search
Back to results

Monitoring and Managing Glucose Levels in People With Pancreatic Cancer (PEGASUS)

Primary Purpose

Pancreatic Cancer, PDAC - Pancreatic Ductal Adenocarcinoma, Hyperglycemia

Status
Not yet recruiting
Phase
Not Applicable
Locations
Study Type
Interventional
Intervention
Endocrinologist-directed target blood glucose level 4-10 mmol/L using data from a continuous glucose monitor (CGM)
Standard Care
Sponsored by
British Columbia Cancer Agency
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Pancreatic Cancer focused on measuring FOLFIRINOX, Continuous Glucose Monitor, Feasibility, Glycemic management, Endocrinologist, Intensive glucose intervention, Pilot, Glucose control

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

  • Histological/cytological diagnosis of pancreatic ductal adenocarcinoma (PDAC).
  • Planned to undergo first-line systemic therapy with FOLFIRINOX.
  • Age greater than or equal to 18 years.
  • Eastern Cooperative Oncology Group (ECOG) performance status 0-1.
  • Adequate bone marrow and organ function as defined by the following laboratory values:

    1. Absolute neutrophil count (ANC) greater than or equal to 1.5 x 10^9/L.
    2. Platelet count greater than or equal to 75 x 10^9/L.
    3. Hemoglobin greater than or equal to 9.0 g/dL.
    4. Estimated glomerular filtration rate (GFR) by Cockroft-Gault equation OR 24 hour urine collection greater than or equal to 40 ml/min.
    5. Creatinine clearance greater than or equal to 40 mL/min using Cockcroft-Gault formula.
    6. Potassium within normal limits, or corrected with supplements.
    7. International normalized ratio (INR) less than or equal to 1.5.
    8. Total serum bilirubin less than or equal to 2 x upper limit of normal (ULN) (any elevated bilirubin should be asymptomatic at enrollment) except for participants with documented Gilbert's syndrome who may only be included if the total bilirubin less than or equal to 3 x ULN or direct bilirubin less than or equal to 1.5 x ULN).
    9. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) less than or equal to 2.5 x ULN (or less than or equal to 5 x ULN if liver metastases are present).
  • Able to understand and voluntarily sign the informed consent form.
  • Able to comply with the study visit schedule and other protocol requirements.
  • Able to swallow oral medications and has no contraindications to subcutaneous insulin injections.
  • Measurable or evaluable disease by Response Evaluation Criteria in Solid Tumours (RECIST) 1.1 at baseline.
  • Life expectancy of more than 90 days as judged by the study doctor.

Exclusion Criteria:

  • Absence of distant or lymph node metastases. Participants with borderline resectable or locally advanced PDAC are not eligible.
  • Received prior systemic therapy (chemotherapy or any other anti-cancer agent) for treatment of metastatic PDAC. Participants who received adjuvant chemotherapy after surgical resection of early stage disease are eligible.
  • Currently receiving anti-cancer therapy (chemotherapy or any other anti-cancer agent).
  • Not fit for combination chemotherapy as judged by the study doctor.
  • Presence of brain metastases.
  • Known diagnosis of type I diabetes where strict glucose control and close Endocrinology follow-up is already indicated.
  • Known diagnosis of type II diabetes and already followed by Endocrinologist.
  • Female participants with a positive pregnancy test.
  • Participants who are not safe to include in the study as judged by the study doctor for any medical or non-medical reason.
  • Unable to comply with study assessments and follow-up.

Sites / Locations

    Arms of the Study

    Arm 1

    Arm 2

    Arm Type

    Experimental

    Other

    Arm Label

    Intensive Glucose Intervention

    Standard Care

    Arm Description

    Participants will receive standard anti-hyperglycemic treatment as guided by an endocrinologist using a combination of data from a continuous glucose monitor (CGM) and standard blood work drawn prior to each cycle of chemotherapy. Treatment will aim to maintain glucose levels between 4 and 10 mmol/L. Participants will have real-time access to their glucose data via the CGM.

    Participants will receive standard anti-hyperglycemic treatment only if blood glucose level is above 15 mmol/L as measured from standard blood work drawn prior to each cycle of chemotherapy. Participants will wear a CGM but will not be able to view their glucose data. Participants may be referred to an endocrinologist at the discretion of their medical oncologist.

    Outcomes

    Primary Outcome Measures

    Frequency of glucose levels maintained within range in Arm 1 compared to Arm 2
    The percentage of time each participant's glucose levels in Arm 1 and Arm 2 remained within the 4-10 mmol/L range during the fourth cycle of FOLFIRINOX treatment as measured by a continuous glucose monitor.

    Secondary Outcome Measures

    Overall response rate (ORR) in each study arm, as defined by RECIST 1.1
    The proportion of participants in each study arm who have a complete response (CR) or partial response (PR) to FOLFIRINOX treatment, as defined by RECIST 1.1.
    Progression-free survival (PFS) in each study arm from the initiation of FOLFIRINOX
    The length of time from the first dose of FOLFIRINOX until the date of progressive disease (PD), as defined by RECIST 1.1, for participants in each study arm.
    Overall survival (OS) in each study arm from the initiation of FOLFIRINOX
    The length of time from the initiation of FOLFIRINOX that participants survive in each study arm.

    Full Information

    First Posted
    November 10, 2021
    Last Updated
    August 2, 2023
    Sponsor
    British Columbia Cancer Agency
    Collaborators
    University of British Columbia
    search

    1. Study Identification

    Unique Protocol Identification Number
    NCT05132244
    Brief Title
    Monitoring and Managing Glucose Levels in People With Pancreatic Cancer
    Acronym
    PEGASUS
    Official Title
    Pancreatic Cancer Glucose Assessment and Regulation Study
    Study Type
    Interventional

    2. Study Status

    Record Verification Date
    August 2023
    Overall Recruitment Status
    Not yet recruiting
    Study Start Date
    October 2023 (Anticipated)
    Primary Completion Date
    October 2025 (Anticipated)
    Study Completion Date
    December 2026 (Anticipated)

    3. Sponsor/Collaborators

    Responsible Party, by Official Title
    Sponsor
    Name of the Sponsor
    British Columbia Cancer Agency
    Collaborators
    University of British Columbia

    4. Oversight

    Studies a U.S. FDA-regulated Drug Product
    No
    Studies a U.S. FDA-regulated Device Product
    No
    Data Monitoring Committee
    No

    5. Study Description

    Brief Summary
    This study will investigate whether or not it is feasible to closely monitor and manage glucose levels in people with pancreatic cancer. It will also investigate what impact glucose management may have on pancreatic cancer. This is a pilot study that will use continuous glucose monitors (CGM) to monitor glucose levels in approximately 50 participants with pancreatic cancer. Participants will receive standard chemotherapy with a combination of up to four drugs to treat their pancreatic cancer: oxaliplatin, irinotecan, 5-fluorouracil, and leucovorin (FOLFIRINOX). To treat high glucose levels, participants will be randomly assigned to one of two groups: Group 1 will receive anti-hyperglycemic treatment as guided by an endocrinologist with the aim of maintaining glucose levels between 4 and 10 mmol/L; Group 2 will receive anti-hyperglycemic treatment if their glucose levels are above 15 mmol/L, which is standard care. Participants in both Groups 1 and 2 will receive standard anti-hyperglycemic treatments: metformin, insulin, glucagon-like peptide-1 (GLP-1) receptor agonists, sodium glucose co-transporter (SGLT2) inhibitors, and dipeptidyl peptidase 4 (DPP-4) inhibitors. After 4 cycles of FOLFIRINOX, the CGM will be removed but any anti-hyperglycemic treatments will continue as needed. If participants discontinue treatment with FOLFIRINOX, they will continue to be followed for survival and subsequent anti-cancer therapy and will continue follow-up for glucose-related concerns at the discretion of their endocrinologist and/or medical oncologist.

    6. Conditions and Keywords

    Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
    Pancreatic Cancer, PDAC - Pancreatic Ductal Adenocarcinoma, Hyperglycemia
    Keywords
    FOLFIRINOX, Continuous Glucose Monitor, Feasibility, Glycemic management, Endocrinologist, Intensive glucose intervention, Pilot, Glucose control

    7. Study Design

    Primary Purpose
    Treatment
    Study Phase
    Not Applicable
    Interventional Study Model
    Parallel Assignment
    Masking
    Participant
    Masking Description
    Participants in Group 1 can view their glucose data in real-time from the continuous glucose monitor (CGM) whereas participants in Group 2 cannot. Participants in Groups 1 and 2 will NOT be masked to the anti-cancer or anti-hypertensive treatment they receive.
    Allocation
    Randomized
    Enrollment
    50 (Anticipated)

    8. Arms, Groups, and Interventions

    Arm Title
    Intensive Glucose Intervention
    Arm Type
    Experimental
    Arm Description
    Participants will receive standard anti-hyperglycemic treatment as guided by an endocrinologist using a combination of data from a continuous glucose monitor (CGM) and standard blood work drawn prior to each cycle of chemotherapy. Treatment will aim to maintain glucose levels between 4 and 10 mmol/L. Participants will have real-time access to their glucose data via the CGM.
    Arm Title
    Standard Care
    Arm Type
    Other
    Arm Description
    Participants will receive standard anti-hyperglycemic treatment only if blood glucose level is above 15 mmol/L as measured from standard blood work drawn prior to each cycle of chemotherapy. Participants will wear a CGM but will not be able to view their glucose data. Participants may be referred to an endocrinologist at the discretion of their medical oncologist.
    Intervention Type
    Procedure
    Intervention Name(s)
    Endocrinologist-directed target blood glucose level 4-10 mmol/L using data from a continuous glucose monitor (CGM)
    Intervention Description
    Standard anti-hyperglycemic treatment given as directed by an endocrinologist to maintain blood glucose level within 4-10 mmol/L based on data from a continuous glucose monitor (CGM) and standard blood work drawn prior to each cycle of chemotherapy. Participants will have access to their glucose data from the CGM.
    Intervention Type
    Other
    Intervention Name(s)
    Standard Care
    Intervention Description
    Standard anti-hyperglycemic treatment given only if blood glucose level is greater than 15 mmol/L as measured from standard blood work drawn prior to each cycle of chemotherapy. Participants will wear a continuous glucose monitor (CGM) but will not have access to their glucose data. Participants may be referred to an endocrinologist at the discretion of their medical oncologist.
    Primary Outcome Measure Information:
    Title
    Frequency of glucose levels maintained within range in Arm 1 compared to Arm 2
    Description
    The percentage of time each participant's glucose levels in Arm 1 and Arm 2 remained within the 4-10 mmol/L range during the fourth cycle of FOLFIRINOX treatment as measured by a continuous glucose monitor.
    Time Frame
    From the Cycle 4 FOLFIRINOX treatment date to the Cycle 5 FOLFIRINOX treatment date (each cycle is typically 14 days).
    Secondary Outcome Measure Information:
    Title
    Overall response rate (ORR) in each study arm, as defined by RECIST 1.1
    Description
    The proportion of participants in each study arm who have a complete response (CR) or partial response (PR) to FOLFIRINOX treatment, as defined by RECIST 1.1.
    Time Frame
    From the date of the screening scan (within 28 days of first dose) until the date of confirmed progression, withdrawal, date of death, or end of study, whichever comes first, assessed up to 43 months.
    Title
    Progression-free survival (PFS) in each study arm from the initiation of FOLFIRINOX
    Description
    The length of time from the first dose of FOLFIRINOX until the date of progressive disease (PD), as defined by RECIST 1.1, for participants in each study arm.
    Time Frame
    From the date of first dose of FOLFIRINOX until the date of confirmed progression, withdrawal, date of death, or end of study, whichever comes first, assessed up to 43 months.
    Title
    Overall survival (OS) in each study arm from the initiation of FOLFIRINOX
    Description
    The length of time from the initiation of FOLFIRINOX that participants survive in each study arm.
    Time Frame
    From the date of first dose of FOLFIRINOX until the date of death or end of study, whichever comes first, assessed up to 43 months.
    Other Pre-specified Outcome Measures:
    Title
    Overall response rate (ORR) in each study arm, as defined by RECIST 1.1 and stratified by prognostic and metabolic gene expression subtypes of PDAC
    Description
    The proportion of participants in each study arm stratified by prognostic and metabolic gene expression subtypes of PDAC who have a complete response (CR) or partial response (PR) to FOLFIRINOX treatment, as defined by RECIST 1.1.
    Time Frame
    From the date of the screening scan (within 28 days of first dose) until the date of confirmed progression, withdrawal, date of death, or end of study, whichever comes first, assessed up to 43 months.
    Title
    Progression-free survival (PFS) in each study arm stratified by prognostic and metabolic gene expression subtypes of PDAC from the initiation of FOLFIRINOX
    Description
    The length of time from the first dose of FOLFIRINOX until the date of progressive disease (PD), as defined by RECIST 1.1, for participants in each study arm stratified by prognostic and metabolic gene expression subtypes of PDAC.
    Time Frame
    From the date of first dose of FOLFIRINOX until the date of confirmed progression, withdrawal, date of death, or end of study, whichever comes first, assessed up to 43 months.
    Title
    Overall survival (OS) in each study arm stratified by prognostic and metabolic gene expression subtypes of PDAC from the initiation of FOLFIRINOX
    Description
    The length of time from the initiation of FOLFIRINOX that participants survive in each study arm stratified by prognostic and metabolic gene expression subtypes of PDAC.
    Time Frame
    From the date of first dose of FOLFIRINOX until the date of death or end of study, whichever comes first, assessed up to 43 months.
    Title
    Overall response rate (ORR) in each study arm, as defined by RECIST 1.1 and stratified by clinical features (i.e. type 2 diabetes), pathological profiles, and genomic profiles
    Description
    The proportion of participants in each study arm stratified by clinical features (i.e. type 2 diabetes), pathological profiles, and genomic profiles who have a complete response (CR) or partial response (PR) to FOLFIRINOX treatment, as defined by RECIST 1.1.
    Time Frame
    From the date of the screening scan (within 28 days of first dose) until the date of confirmed progression, withdrawal, date of death, or end of study, whichever comes first, assessed up to 43 months.
    Title
    Progression-free survival (PFS) in each study arm stratified by clinical features (i.e. type 2 diabetes), pathological profiles, and genomic profiles from the initiation of FOLFIRINOX
    Description
    The length of time from the first dose of FOLFIRINOX until the date of progressive disease (PD), as defined by RECIST 1.1, for participants in each study arm stratified by clinical features (i.e. type 2 diabetes), pathological profiles, and genomic profiles.
    Time Frame
    From the date of first dose of FOLFIRINOX until the date of confirmed progression, withdrawal, date of death, or end of study, whichever comes first, assessed up to 43 months.
    Title
    Overall survival (OS) in each study arm stratified by clinical features (i.e. type 2 diabetes), pathological profiles, and genomic profiles from the initiation of FOLFIRINOX
    Description
    The length of time from the initiation of FOLFIRINOX that participants survive in each study arm stratified by clinical features (i.e. type 2 diabetes), pathological profiles, and genomic profiles.
    Time Frame
    From the date of first dose of FOLFIRINOX until the date of death or end of study, whichever comes first, assessed up to 43 months.
    Title
    Amount of insulin, measured in molar, for participants in each study arm from screening until the end of study visit
    Description
    The quantity of insulin, measured in molar from immunoassays, from blood samples collected throughout the study for the participants in each study arm.
    Time Frame
    From the date of screening blood sample collection until the date of the end of study blood sample collection (an average of 6 months).
    Title
    Amount of proinsulin, measured in molar, for participants in each study arm from screening until the end of study visit
    Description
    The quantity of proinsulin, measured in molar from immunoassays, from blood samples collected throughout the study for the participants in each study arm.
    Time Frame
    From the date of the screening blood sample collection until the date of the end of study blood sample collection (an average of 6 months).
    Title
    Amount of C-peptide, measured in molar, for participants in each study arm from screening until the end of study visit
    Description
    The quantity of C-peptide, measured in molar from immunoassays, from blood samples collected throughout the study for the participants in each study arm.
    Time Frame
    From the date of the screening blood sample collection until the date of the end of study blood sample collection (an average of 6 months).
    Title
    Amount of circulating biomarkers, measured in molar, for participants in each study arm from screening until the end of study visit
    Description
    The quantity of circulating biomarkers, measured in molar from immunoassays, from blood samples collected throughout the study for the participants in each study arm.
    Time Frame
    From the date of the screening blood sample collection until the date of the end of study blood sample collection (an average of 6 months).

    10. Eligibility

    Sex
    All
    Minimum Age & Unit of Time
    18 Years
    Accepts Healthy Volunteers
    No
    Eligibility Criteria
    Inclusion Criteria: Histological/cytological diagnosis of pancreatic ductal adenocarcinoma (PDAC). Planned to undergo first-line systemic therapy with FOLFIRINOX. Age greater than or equal to 18 years. Eastern Cooperative Oncology Group (ECOG) performance status 0-1. Adequate bone marrow and organ function as defined by the following laboratory values: Absolute neutrophil count (ANC) greater than or equal to 1.5 x 10^9/L. Platelet count greater than or equal to 75 x 10^9/L. Hemoglobin greater than or equal to 9.0 g/dL. Estimated glomerular filtration rate (GFR) by Cockroft-Gault equation OR 24 hour urine collection greater than or equal to 40 ml/min. Creatinine clearance greater than or equal to 40 mL/min using Cockcroft-Gault formula. Potassium within normal limits, or corrected with supplements. International normalized ratio (INR) less than or equal to 1.5. Total serum bilirubin less than or equal to 2 x upper limit of normal (ULN) (any elevated bilirubin should be asymptomatic at enrollment) except for participants with documented Gilbert's syndrome who may only be included if the total bilirubin less than or equal to 3 x ULN or direct bilirubin less than or equal to 1.5 x ULN). Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) less than or equal to 2.5 x ULN (or less than or equal to 5 x ULN if liver metastases are present). Able to understand and voluntarily sign the informed consent form. Able to comply with the study visit schedule and other protocol requirements. Able to swallow oral medications and has no contraindications to subcutaneous insulin injections. Measurable or evaluable disease by Response Evaluation Criteria in Solid Tumours (RECIST) 1.1 at baseline. Life expectancy of more than 90 days as judged by the study doctor. Exclusion Criteria: Absence of distant or lymph node metastases. Participants with borderline resectable or locally advanced PDAC are not eligible. Received prior systemic therapy (chemotherapy or any other anti-cancer agent) for treatment of metastatic PDAC. Participants who received adjuvant chemotherapy after surgical resection of early stage disease are eligible. Currently receiving anti-cancer therapy (chemotherapy or any other anti-cancer agent). Not fit for combination chemotherapy as judged by the study doctor. Presence of brain metastases. Known diagnosis of type I diabetes where strict glucose control and close Endocrinology follow-up is already indicated. Known diagnosis of type II diabetes and already followed by Endocrinologist. Female participants with a positive pregnancy test. Participants who are not safe to include in the study as judged by the study doctor for any medical or non-medical reason. Unable to comply with study assessments and follow-up.
    Central Contact Person:
    First Name & Middle Initial & Last Name or Official Title & Degree
    Daniel Renouf, MD, MPH
    Phone
    604-877-6000
    Ext
    672445
    Email
    drenouf@bccancer.bc.ca
    Overall Study Officials:
    First Name & Middle Initial & Last Name & Degree
    Daniel Renouf, MD, MPH
    Organizational Affiliation
    BC Cancer
    Official's Role
    Principal Investigator

    12. IPD Sharing Statement

    Citations:
    PubMed Identifier
    19485704
    Citation
    Becker S, Dossus L, Kaaks R. Obesity related hyperinsulinaemia and hyperglycaemia and cancer development. Arch Physiol Biochem. 2009 May;115(2):86-96. doi: 10.1080/13813450902878054.
    Results Reference
    background
    PubMed Identifier
    23943783
    Citation
    Dawson DW, Hertzer K, Moro A, Donald G, Chang HH, Go VL, Pandol SJ, Lugea A, Gukovskaya AS, Li G, Hines OJ, Rozengurt E, Eibl G. High-fat, high-calorie diet promotes early pancreatic neoplasia in the conditional KrasG12D mouse model. Cancer Prev Res (Phila). 2013 Oct;6(10):1064-73. doi: 10.1158/1940-6207.CAPR-13-0065. Epub 2013 Aug 13.
    Results Reference
    background
    PubMed Identifier
    24311953
    Citation
    Harris D, Barts A, Connors J, Dahl M, Elliott T, Kong J, Keane T, Thompson D, Stafford S, Ur E, Sirrs S. Glucocorticoid-induced hyperglycemia is prevalent and unpredictable for patients undergoing cancer therapy: an observational cohort study. Curr Oncol. 2013 Dec;20(6):e532-8. doi: 10.3747/co.20.1499.
    Results Reference
    background
    PubMed Identifier
    18328435
    Citation
    Hart AR, Kennedy H, Harvey I. Pancreatic cancer: a review of the evidence on causation. Clin Gastroenterol Hepatol. 2008 Mar;6(3):275-82. doi: 10.1016/j.cgh.2007.12.041.
    Results Reference
    background
    PubMed Identifier
    17764494
    Citation
    Hassan MM, Bondy ML, Wolff RA, Abbruzzese JL, Vauthey JN, Pisters PW, Evans DB, Khan R, Chou TH, Lenzi R, Jiao L, Li D. Risk factors for pancreatic cancer: case-control study. Am J Gastroenterol. 2007 Dec;102(12):2696-707. doi: 10.1111/j.1572-0241.2007.01510.x. Epub 2007 Aug 31.
    Results Reference
    background
    PubMed Identifier
    18637486
    Citation
    Hjartaker A, Langseth H, Weiderpass E. Obesity and diabetes epidemics: cancer repercussions. Adv Exp Med Biol. 2008;630:72-93. doi: 10.1007/978-0-387-78818-0_6.
    Results Reference
    background
    PubMed Identifier
    15886696
    Citation
    Huxley R, Ansary-Moghaddam A, Berrington de Gonzalez A, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer. 2005 Jun 6;92(11):2076-83. doi: 10.1038/sj.bjc.6602619.
    Results Reference
    background
    PubMed Identifier
    31481506
    Citation
    Karasinska JM, Topham JT, Kalloger SE, Jang GH, Denroche RE, Culibrk L, Williamson LM, Wong HL, Lee MKC, O'Kane GM, Moore RA, Mungall AJ, Moore MJ, Warren C, Metcalfe A, Notta F, Knox JJ, Gallinger S, Laskin J, Marra MA, Jones SJM, Renouf DJ, Schaeffer DF. Altered Gene Expression along the Glycolysis-Cholesterol Synthesis Axis Is Associated with Outcome in Pancreatic Cancer. Clin Cancer Res. 2020 Jan 1;26(1):135-146. doi: 10.1158/1078-0432.CCR-19-1543. Epub 2019 Sep 3.
    Results Reference
    background
    PubMed Identifier
    28647837
    Citation
    Kawada K, Toda K, Sakai Y. Targeting metabolic reprogramming in KRAS-driven cancers. Int J Clin Oncol. 2017 Aug;22(4):651-659. doi: 10.1007/s10147-017-1156-4. Epub 2017 Jun 24.
    Results Reference
    background
    PubMed Identifier
    27518362
    Citation
    Kenner BJ, Chari ST, Maitra A, Srivastava S, Cleeter DF, Go VL, Rothschild LJ, Goldberg AE. Early Detection of Pancreatic Cancer-a Defined Future Using Lessons From Other Cancers: A White Paper. Pancreas. 2016 Sep;45(8):1073-9. doi: 10.1097/MPA.0000000000000701.
    Results Reference
    background
    PubMed Identifier
    27584663
    Citation
    Kleeff J, Costello E, Jackson R, Halloran C, Greenhalf W, Ghaneh P, Lamb RF, Lerch MM, Mayerle J, Palmer D, Cox T, Rawcliffe CL, Strobel O, Buchler MW, Neoptolemos JP. The impact of diabetes mellitus on survival following resection and adjuvant chemotherapy for pancreatic cancer. Br J Cancer. 2016 Sep 27;115(7):887-94. doi: 10.1038/bjc.2016.277. Epub 2016 Sep 1.
    Results Reference
    background
    PubMed Identifier
    18382097
    Citation
    Koorstra JB, Hustinx SR, Offerhaus GJ, Maitra A. Pancreatic carcinogenesis. Pancreatology. 2008;8(2):110-25. doi: 10.1159/000123838. Epub 2008 Apr 1.
    Results Reference
    background
    PubMed Identifier
    23201164
    Citation
    Kopp JL, von Figura G, Mayes E, Liu FF, Dubois CL, Morris JP 4th, Pan FC, Akiyama H, Wright CV, Jensen K, Hebrok M, Sander M. Identification of Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer Cell. 2012 Dec 11;22(6):737-50. doi: 10.1016/j.ccr.2012.10.025. Epub 2012 Nov 29.
    Results Reference
    background
    PubMed Identifier
    27557308
    Citation
    Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K; International Agency for Research on Cancer Handbook Working Group. Body Fatness and Cancer--Viewpoint of the IARC Working Group. N Engl J Med. 2016 Aug 25;375(8):794-8. doi: 10.1056/NEJMsr1606602. No abstract available.
    Results Reference
    background
    PubMed Identifier
    15051286
    Citation
    Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004 Mar 27;363(9414):1049-57. doi: 10.1016/S0140-6736(04)15841-8.
    Results Reference
    background
    PubMed Identifier
    11461162
    Citation
    McCarty MF. Insulin secretion as a determinant of pancreatic cancer risk. Med Hypotheses. 2001 Aug;57(2):146-50. doi: 10.1054/mehy.2001.1316.
    Results Reference
    background
    PubMed Identifier
    18395079
    Citation
    Pannala R, Leirness JB, Bamlet WR, Basu A, Petersen GM, Chari ST. Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology. 2008 Apr;134(4):981-7. doi: 10.1053/j.gastro.2008.01.039. Epub 2008 Jan 18.
    Results Reference
    background
    PubMed Identifier
    35121966
    Citation
    Pleasance E, Titmuss E, Williamson L, Kwan H, Culibrk L, Zhao EY, Dixon K, Fan K, Bowlby R, Jones MR, Shen Y, Grewal JK, Ashkani J, Wee K, Grisdale CJ, Thibodeau ML, Bozoky Z, Pearson H, Majounie E, Vira T, Shenwai R, Mungall KL, Chuah E, Davies A, Warren M, Reisle C, Bonakdar M, Taylor GA, Csizmok V, Chan SK, Zong Z, Bilobram S, Muhammadzadeh A, D'Souza D, Corbett RD, MacMillan D, Carreira M, Choo C, Bleile D, Sadeghi S, Zhang W, Wong T, Cheng D, Brown SD, Holt RA, Moore RA, Mungall AJ, Zhao Y, Nelson J, Fok A, Ma Y, Lee MKC, Lavoie JM, Mendis S, Karasinska JM, Deol B, Fisic A, Schaeffer DF, Yip S, Schrader K, Regier DA, Weymann D, Chia S, Gelmon K, Tinker A, Sun S, Lim H, Renouf DJ, Laskin J, Jones SJM, Marra MA. Pan-cancer analysis of advanced patient tumors reveals interactions between therapy and genomic landscapes. Nat Cancer. 2020 Apr;1(4):452-468. doi: 10.1038/s43018-020-0050-6. Epub 2020 Apr 13.
    Results Reference
    background
    PubMed Identifier
    10997604
    Citation
    Polonsky KS. Dynamics of insulin secretion in obesity and diabetes. Int J Obes Relat Metab Disord. 2000 Jun;24 Suppl 2:S29-31. doi: 10.1038/sj.ijo.0801273.
    Results Reference
    background
    PubMed Identifier
    30215162
    Citation
    Sharma A, Chari ST. Pancreatic Cancer and Diabetes Mellitus. Curr Treat Options Gastroenterol. 2018 Dec;16(4):466-478. doi: 10.1007/s11938-018-0197-8.
    Results Reference
    background
    PubMed Identifier
    29723506
    Citation
    Sharma A, Smyrk TC, Levy MJ, Topazian MA, Chari ST. Fasting Blood Glucose Levels Provide Estimate of Duration and Progression of Pancreatic Cancer Before Diagnosis. Gastroenterology. 2018 Aug;155(2):490-500.e2. doi: 10.1053/j.gastro.2018.04.025. Epub 2018 Apr 30.
    Results Reference
    background
    PubMed Identifier
    27903154
    Citation
    Shlomai G, Neel B, LeRoith D, Gallagher EJ. Type 2 Diabetes Mellitus and Cancer: The Role of Pharmacotherapy. J Clin Oncol. 2016 Dec 10;34(35):4261-4269. doi: 10.1200/JCO.2016.67.4044. Epub 2016 Nov 7.
    Results Reference
    background
    PubMed Identifier
    33433946
    Citation
    Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin. 2021 Jan;71(1):7-33. doi: 10.3322/caac.21654. Epub 2021 Jan 12. Erratum In: CA Cancer J Clin. 2021 Jul;71(4):359.
    Results Reference
    background
    PubMed Identifier
    28700945
    Citation
    Templeman NM, Flibotte S, Chik JHL, Sinha S, Lim GE, Foster LJ, Nislow C, Johnson JD. Reduced Circulating Insulin Enhances Insulin Sensitivity in Old Mice and Extends Lifespan. Cell Rep. 2017 Jul 11;20(2):451-463. doi: 10.1016/j.celrep.2017.06.048.
    Results Reference
    background
    PubMed Identifier
    28390156
    Citation
    Tsujimoto T, Kajio H, Sugiyama T. Association between hyperinsulinemia and increased risk of cancer death in nonobese and obese people: A population-based observational study. Int J Cancer. 2017 Jul 1;141(1):102-111. doi: 10.1002/ijc.30729. Epub 2017 Apr 22.
    Results Reference
    background
    PubMed Identifier
    32580961
    Citation
    Wang F, Qi XM, Wertz R, Mortensen M, Hagen C, Evans J, Sheinin Y, James M, Liu P, Tsai S, Thomas J, Mackinnon A, Dwinell M, Myers CR, Bartrons Bach R, Fu L, Chen G. p38gamma MAPK Is Essential for Aerobic Glycolysis and Pancreatic Tumorigenesis. Cancer Res. 2020 Aug 15;80(16):3251-3264. doi: 10.1158/0008-5472.CAN-19-3281. Epub 2020 Jun 24.
    Results Reference
    background
    PubMed Identifier
    23798995
    Citation
    Wang M, Li J, Lim GE, Johnson JD. Is dynamic autocrine insulin signaling possible? A mathematical model predicts picomolar concentrations of extracellular monomeric insulin within human pancreatic islets. PLoS One. 2013 Jun 14;8(6):e64860. doi: 10.1371/journal.pone.0064860. Print 2013.
    Results Reference
    background
    PubMed Identifier
    31569510
    Citation
    Yan L, Raj P, Yao W, Ying H. Glucose Metabolism in Pancreatic Cancer. Cancers (Basel). 2019 Sep 29;11(10):1460. doi: 10.3390/cancers11101460.
    Results Reference
    background
    PubMed Identifier
    25403204
    Citation
    Yuan C, Rubinson DA, Qian ZR, Wu C, Kraft P, Bao Y, Ogino S, Ng K, Clancy TE, Swanson RS, Gorman MJ, Brais LK, Li T, Stampfer MJ, Hu FB, Giovannucci EL, Kulke MH, Fuchs CS, Wolpin BM. Survival among patients with pancreatic cancer and long-standing or recent-onset diabetes mellitus. J Clin Oncol. 2015 Jan 1;33(1):29-35. doi: 10.1200/JCO.2014.57.5688. Epub 2014 Nov 17.
    Results Reference
    background
    PubMed Identifier
    31378465
    Citation
    Zhang AMY, Magrill J, de Winter TJJ, Hu X, Skovso S, Schaeffer DF, Kopp JL, Johnson JD. Endogenous Hyperinsulinemia Contributes to Pancreatic Cancer Development. Cell Metab. 2019 Sep 3;30(3):403-404. doi: 10.1016/j.cmet.2019.07.003. Epub 2019 Aug 1. No abstract available.
    Results Reference
    background

    Learn more about this trial

    Monitoring and Managing Glucose Levels in People With Pancreatic Cancer

    We'll reach out to this number within 24 hrs