D. T cells specific for both groups of epitopes could be expanded from the peripheral blood of subjects with established T1D and at-risk subjects. Expanded neo-epitope specific T cells primarily produced interferon gamma in both groups, but a greater proportion of T cells were interferon gamma positive in subjects with T1D, including some poly-functional cells that also produced IL-4. Based on direct surface phenotyping, neo-epitope specific T cells exhibited diverse combinations of chemokine receptors. However, the largest proportion had markers associated with a Th1-like phenotype. Notably, DQ8 restricted responses to PTM IA2 were over-represented in subjects with lower residual beta cell function. Neo-epitope specific T cells were present in at-risk subjects, and those with multiple autoantibodies have higher interferon gamma to IL-4 ratios than those with single autoantibodies, suggesting a shift in polarization during progression., Discussion: These results reinforce the relevance of PTM neo-epitopes in human disease and suggest that distinct responses to neo-antigens promote a more rapid decline in beta cell function., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Nguyen, Arribas-Layton, Chow, Speake, Kwok, Hessner, Greenbaum and James.)
- Published
- 2023
- Full Text
- View/download PDF
41. IL-6-targeted therapies to block the cytokine or its receptor drive distinct alterations in T cell function.
- Author
-
Speake C, Habib T, Lambert K, Hundhausen C, Lord S, Dufort MJ, Skinner SO, Hu A, Kinsman M, Jones BE, Maerz MD, Tatum M, Hocking AM, Nepom GT, Greenbaum CJ, and Buckner JH
- Subjects
- Humans, Signal Transduction, Phosphorylation, Cytokines pharmacology, Receptors, Antigen, T-Cell
- Abstract
Therapeutics that inhibit IL-6 at different points in its signaling pathway are in clinical use, yet whether the immunological effects of these interventions differ based on their molecular target is unknown. We performed short-term interventions in individuals with type 1 diabetes using anti-IL-6 (siltuximab) or anti-IL-6 receptor (IL-6R; tocilizumab) therapies and investigated the impact of this in vivo blockade on T cell fate and function. Immune outcomes were influenced by the target of the therapeutic intervention (IL-6 versus IL-6R) and by peak drug concentration. Tocilizumab reduced ICOS expression on T follicular helper cell populations and T cell receptor-driven (TCR-driven) STAT3 phosphorylation. Siltuximab reversed resistance to Treg-mediated suppression and increased TCR-driven phosphorylated STAT3 and production of IL-10, IL-21, and IL-27 by T effectors. Together, these findings indicate that the context of IL-6 blockade in vivo drives distinct T cell-intrinsic changes that may influence therapeutic outcomes.
- Published
- 2022
- Full Text
- View/download PDF
42. IL-6-Driven pSTAT1 Response Is Linked to T Cell Features Implicated in Early Immune Dysregulation.
- Author
-
Lambert K, Diggins KE, Jones BE, Hundhausen C, Maerz MD, Hocking AM, Sanda S, Greenbaum CJ, Linsley PS, Cerosaletti K, and Buckner JH
- Subjects
- Apoptosis, Cytokines, Humans, Immune System Diseases etiology, Immune System Diseases pathology, Interleukin-6 metabolism, Interleukin-6 pharmacology, STAT1 Transcription Factor metabolism, Signal Transduction, T-Lymphocytes metabolism, T-Lymphocytes pathology
- Abstract
Elevated levels and enhanced sensing of the pro-inflammatory cytokine interleukin-6 (IL-6) are key features of many autoimmune and inflammatory diseases. To better understand how IL-6 signaling may influence human T cell fate, we investigated the relationships between levels of components of the IL-6R complex, pSTAT responses, and transcriptomic and translational changes in CD4
+ and CD8+ T cell subsets from healthy individuals after exposure to IL-6. Our findings highlight the striking heterogeneity in mbIL-6R and gp130 expression and IL-6-driven pSTAT1/3 responses across T cell subsets. Increased mbIL-6R expression correlated with enhanced signaling via pSTAT1 with less impact on pSTAT3, most strikingly in CD4+ naïve T cells. Additionally, IL-6 rapidly induced expression of transcription factors and surface receptors expressed by T follicular helper cells and altered expression of markers of apoptosis. Importantly, many of the features associated with the level of mbIL-6R expression on T cells were recapitulated both in the setting of tocilizumab therapy and when comparing donor CD4+ T cells harboring the genetic variant, IL6R Asp358Ala (rs2228145), known to alter mbIL-6R expression on T cells. Collectively, these findings should be taken into account as we consider the role of IL-6 in disease pathogenesis and translating IL-6 biology into effective therapies for T cell-mediated autoimmune disease., Competing Interests: JB is a Scientific Co-Founder and Scientific Advisory Board member of GentiBio, a consultant for Bristol-Myers Squibb and Hotspot Therapeutics, and has past and current research projects sponsored by Amgen, Bristol-Myers Squib, Janssen, Novo Nordisk, and Pfizer. She is a member of the Type 1 Diabetes TrialNet Study Group, a partner of the Allen Institute for Immunology, and a member of the Scientific Advisory Boards for the La Jolla Institute for Allergy and Immunology and BMS Immunology. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Lambert, Diggins, Jones, Hundhausen, Maerz, Hocking, Sanda, Greenbaum, Linsley, Cerosaletti and Buckner.)- Published
- 2022
- Full Text
- View/download PDF
43. Rationale and Design for the Diabetes RElated to Acute Pancreatitis and Its Mechanisms Study: A Prospective Cohort Study From the Type 1 Diabetes in Acute Pancreatitis Consortium.
- Author
-
Hart PA, Papachristou GI, Park WG, Dyer AM, Chinchilli VM, Afghani E, Akshintala VS, Andersen DK, Buxbaum JL, Conwell DL, Dungan KM, Easler JJ, Fogel EL, Greenbaum CJ, Kalyani RR, Korc M, Kozarek R, Laughlin MR, Lee PJ, Maranki JL, Pandol SJ, Phillips AE, Serrano J, Singh VK, Speake C, Tirkes T, Toledo FGS, Trikudanathan G, Vege SS, Wang M, Yazici C, Zaheer A, Forsmark CE, Bellin MD, and Yadav D
- Subjects
- Acute Disease, Humans, Incidence, Prospective Studies, Diabetes Mellitus, Type 1 complications, Diabetes Mellitus, Type 1 diagnosis, Diabetes Mellitus, Type 1 epidemiology, Pancreatitis complications, Pancreatitis epidemiology
- Abstract
Abstract: Acute pancreatitis (AP) is a disease characterized by an acute inflammatory phase followed by a convalescent phase. Diabetes mellitus (DM) was historically felt to be a transient phenomenon related to acute inflammation; however, it is increasingly recognized as an important late and chronic complication. There are several challenges that have prevented precisely determining the incidence rate of DM after AP and understanding the underlying mechanisms. The DREAM (Diabetes RElated to Acute Pancreatitis and its Mechanisms) Study is a prospective cohort study designed to address these and other knowledge gaps to provide the evidence needed to screen for, prevent, and treat DM after AP. In the following article, we summarize literature regarding the epidemiology of DM after AP and provide the rationale and an overview of the DREAM study., Competing Interests: E.A. is on the advisory board for Nestle. M.D.B. receives research support from Viacyte and Dexcom and is on advisory board for Insulet. K.M.D. receives research support from Sanofi, Viacyte, Abbott, and Dexcom; has consulting activities with Eli Lilly, Boehringer Ingelheim, Elsevier; and receives honoraria from UptoDate, Elsevier, Medscape, and Academy for Continued Healthcare Learning. C.E.F. receives research support from AbbVie and has consulting activities with Nestle. G.I.P. receives research support from AbbVie and has consulting activities with Nestle. C.S. is on the advisory board for Vertex Pharmaceuticals. F.G.S.T. receives research support from Dompé Pharmaceuticals and has consulting activities with Sanofi, Eli Lilly, and AstraZeneca. The other authors declare no conflict of interest., (Copyright © 2022 Written work prepared by employees of the Federal Government as part of their official duties is, under the U.S. Copyright Act, a “work of the United States Government” for which copyright protection under Title 17 of the United States Code is not available. As such, copyright does not extend to the contributions of employees of the Federal Government.)
- Published
- 2022
- Full Text
- View/download PDF
44. Assessing the Pathophysiology of Hyperglycemia in the Diabetes RElated to Acute Pancreatitis and Its Mechanisms Study: From the Type 1 Diabetes in Acute Pancreatitis Consortium.
- Author
-
Dungan KM, Hart PA, Andersen DK, Basina M, Chinchilli VM, Danielson KK, Evans-Molina C, Goodarzi MO, Greenbaum CJ, Kalyani RR, Laughlin MR, Pichardo-Lowden A, Pratley RE, Serrano J, Sims EK, Speake C, Yadav D, Bellin MD, and Toledo FGS
- Subjects
- Acute Disease, Blood Glucose, Glucose, Humans, Incretins metabolism, Insulin metabolism, Pancreatic Polypeptide, Diabetes Mellitus, Type 1 complications, Diabetes Mellitus, Type 1 diagnosis, Hyperglycemia complications, Insulin Resistance, Pancreatitis complications, Pancreatitis diagnosis
- Abstract
Objectives: The metabolic abnormalities that lead to diabetes mellitus (DM) after an episode of acute pancreatitis (AP) have not been extensively studied. This article describes the objectives, hypotheses, and methods of mechanistic studies of glucose metabolism that comprise secondary outcomes of the DREAM (Diabetes RElated to Acute pancreatitis and its Mechanisms) Study., Methods: Three months after an index episode of AP, participants without preexisting DM will undergo baseline testing with an oral glucose tolerance test. Participants will be followed longitudinally in three subcohorts with distinct metabolic tests. In the first and largest subcohort, oral glucose tolerance tests will be repeated 12 months after AP and annually to assess changes in β-cell function, insulin secretion, and insulin sensitivity. In the second, mixed meal tolerance tests will be performed at 3 and 12 months, then annually, and following incident DM to assess incretin and pancreatic polypeptide responses. In the third, frequently sampled intravenous glucose tolerance tests will be performed at 3 months and 12 months to assess the first-phase insulin response and more precisely measure β-cell function and insulin sensitivity., Conclusions: The DREAM study will comprehensively assess the metabolic and endocrine changes that precede and lead to the development of DM after AP., Competing Interests: K.M.D. has declared research support from Sanofi, Viacyte, Abbott, and Dexcom; consulting activities with Eli Lilly, Boehringer Ingelheim, Elsevier, and Dexcom; and honoraria from UptoDate, Medscape, Academy for Continued Healthcare Learning, and Cardiometabolic Health Congress. M.D.B. has declared research support from Viacyte and Dexcom and consulting activities with Insulet (advisory board). C.E.-M. has received research support from Lilly, Astellas Pharma, Bristol Myers Squibb, and Nimbus Therapeutics and consulting/advisory fees from Avotres, Inc, DiogenX, Isla Technologies, Provention Bio, Inc, MaiCell Therapeutics, and Dompe. R.E.P. has received grants (directed to his institution) from Hanmi Pharmaceutical Co, Ltd, Janssen, Metavention, Novo Nordisk, Poxel SA, and Sanofi; has received consulting fees (directed to his institution) from AstraZeneca, Corcept Therapeutics Incorporated, Glytec LLC, Hanmi Pharmaceutical Co, Ltd, Janssen, Merck & Co, Inc., Mundipharma, Novo Nordisk, Pfizer, Inc, Sanofi, Scohia Pharma, Inc, and Sun Pharmaceutical Industries; and has received support for attending meetings/travel (directed to his institution or to the travel provider) from AstraZeneca, Glytec LLC, Merck & Co, Inc, Mundipharma, Novo Nordisk, and Pfizer, Inc. E.K.S. has received honoraria from Medscape. C.S. has participated on an advisory board for Vertex Pharmaceuticals. F.G.S.T. has declared research support from Dompé Pharmaceuticals and previous consulting activities with Sanofi, Eli Lilly, and AstraZeneca. The other authors declare no conflict of interest., (Copyright © 2022 Written work prepared by employees of the Federal Government as part of their official duties is, under the U.S. Copyright Act, a “work of the United States Government” for which copyright protection under Title 17 of the United States Code is not available. As such, copyright does not extend to the contributions of employees of the Federal Government.)
- Published
- 2022
- Full Text
- View/download PDF
45. Recruitment and Retention Strategies for the Diabetes RElated to Acute Pancreatitis and Its Mechanisms Study: From the Type 1 Diabetes in Acute Pancreatitis Consortium.
- Author
-
Yazici C, Dyer AM, Conwell DL, Afghani E, Andersen DK, Basina M, Bellin MD, Boone LR, Casu A, Easler JJ, Greenbaum CJ, Hart PA, Jeon CY, Lee PJ, Meier S, Papachristou GI, Raja-Khan NT, Saeed ZI, Serrano J, Yadav D, and Fogel EL
- Subjects
- Acute Disease, Humans, Prospective Studies, Diabetes Mellitus, Type 1 complications, Diabetes Mellitus, Type 1 therapy, Pancreatitis complications, Pancreatitis diagnosis
- Abstract
Abstract: Recruitment and retention of patients with acute pancreatitis (AP) in clinical studies can be challenging. While some obstacles are similar to other clinical conditions, some are unique to AP. Identifying potential barriers early and developing targeted solutions can help optimize recruitment and retention in AP studies. Such pre-emptive and detailed planning can help prospective, longitudinal studies focus on exocrine and endocrine complications of AP in accurately measuring outcomes. This article highlights the challenges in recruitment and retention strategies in AP studies and reviews available resources to create opportunities to address them. We describe the multifaceted approach used by the Recruitment and Retention Committee of the Type 1 Diabetes in Acute Pancreatitis Consortium, which builds upon earlier experiences to develop a recruitment and retention plan for the DREAM (Diabetes RElated to Acute pancreatitis and its Mechanisms) study., Competing Interests: E.A. is part of advisory board for Nestle. M.D.B. receives research support from Viacyte and Dexcom and is part of advisory board for Insulet. AC is part of advisory board for GSK and expert opinion for Guidepoint. P.J.L. is part of advisory board for AbbVie. G.I.P. receives research support from AbbVie, has consulting activities with Olympus and Nestle, and receives equity with Ariel Precision Medicine. The other authors declare no conflict of interest., (Copyright © 2022 Written work prepared by employees of the Federal Government as part of their official duties is, under the U.S. Copyright Act, a “work of the United States Government” for which copyright protection under Title 17 of the United States Code is not available. As such, copyright does not extend to the contributions of employees of the Federal Government.)
- Published
- 2022
- Full Text
- View/download PDF
46. Citrullination of glucokinase is linked to autoimmune diabetes.
- Author
-
Yang ML, Horstman S, Gee R, Guyer P, Lam TT, Kanyo J, Perdigoto AL, Speake C, Greenbaum CJ, Callebaut A, Overbergh L, Kibbey RG, Herold KC, James EA, and Mamula MJ
- Subjects
- Animals, Citrullination, Glucose metabolism, Humans, Inflammation metabolism, Insulin metabolism, Liver metabolism, Mice, Mice, Inbred NOD, Diabetes Mellitus, Type 1 metabolism, Glucokinase genetics
- Abstract
Inflammation, including reactive oxygen species and inflammatory cytokines in tissues amplify various post-translational modifications of self-proteins. A number of post-translational modifications have been identified as autoimmune biomarkers in the initiation and progression of Type 1 diabetes. Here we show the citrullination of pancreatic glucokinase as a result of inflammation, triggering autoimmunity and affecting glucokinase biological functions. Glucokinase is expressed in hepatocytes to regulate glycogen synthesis, and in pancreatic beta cells as a glucose sensor to initiate glycolysis and insulin signaling. We identify autoantibodies and autoreactive CD4
+ T cells to glucokinase epitopes in the circulation of Type 1 diabetes patients and NOD mice. Finally, citrullination alters glucokinase biologic activity and suppresses glucose-stimulated insulin secretion. Our study define glucokinase as a Type 1 diabetes biomarker, providing new insights of how inflammation drives post-translational modifications to create both neoautoantigens and affect beta cell metabolism., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
47. Characterising the age-dependent effects of risk factors on type 1 diabetes progression.
- Author
-
So M, O'Rourke C, Ylescupidez A, Bahnson HT, Steck AK, Wentworth JM, Bruggeman BS, Lord S, Greenbaum CJ, and Speake C
- Subjects
- Adolescent, Adult, Autoantibodies, Child, Child, Preschool, Disease Progression, Genetic Predisposition to Disease, HLA-DR3 Antigen, Humans, Infant, Male, Middle Aged, Risk Factors, Young Adult, Diabetes Mellitus, Type 1
- Abstract
Aims/hypothesis: Age is known to be one of the most important stratifiers of disease progression in type 1 diabetes. However, what drives the difference in rate of progression between adults and children is poorly understood. Evidence suggests that many type 1 diabetes disease predictors do not have the same effect across the age spectrum. Without a comprehensive analysis describing the varying risk profiles of predictors over the age continuum, researchers and clinicians are susceptible to inappropriate assessment of risk when examining populations of differing ages. We aimed to systematically assess and characterise how the effect of key type 1 diabetes risk predictors changes with age., Methods: Using longitudinal data from single- and multiple-autoantibody-positive at-risk individuals recruited between the ages of 1 and 45 years in TrialNet's Pathway to Prevention Study, we assessed and visually characterised the age-varying effect of key demographic, immune and metabolic predictors of type 1 diabetes by employing a flexible spline model. Two progression outcomes were defined: participants with single autoantibodies (n=4893) were analysed for progression to multiple autoantibodies or type 1 diabetes, and participants with multiple autoantibodies were analysed (n=3856) for progression to type 1 diabetes., Results: Several predictors exhibited significant age-varying effects on disease progression. Amongst single-autoantibody participants, HLA-DR3 (p=0.007), GAD65 autoantibody positivity (p=0.008), elevated BMI (p=0.007) and HOMA-IR (p=0.002) showed a significant increase in effect on disease progression with increasing age. Insulin autoantibody positivity had a diminishing effect with older age in single-autoantibody-positive participants (p<0.001). Amongst multiple-autoantibody-positive participants, male sex (p=0.002) was associated with an increase in risk for progression, and HLA DR3/4 (p=0.05) showed a decreased effect on disease progression with older age. In both single- and multiple-autoantibody-positive individuals, significant changes in HR with age were seen for multiple measures of islet function. Risk estimation using prediction risk score Index60 was found to be better at a younger age for both single- and multiple-autoantibody-positive individuals (p=0.007 and p<0.001, respectively). No age-varying effect was seen for prediction risk score DPTRS (p=0.861 and p=0.178, respectively). Multivariable analyses suggested that incorporating the age-varying effect of the individual components of these validated risk scores has the potential to enhance the risk estimate., Conclusions/interpretation: Analysing the age-varying effect of disease predictors improves understanding and prediction of type 1 diabetes disease progression, and should be leveraged to refine prediction models and guide mechanistic studies., (© 2022. Crown.)
- Published
- 2022
- Full Text
- View/download PDF
48. Deep immune phenotyping reveals similarities between aging, Down syndrome, and autoimmunity.
- Author
-
Lambert K, Moo KG, Arnett A, Goel G, Hu A, Flynn KJ, Speake C, Wiedeman AE, Gersuk VH, Linsley PS, Greenbaum CJ, Long SA, Partridge R, Buckner JH, and Khor B
- Subjects
- Aging, Autoimmunity genetics, CD8-Positive T-Lymphocytes, Humans, Immunophenotyping, Autoimmune Diseases, Diabetes Mellitus, Type 1, Down Syndrome genetics
- Abstract
Individuals with Down syndrome show cellular and clinical features of dysregulated aging of the immune system, including a shift from naïve to memory T cells and increased incidence of autoimmunity. However, a quantitative understanding of how various immune compartments change with age in Down syndrome remains lacking. Here, we performed deep immunophenotyping of a cohort of individuals with Down syndrome across the life span, selecting for autoimmunity-free individuals. We simultaneously interrogated age- and sex-matched healthy controls and people with type 1 diabetes as a representative autoimmune disease. We built an analytical software, IMPACD (Iterative Machine-assisted Permutational Analysis of Cytometry Data), that enabled us to rapidly identify many features of immune dysregulation in Down syndrome shared with other autoimmune diseases. We found quantitative and qualitative dysregulation of naïve CD4
+ and CD8+ T cells in individuals with Down syndrome and identified interleukin-6 as a candidate driver of some of these changes, thus extending the consideration of immunopathologic cytokines in Down syndrome beyond interferons. We used immune cellular composition to generate three linear models of aging (immune clocks) trained on control participants. All three immune clocks demonstrated advanced immune aging in individuals with Down syndrome. One of these clocks, informed by Down syndrome–relevant biology, also showed advanced immune aging in individuals with type 1 diabetes. Orthologous RNA sequencing–derived immune clocks also demonstrated advanced immune aging in individuals with Down syndrome. Together, our findings demonstrate an approach to studying immune aging in Down syndrome that may have implications in other autoimmune diseases.- Published
- 2022
- Full Text
- View/download PDF
49. Autoreactive T cell receptors with shared germline-like α chains in type 1 diabetes.
- Author
-
Linsley PS, Barahmand-Pour-Whitman F, Balmas E, DeBerg HA, Flynn KJ, Hu AK, Rosasco MG, Chen J, O'Rourke C, Serti E, Gersuk VH, Motwani K, Seay HR, Brusko TM, Kwok WW, Speake C, Greenbaum CJ, Nepom GT, and Cerosaletti K
- Subjects
- Adolescent, Adult, Female, Humans, Male, Young Adult, Diabetes Mellitus, Type 1 genetics, Germ Cells metabolism, Immunoglobulin alpha-Chains metabolism, Receptors, Antigen, T-Cell metabolism
- Abstract
Human islet antigen reactive CD4+ memory T cells (IAR T cells) play a key role in the pathogenesis of autoimmune type 1 diabetes (T1D). Using single-cell RNA sequencing (scRNA-Seq) to identify T cell receptors (TCRs) in IAR T cells, we have identified a class of TCRs that share TCRα chains between individuals ("public" chains). We isolated IAR T cells from blood of healthy, new-onset T1D and established T1D donors using multiplexed CD154 enrichment and identified paired TCRαβ sequences from 2767 individual cells. More than a quarter of cells shared TCR junctions between 2 or more cells ("expanded"), and 29/47 (~62%) of expanded TCRs tested showed specificity for islet antigen epitopes. Public TCRs sharing TCRα junctions were most prominent in new-onset T1D. Public TCR sequences were more germline like than expanded unique, or "private," TCRs, and had shorter junction sequences, suggestive of fewer random nucleotide insertions. Public TCRα junctions were often paired with mismatched TCRβ junctions in TCRs; remarkably, a subset of these TCRs exhibited cross-reactivity toward distinct islet antigen peptides. Our findings demonstrate a prevalent population of IAR T cells with diverse specificities determined by TCRs with restricted TCRα junctions and germline-constrained antigen recognition properties. Since these "innate-like" TCRs differ from previously described immunodominant TCRβ chains in autoimmunity, they have implications for fundamental studies of disease mechanisms. Self-reactive restricted TCRα chains and their associated epitopes should be considered in fundamental and translational investigations of TCRs in T1D.
- Published
- 2021
- Full Text
- View/download PDF
50. IL-6 receptor blockade does not slow β cell loss in new-onset type 1 diabetes.
- Author
-
Greenbaum CJ, Serti E, Lambert K, Weiner LJ, Kanaparthi S, Lord S, Gitelman SE, Wilson DM, Gaglia JL, Griffin KJ, Russell WE, Raskin P, Moran A, Willi SM, Tsalikian E, DiMeglio LA, Herold KC, Moore WV, Goland R, Harris M, Craig ME, Schatz DA, Baidal DA, Rodriguez H, Utzschneider KM, Nel HJ, Soppe CL, Boyle KD, Cerosaletti K, Keyes-Elstein L, Long SA, Thomas R, McNamara JG, Buckner JH, and Sanda S
- Subjects
- Adolescent, Child, Diabetes Mellitus, Type 1 pathology, Double-Blind Method, Female, Humans, Male, B-Lymphocyte Subsets metabolism, Diabetes Mellitus, Type 1 genetics, Receptors, Interleukin-6 antagonists & inhibitors
- Abstract
BackgroundIL-6 receptor (IL-6R) signaling drives development of T cell populations important to type 1 diabetes pathogenesis. We evaluated whether blockade of IL-6R with monoclonal antibody tocilizumab would slow loss of residual β cell function in newly diagnosed type 1 diabetes patients.MethodsWe conducted a multicenter, randomized, placebo-controlled, double-blind trial with tocilizumab in new-onset type 1 diabetes. Participants were screened within 100 days of diagnosis. Eligible participants were randomized 2:1 to receive 7 monthly doses of tocilizumab or placebo. The primary outcome was the change from screening in the mean AUC of C-peptide collected during the first 2 hours of a mixed meal tolerance test at week 52 in pediatric participants (ages 6-17 years).ResultsThere was no statistical difference in the primary outcome between tocilizumab and placebo. Immunophenotyping showed reductions in downstream signaling of the IL-6R in T cells but no changes in CD4 memory subsets, Th17 cells, Tregs, or CD4+ T effector cell resistance to Treg suppression. A DC subset decreased during therapy but regressed to baseline once therapy stopped. Tocilizumab was well tolerated.ConclusionTocilizumab reduced T cell IL-6R signaling but did not modulate CD4+ T cell phenotypes or slow loss of residual β cell function in newly diagnosed individuals with type 1 diabetes.Trial RegistrationClinicalTrials.gov NCT02293837.FundingNIH National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and National Institute of Allergy and Infectious Diseases (NIAID) UM1AI109565, UL1TR000004 from NIH/National Center for Research Resources (NCRR) Clinical and Translational Science Award (CTSA), NIH/NIDDK P30DK036836, NIH/NIDDK U01DK103266, NIH/NIDDK U01DK103266, 1UL1TR000064 from NIH/NCRR CTSA, NIH/National Center for Advancing Translational Sciences (NCATS) UL1TR001878, UL1TR002537 from NIH/CTSA; National Health and Medical Research Council Practitioner Fellowship (APP1136735), NIH/NIDDK U01-DK085476, NIH/CTSA UL1-TR002494, Indiana Clinical and Translational Science Institute Award UL1TR002529, Vanderbilt Institute for Clinical and Translational Research UL1TR000445. NIH/NCATS UL1TR003142, NIH/CTSA program UL1-TR002494, Veteran Affairs Administration, and 1R01AI132774.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.