EQW, DAPA, EQW/DAPA, DAPA/MET ER and PHEN/TPM ER in Obese Women With PolycysticOvary Syndrome (PCOS)

January 9, 2021 updated by: Karen Elkind-Hirsch, Woman's

Comparison of Dapagliflozin (DAPA) and Once-weekly Exenatide (EQW), Co-administered or Alone, DAPA/ Glucophage (DAPA/MET ER) and Phentermine/Topiramate (PHEN/TPM) ER on Metabolic Profiles and Body Composition in Obese PCOS Women

This is a randomized, single-blind, parallel 5 treatment group 24-week trial designed to directly compare the therapeutic effects of exenatide once weekly (EQW), dapagliflozin (DAPA), EQW plus DAPA, combined DAPA/metformin extended release (XR) and the weight loss medication, phentermine/topiramate extended release (PHEN/TPM ER) on metabolic and endocrinological parameters in overweight/obese non-diabetic women with PCOS. In this study, we will examine the efficacy of these therapies on metabolic parameters, body weight and body composition, anthropometric measurements, and reproductive function in a well-defined group of pre-menopausal overweight/obese, non-diabetic women with PCOS, focusing on their relationship to insulin resistance and obesity. We hope to determine which treatment(s) addressing the multifaceted disturbances of individual subgroups emerge as the preferable therapy.

Study Overview

Detailed Description

Polycystic ovary syndrome is a heterogeneous condition characterized by disordered reproductive and metabolic function which accounts for the myriad of clinical features including androgen excess, chronic anovulation, insulin resistance adiposity, and dyslipidemia. This syndrome is highly prevalent, affecting between 8 and 18% of the female population, depending on the diagnostic criteria used. Hyperandrogenism, ovarian dysfunction and metabolic abnormalities - the main determinants of PCOS - all appear to be involved in a synergistic way in the pathophysiology of PCOS. Women with PCOS are more likely to be obese: between 38 and 88% of women with PCOS are overweight or obese, although PCOS can also manifest in lean women. Obesity, particularly abdominal obesity, plays a central role in the development of PCOS, and exacerbates the reproductive and metabolic dysfunction. Rather than absolute body weight, it is the distribution of fat that is important with central (visceral) adiposity being a risk factor. Visceral adipose tissue is more metabolically active than subcutaneous fat and the amount of visceral fat correlates with insulin resistance and hyperinsulinemia. Weight gain is also often an important pathogenic factor, with PCOS usually becoming clinically manifest in women with a presumable genetic predisposition for PCOS who subsequently gain weight. Therefore, environmental (particularly dietary) factors are important. However, BMI is also influenced by genetic factors such as the fat mass and obesity-associated protein, and obesity is a highly heritable condition. Therefore, the weight gain responsible for the manifestation of PCOS in many women with this condition is itself influenced by genetic factors. Ethnicity, genetic background, personal and family history, degree of obesity must all be taken into account because they might aggravate or even trigger metabolic disturbances women with PCOS. Moreover, the incidence of glucose intolerance, dyslipidemia, gestational diabetes, and type 2 diabetes (DM2) is increased in women with PCOS at all weight levels and at a young age. PCOS may be a more important risk factor than ethnicity or race for glucose intolerance in young women. The exact factors responsible for this excess risk in women with PCOS have not been identified; family history of DM 2, obesity, insulin resistance, beta cell (ß-cell) secretory dysfunction, and hyperandrogenism are possible candidates. With better understanding of its pathophysiology, the metabolic consequences of the syndrome are now evident. Therefore, these patients need to be followed up even after their presenting complaint has been adequately resolved.

Lifestyle modification is a key component for the improvement of reproductive function for overweight, anovulatory women with PCOS. Even a modest weight loss 5% of total body weight can restore ovulation in overweight women with PCOS. Features of PCOS (e.g., hirsutism, testosterone levels, insulin resistance, menstrual cyclicity and ovulation) showed marked improvements, and PCOS frequently resolved after substantial weight loss induced by bariatric surgery. Recently a number of anti-diabetes medications have been approved which facilitate weight loss and improve the underlying insulin resistance. We reported that treatment with the glucagon like peptide -1 (GLP-1) agonist, exenatide for 24 weeks was superior to single agent metformin treatment in improving insulin action and reducing body weight and hyperandrogenism in obese women with PCOS. We further observed exenatide treatment significantly improved first-phase insulin responses to oral glucose administration. Since aberrant first-phase insulin secretion and impaired suppression of endogenous glucose production are major contributors to postprandial hyperglycemia and development of type 2 diabetes, the effects of exenatide once weekly [EQW (2 mg)] to target these defects, and normalize glucose excursions are likely to be clinically significant in obese patients with PCOS. Sodium/glucose cotransporter 2 (SGLT-2) inhibitors are the newest class of medications for diabetes management that have not been investigated for use in the women with PCOS. The SGLT2 inhibitor, dapagliflozin [DAPA (10 mg/day)] has an insulin-independent action, promotes weight loss, has a low incidence of hypoglycemia, and complements the action of other antidiabetic agents. The loss of glucose with attendant caloric loss contributes to weight loss; in addition, improvements in β cell function have been seen. Because the SGLT2 inhibitors have a distinct mechanism of action that is independent of insulin secretion, the efficacy of this class of drugs is not anticipated to decline in the presence of severe insulin resistance. The weight loss seen with SGLT2 inhibitors is similar to that seen with glucagon-like peptide 1 agonists, and may be more acceptable because they are oral agents. The resulting weight loss will further assist in decreasing insulin resistance, leading to increased glucose disposal thus contributing to an increased insulin secretion-insulin sensitivity (ISSI) index, the primary outcomes measure.

The non-diabetic female with PCOS offers a unique model to study the relationship between insulin resistance and adiposity. Women with PCOS demonstrate abnormal body composition characterized by a greater percent body fat, body fat mass, and increased ratio of fat to lean mass (F/L ratio). Studies using dual-energy X-ray absorptiometry (DEXA) methodology report a higher degree of metabolic dysfunction in patients with PCOS which appears be directly associated with their higher F/L ratio. Given that monotherapy and combined therapy with exenatide once weekly (EQW),and dapagliflozin (DAPA) along with DAPA/ metformin XR therapy are associated with weight loss introduces a confounder to the current study since it prevents distinguishing direct effects of the compounds on β-cell function vs. effects due to reduced adiposity. To control for loss of body mass and provide appropriate intervention in the remaining study arm we propose the use of a comparator weight loss drug alone, combination phentermine (PHEN)/topiramate (TPM) extended release (ER). To avoid the confounding relationship of body fat and insulin resistance, we will enroll only obese non-diabetic women with PCOS. All patients will receive diet and lifestyle counselling, including advice on exercise, according to usual clinical routine commencing during the lead-in period and continuing throughout the study We propose a randomized, single-blind, parallel 5 treatment group 24-week trial designed to directly compare the therapeutic effects of exenatide once weekly (EQW), dapagliflozin (DAPA), EQW plus DAPA, combined DAPA/metformin ER and the weight loss medication, phentermine/topiramate extended release (PHEN/TPM ER) on metabolic and endocrinological parameters in obese non-diabetic women with PCOS.

Study Type

Interventional

Enrollment (Actual)

119

Phase

  • Phase 3

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

    • Louisiana
      • Baton Rouge, Louisiana, United States, 70817
        • Woman's Hospital

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

16 years to 43 years (Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

Female

Description

Inclusion Criteria:

  1. Non-diabetic women (18-45 years)
  2. PCOS- NIH criteria hyperandrogenism and irregular menses
  3. Obese class I, II, and III (BMI >30<45)
  4. Willing to use effective contraception consistently during therapy which is defined as:

    1. an intrauterine device, tubal sterilization, or male partner vasectomy, or
    2. combination of two barrier methods with one being male condom.
  5. Written consent for participation in the study

Exclusion Criteria:

  1. Presence of significant systemic disease, heart problems including congestive heart failure, unstable angina or acute myocardial infarction, current infectious liver disease, acute stroke or transient ischemic attacks, history of pancreatitis, or diabetes mellitus (Type 1 or 2)
  2. Any hepatic diseases in the past (infectious liver disease, viral hepatitis, toxic hepatic damage, jaundice of unknown etiology) or severe hepatic insufficiency and/or significant abnormal liver function tests defined as aspartate aminotransferase (AST) >3x upper limit of normal (ULN) and/or alanine aminotransferase (ALT) >3x ULN
  3. Renal impairment (e.g., serum creatinine levels ≥1.4 mg/dL for women, or estimated glomerular filtration rate (eGFR) <60 mL/min/1.73 m2) or history of unstable or rapidly progressing renal disease or end stage renal disease. Patients with a history of nephrolithiasis are also excluded due to increased association with kidney stone formation.
  4. Uncontrolled thyroid disease (documented normal TSH), Cushing's syndrome, congenital adrenal hyperplasia or hyperprolactinemia
  5. Significantly elevated triglyceride levels (fasting triglyceride > 400 mg/dL)
  6. Untreated or poorly controlled hypertension (sitting blood pressure > 160/95 mm Hg)
  7. Use of hormonal medications, lipid-lowering (statins, etc.), anti-obesity drugs or weight loss medications (prescription or OTC) and medications known to exacerbate glucose tolerance (such as isotretinoin, hormonal contraceptives, gonadotropin-releasing hormone agonists, glucocorticoids, anabolic steroids, C-19 progestins) stopped for at least 8 weeks. Use of anti-androgens that act peripherally to reduce hirsutism such as 5-alpha reductase inhibitors (finasteride, spironolactone, flutamide) stopped for at least 4 weeks
  8. Prior history of a malignant disease requiring chemotherapy
  9. Patients at risk for volume depletion due to co-existing conditions or concomitant medications, such as loop diuretics should have careful monitoring of their volume status
  10. History of unexplained microscopic or gross hematuria, or microscopic hematuria at visit 1, confirmed by a follow-up sample at next scheduled visit.
  11. Presence of hypersensitivity to dapagliflozin or other SGLT2 inhibitors (e.g. anaphylaxis, angioedema, exfoliative skin conditions
  12. Known hypersensitivity or contraindications to use GLP1 receptor agonists (exenatide, liraglutide)
  13. Use of metformin, thiazolidinediones, GLP-1 receptor agonists, dipeptidyl peptidase 4 (DPP-4) inhibitors, SGLT2 inhibitors stopped for at least 4 weeks.
  14. Prior use of medication to treat diabetes except gestational diabetes
  15. Eating disorders (anorexia, bulimia) or gastrointestinal disorders
  16. Suspected pregnancy (documented negative serum pregnancy test), desiring pregnancy in next 6 months, breastfeeding, or known pregnancy in last 2 months
  17. Active or prior history of substance abuse (smoke or tobacco use within past 3 years) or significant intake of alcohol
  18. Having a history of bariatric surgery
  19. Patient not willing to use two barrier method contraception during study period (unless sterilized or have an IUD)
  20. Patients with glaucoma or history of increased intraocular pressure, or use of any medications to treat increased intraocular pressure
  21. Debilitating psychiatric disorder such as psychosis or neurological condition that might confound outcome variables. Patients with a history of bipolar disorder or psychosis, greater than one lifetime, episode of major depression, current depression of moderate or greater severity (PHQ-9score of 10 or more), presence or history of suicidal behavior or ideation with some intent to act on it, or antidepressant use that has not been stable for at least 3 months will also be excluded.
  22. Inability or refusal to comply with protocol
  23. Current participation or participation in an experimental drug study in previous three months

    -

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: Single

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Exenatide once weekly (EQW )
EQW- 2 mg subcutaneous (SC) injection once every seven days for 24 weeks
2 mg SC injection every 7 days
Other Names:
  • • Bydureon
  • • Long acting glucagon like peptide 1 (GLP1) agonist
Experimental: Dapagliflozin (DAPA)
DAPA-10 mg oral pill once daily in am for 24 weeks
One pill (10 mg) by mouth daily (QD) in am
Other Names:
  • • Farxiga
  • • SGLT2 inhibitor
Experimental: EQW plus DAPA
EQW- 2 mg SC injection once every seven days for 24 weeks DAPA-10 mg oral pill once daily in am daily for 24 weeks
2 mg SC injection every 7 days One pill (10 mg) by mouth QD in am
Other Names:
  • Bydureon plus Farxiga
  • Long acting GLP1 agonist plus SGLT2 inhibitor
Experimental: Dapagliflozin plus Glucophage (MET ER)
Combination DAPA / MET ER-10 mg /2000 mg oral pill daily with food for 24 weeks
DAPA/MET ER-5 mg /1000 mg - 1 pill by mouth in am with food for 4 weeks DAPA/MET ER-10 mg /2000 mg - 2 pills in am by mouth with food -final dose
Other Names:
  • Xigduo
  • •Combination SGLT2 inhibitor and metformin ER
Active Comparator: Phentermine /Topiramate (PHEN/ TPM) ER
Combination Phentermine /Topiramate ER -7.5 mg/46mg pill once daily in am for 24 weeks
PHEN 3.75/TPM ER 23mg - 1 pill by mouth once daily in am for 2 weeks. After 2 weeks, PHEN 7.5mg/TPM ER 46mg- 1 pill by mouth once daily in am
Other Names:
  • Qsymia

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Oral Disposition (Insulin Sensitivity-insulin Secretion) Index
Time Frame: 24 weeks of treatment
An estimation of β-cell compensatory function, the insulin secretion-sensitivity index (IS-SI) will be derived by applying the concept of the oral disposition index to measurements obtained during the 2-h OGTT and calculated as the index of insulin secretion factored by insulin sensitivity (ΔINS/ΔPG 30 x Matsuda SIOGTT) from the OGTT. A higher score shows improved pancreatic insulin responsiveness relative to resistance.
24 weeks of treatment

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Absolute Body Weight
Time Frame: 24 weeks of treatment
Treatment effect on body weight at 24 weeks of treatment
24 weeks of treatment
Body Mass Index (BMI)
Time Frame: 24 weeks of treatment
Treatment efficacy in reducing body mass at 24 weeks of treatment
24 weeks of treatment
Change in Percent Body Weight
Time Frame: Change from baseline (time 0) to study end (24 weeks)
Treatment effect on change in percent body weight from baseline
Change from baseline (time 0) to study end (24 weeks)
Central Adiposity (Waist Circumference)
Time Frame: 24 weeks of treatment
Treatment effect on loss of central adiposity after 24 weeks
24 weeks of treatment
Waist-to-Hip Ratio (WHR)
Time Frame: 24 weeks of treatment
Treatment impact on central adiposity after 24 weeks
24 weeks of treatment
Waist-to-Height Ratio (WHtR)
Time Frame: 24 weeks of treatment
Treatment impact on WHtR which is a measure of central adiposity
24 weeks of treatment
Total Fat Mass (kg) Evaluated by DEXA
Time Frame: 24 weeks of treatment
Treatment impact on total fat mass by DEXA
24 weeks of treatment
Total Body Fat (%) by DEXA
Time Frame: 24 weeks of treatment
Treatment impact on percent total body fat by DEXA
24 weeks of treatment
Android-Gynoid Ratio (AGR) as Determined by DEXA
Time Frame: 24 weeks of treatment
treatment impact on measure of central adiposity as determined by android/gynoid ratio
24 weeks of treatment
Trunk/Leg Fat Ratio by DEXA
Time Frame: 24 weeks of treatment
Treatment impact on trunk/limb ratio (measure of central adiposity) by DEXA
24 weeks of treatment
Fasting Blood Glucose
Time Frame: 24 weeks of treatment
Treatment impact on fasting concentration of glucose in the blood
24 weeks of treatment
OGTT Mean Blood Glucose (MBG)
Time Frame: 24 weeks of treatment
Treatment effect on MBG measured during the oral glucose tolerance test
24 weeks of treatment
Fasting Insulin Sensitivity (HOMA-IR)
Time Frame: 24 weeks of treatment
Treatment effect on the ratio HOMA-IR which is insulin resistance measure derived from fasting blood glucose and insulin and is calculated by insulin (mU/ml)*glucose (mmol/L)/22,5. The higher thenumber the more insulin resistant.
24 weeks of treatment
Matsuda Sensitivity Index Derived From the OGTT(SI OGTT)
Time Frame: 24 weeks of treatment
The SI IOGTT is a measure of peripheral insulin sensitivity derived from the values of Insulin (microunits per milliliter) and Glucose (milligrams per deciliter) obtained from the OGTT and the corresponding fasting values. SI (OGTT) = 10,000/ [(G fasting x I fasting) x (G OGTTmean x I OGTTmean)], where fasting glucose and insulin data are taken from time 0 of the OGTT and mean data represent the average glucose and insulin values obtained during the entire OGTT. The square root is used to correct for nonlinear distribution of insulin, and 10,000 is a scaling factor in the equation. The higher value, the more sensitive to insulin.
24 weeks of treatment
Corrected First Phase Insulin Secretion (IGI/HOMA-IR)
Time Frame: 24 weeks of treatment
Treatment effect on insulin secretion from 0 to 30 minutes after glucose load corrected for by fasting insulin sensitivity. A higher score shows improved first phase insulin secretion in response to glucose
24 weeks of treatment
Total Cholesterol Levels
Time Frame: 24 weeks of treatment
Treatment effect on blood concentrations of total cholesterol
24 weeks of treatment
Triglyceride (TRG) Levels
Time Frame: 24 weeks of treatment
Treatment effect on blood concentrations of triglycerides
24 weeks of treatment
Total Testosterone Concentrations
Time Frame: 24 weeks of treatment
Treatment effect on blood concentrations of total testosterone
24 weeks of treatment
Dehydroepiandrosterone Sulfate (DHEA-S) Levels
Time Frame: 24 weeks of treatment
Treatment effect on blood concentrations of DHEA-S
24 weeks of treatment
Free Androgen Index (FAI)
Time Frame: 24 weeks of treatment
Treatment effect on FAI calculated from total testosterone divided by sex hormone binding globulin (SHBG) levels. A higher score indicates a worse outcome.
24 weeks of treatment
Systolic Blood Pressure (SBP)
Time Frame: 24 weeks treatment
Treatment effect on SBP after 24 weeks of treatment
24 weeks treatment
Diastolic Blood Pressure (DBP)
Time Frame: 24 weeks of treatment
Treatment effect on DBP after 24 weeks
24 weeks of treatment

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Sponsor

Collaborators

Investigators

  • Principal Investigator: Karen Elkind-Hirsch, PhD, Woman's Hospital, Louisiana
  • Study Director: Drake Bellanger, MD, Woman's Hospital, Louisiana

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

March 22, 2016

Primary Completion (Actual)

July 22, 2020

Study Completion (Actual)

October 9, 2020

Study Registration Dates

First Submitted

December 15, 2015

First Submitted That Met QC Criteria

December 15, 2015

First Posted (Estimate)

December 18, 2015

Study Record Updates

Last Update Posted (Actual)

January 29, 2021

Last Update Submitted That Met QC Criteria

January 9, 2021

Last Verified

January 1, 2021

More Information

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Obesity

Clinical Trials on Exenatide once weekly (EQW )

3
Subscribe