the primary glucose-lowering - care.diabetesjournals.org · 90% power to detect a difference in the...

8
The Primary Glucose-Lowering Effect of Metformin Resides in the Gut, Not the Circulation: Results From Short-term Pharmacokinetic and 12-Week Dose-Ranging Studies Diabetes Care 2016;39:198205 | DOI: 10.2337/dc15-0488 OBJECTIVE Delayed-release metformin (Met DR) is formulated to deliver the drug to the lower bowel to leverage the gut-based mechanisms of metformin action with lower plasma exposure. Met DR was assessed in two studies. Study 1 compared the bioavailability of single daily doses of Met DR to currently available immediate- release metformin (Met IR) and extended-release metformin (Met XR) in otherwise healthy volunteers. Study 2 assessed glycemic control in subjects with type 2 di- abetes (T2DM) over 12 weeks. RESEARCH DESIGN AND METHODS Study 1 was a phase 1, randomized, four-period crossover study in 20 subjects. Study 2 was a 12-week, phase 2, multicenter, placebo-controlled, dose-ranging study in 240 subjects with T2DM randomized to receive Met DR 600, 800, or 1,000 mg administered once daily; blinded placebo; or unblinded Met XR 1,000 or 2,000 mg (reference). RESULTS The bioavailability of 1,000 mg Met DR b.i.d. was 50% that of Met IR and Met XR (study 1). In study 2, 600, 800, and 1,000 mg Met DR q.d. produced statistically signicant, clinically relevant, and sustained reductions in fasting plasma glucose (FPG) levels over 12 weeks compared with placebo, with an 40% increase in potency compared with Met XR. The placebo-subtracted changes from baseline in HbA 1c level at 12 weeks were consistent with changes in FPG levels. All treatments were generally well tolerated, and adverse events were consistent with Gluco- phage/Glucophage XR prescribing information. CONCLUSIONS Dissociation of the glycemic effect from plasma exposure with gut-restricted Met DR provides strong evidence for a predominantly lower bowel-mediated mecha- nism of metformin action. 1 University of North Carolina School of Medicine, Chapel Hill, NC 2 University of Texas Health Science Center, San Antonio, TX 3 Dallas Diabetes and Endocrine Center, Dallas, TX 4 Elcelyx Therapeutics, San Diego, CA Corresponding author: Mark Fineman, mark. [email protected]. Received 6 March 2015 and accepted 16 July 2015. Clinical trial reg. nos. NCT02291510 and NCT01819272, clinicaltrials.gov. This article contains Supplementary Data online at http://care.diabetesjournals.org/lookup/ suppl/doi:10.2337/dc15-0488/-/DC1. A slide set summarizing this article is available online. © 2016 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for prot, and the work is not altered. See accompanying article, p. 187. John B. Buse, 1 Ralph A. DeFronzo, 2 Julio Rosenstock, 3 Terri Kim, 4 Colleen Burns, 4 Sharon Skare, 4 Alain Baron, 4 and Mark Fineman 4 198 Diabetes Care Volume 39, February 2016 CLIN CARE/EDUCATION/NUTRITION/PSYCHOSOCIAL

Upload: vongoc

Post on 24-Jul-2019

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Primary Glucose-Lowering - care.diabetesjournals.org · 90% power to detect a difference in the area under the curve (AUC) of at least 25% between 1,000 mg b.i.d. Met DR and 2,000

The Primary Glucose-LoweringEffect of Metformin Residesin the Gut, Not the Circulation:Results From Short-termPharmacokinetic and 12-WeekDose-Ranging StudiesDiabetes Care 2016;39:198–205 | DOI: 10.2337/dc15-0488

OBJECTIVE

Delayed-release metformin (Met DR) is formulated to deliver the drug to the lowerbowel to leverage the gut-based mechanisms of metformin action with lowerplasma exposure. Met DR was assessed in two studies. Study 1 compared thebioavailability of single daily doses of Met DR to currently available immediate-releasemetformin (Met IR) and extended-releasemetformin (Met XR) in otherwisehealthy volunteers. Study 2 assessed glycemic control in subjects with type 2 di-abetes (T2DM) over 12 weeks.

RESEARCH DESIGN AND METHODS

Study 1 was a phase 1, randomized, four-period crossover study in 20 subjects.Study 2 was a 12-week, phase 2, multicenter, placebo-controlled, dose-rangingstudy in 240 subjects with T2DM randomized to receive Met DR 600, 800, or 1,000mg administered once daily; blinded placebo; or unblinded Met XR 1,000 or 2,000mg (reference).

RESULTS

The bioavailability of 1,000 mgMet DR b.i.d. was∼50% that of Met IR andMet XR(study 1). In study 2, 600, 800, and 1,000 mg Met DR q.d. produced statisticallysignificant, clinically relevant, and sustained reductions in fasting plasma glucose(FPG) levels over 12 weeks compared with placebo, with an ∼40% increase inpotency compared withMet XR. The placebo-subtracted changes from baseline inHbA1c level at 12weeks were consistent with changes in FPG levels. All treatmentswere generally well tolerated, and adverse events were consistent with Gluco-phage/Glucophage XR prescribing information.

CONCLUSIONS

Dissociation of the glycemic effect from plasma exposure with gut-restricted MetDR provides strong evidence for a predominantly lower bowel-mediated mecha-nism of metformin action.

1University of North Carolina School of Medicine,Chapel Hill, NC2University of Texas Health Science Center, SanAntonio, TX3Dallas Diabetes and Endocrine Center, Dallas, TX4Elcelyx Therapeutics, San Diego, CA

Corresponding author: Mark Fineman,[email protected].

Received 6 March 2015 and accepted 16 July2015.

Clinical trial reg. nos. NCT02291510 andNCT01819272, clinicaltrials.gov.

This article contains Supplementary Data onlineat http://care.diabetesjournals.org/lookup/suppl/doi:10.2337/dc15-0488/-/DC1.

A slide set summarizing this article is availableonline.

© 2016 by the American Diabetes Association.Readersmayuse this article as longas thework isproperly cited, the use is educational and not forprofit, and the work is not altered.

See accompanying article, p. 187.

John B. Buse,1 Ralph A. DeFronzo,2

Julio Rosenstock,3 Terri Kim,4

Colleen Burns,4 Sharon Skare,4

Alain Baron,4 and Mark Fineman4

198 Diabetes Care Volume 39, February 2016

CLINCARE/ED

UCATION/N

UTR

ITION/PSY

CHOSO

CIAL

Page 2: The Primary Glucose-Lowering - care.diabetesjournals.org · 90% power to detect a difference in the area under the curve (AUC) of at least 25% between 1,000 mg b.i.d. Met DR and 2,000

Although metformin was introduced as atreatment for type 2diabetes (T2DM).50years ago, the mechanism of metforminaction is still debated (1). Historically, theglucose-lowering actions of metforminhave been attributed to its effects on mi-tochondrial function, AMPK, and glucagonreceptor–stimulated adenylate cyclase inthe liver and skeletal muscle, albeit atsuprapharmacological doses (2,3). A re-cent study (4) in rodents suggests that in-travenous metformin inhibits the redoxshuttle enzyme mitochondrial glycero-phosphate dehydrogenase, resulting inan altered hepatocellular redox state, re-duced conversion of lactate and glycerolto glucose, and decreased hepatic gluco-neogenesis at lower doses than are re-quired to affect AMPK. However, reports(5,6) that short-term intravenous metfor-min administration is less effective thanoral administration in rats and humanshave suggested that the gut may be im-portant for the glucose-lowering action ofmetformin. Gut effects include secretionof the enteroendocrine L-cell productsglucagon-like peptide 1 (GLP-1) and pep-tide YY, bile acid metabolism, and the gutmicrobiome (7,8).When currently available metformin

formulations (immediate-release met-formin [Met IR] and extended-releasemetformin [Met XR]) are orally adminis-tered, the absolute bioavailability is;50% of the total dose with the majorityof absorption occurring in the duode-num and jejunum (9–11). Importantly,as metformin is not metabolized in thegut (11), ;50% of a typical therapeuticdose is delivered to the distal small in-testine where it accumulates in the gutmucosa at concentrations up to 300times greater than concentrations inplasma (12). After a single dose of orallyadministered Met IR,;30% of the doseis recovered in the feces (10). Given thatmetformin absorption is transporterrate limited, lower doses (,1,000 mg)have higher bioavailability (11) but areless effective (13). Thus, we speculatedthat higher doses of metformin ($1,500mg) are necessary to “overwhelm” thetransporters in the proximal small intes-tine and deliver optimally effectivedoses of metformin to the lower bowel.The fact that there is a clear dose re-sponse for metformin while the phar-macokinetic (PK)/pharmacodynamicrelationship is weak (14) also supportsthe concept that presystemicmechanisms

may be important to its glucose-loweringeffect.

We tested the hypothesis that gut ex-posure to metformin predominantly ac-counts for its glucose-lowering effect byusing a delayed-release metformin (MetDR) formulation that targets the ileum, aregion of the gut where the absorptionof metformin is low (9,11). Met DR tar-gets the ileum through pH-dependentdissolution of the tablet without modi-fying the structure of the metforminmolecule. In this report, we describetwo studies demonstrating that MetDR has lower bioavailability comparedwith Met IR and Met XR in otherwisehealthy subjects (study 1) and that thedelivery of low doses of metformin(600–1,000 mg) to the lower bowel isat least as effective as similar doses ofMet XR in lowering plasma glucose levelsover 12 weeks in subjects with T2DM(study 2).

RESEARCH DESIGN AND METHODS

Met DR tablets were produced accord-ing to current good manufacturingpractices and comprise a Met IR hydro-chloride (HCl) core overlaid with a pro-prietary enteric coat. The enteric coat,which includes Eudragit polymers andother commonly used excipients, delaysdisintegration and dissolution of thetablet until it reaches a pH of 6.5 inthe distal small intestine and beyond.Tablet strengths used included 300and 500 mg metformin HCl. Placebotablets were visually identical but con-tained no active ingredient. Met IR andMet XR tablets used as comparatortreatments were commercially avail-able products (Glucophage; Bristol-Myers Squibb, New York, NY). Bothstudy protocols were performed in ac-cordance with good clinical practice andwere approved by the institutional re-view boards. All subjects provided writ-ten informed consent prior to studyenrollment.

Study 1 (clinical trial reg. no.NCT02291510, clinicaltrials.gov) was arandomized, four-period, crossover PKstudy in 20 otherwise healthy maleand female subjects who were between19 and 65 years of age and had a BMI of25–35 kg/m2. All subjects received 1 dayof dosing for each of four treatments(500 mg Met DR b.i.d., 1,000 mg Met DRb.i.d., 1,000mgMet IR b.i.d., and2,000mgMet XR q.d.) in a randomized (1:1:1:1)

sequence, separated by a washout pe-riod of 3–7 days. Met XR was adminis-tered once with the evening meal perthe prescribing information (15), andtwice-daily doses of Met DR and Met IRwere administered 12 h apart aftermeals. Plasma metformin concentra-tions were measured over a 36.5-h pe-riod that included five standardizedmeals. PK parameters were determinedusing noncompartmental analysis meth-ods based on the individual data forplasma metformin concentration overtime for each subject. PK parameterswere analyzed in the evaluable popula-tion (randomized subjects who com-pleted all treatment periods consistentwith protocol procedures) using amixed-effects model on a natural logscale with fixed effects for treatment,period, and sequence and subject withinsequence as a random effect. Resultswere exponentiated to obtain the geo-metric least squares (LS) mean ratios andcorresponding 90% CIs on the originalscale. Using two one-sided t tests withan a = 0.05 and an assumption that thewithin-subject variation would be atleast 25%, this sample size provided90% power to detect a difference in thearea under the curve (AUC) of at least25% between 1,000 mg b.i.d. Met DRand 2,000 mg q.d. Met XR.

Study 2 (clinical trial reg. no.NCT01819272, clinicaltrials.gov) was aphase 2, 12-week, randomized, pla-cebo-controlled, dose-response studyconducted in 240 subjects with T2DMand an estimated glomerular filtrationrate (eGFR) of $60 mL/min/1.73 m2

based on the Modification of Diet in Re-nal Disease equation. Subjects weremale or female, between 18 and 65years of age, had a BMI of 25–45 kg/m2,and had their T2DM treated withdiet and exercise alone or with metfor-min and/or a DPP-4 inhibitor (DPP-4i)only. Subjects were washed out of thesemedications for 14–17 days. Other inclu-sion criteria included an HbA1c level of7.0–9.5% if treated with diet and exer-cise alone or 6.0–9.5% if treated withmetformin/DPP-4i, serum creatinine,1.5 mg/dL (male) or ,1.4 mg/dL (fe-male), and an eGFR of $60 mL/min/1.73 m2 prior to randomization. Subjectswere randomized (1:1:1:1:1:1) to one ofsix treatment arms in a balancedmannerstratified by screening HbA1c level (,8%vs. $8%).

care.diabetesjournals.org Buse and Associates 199

Page 3: The Primary Glucose-Lowering - care.diabetesjournals.org · 90% power to detect a difference in the area under the curve (AUC) of at least 25% between 1,000 mg b.i.d. Met DR and 2,000

The double-blind treatment consistedof indistinguishable placebo, or 600,800, or 1,000 mg Met DR once daily inthe morning. Dosing with the morningmeal was selected based on a previoustrial demonstrating that once-daily dos-ing in the morning resulted in lower bio-availabilitywith equivalent fasting plasmaglucose (FPG) lowering than the same to-tal daily dose (1,000 mg) administeredwith the evening meal or split betweenthe morning and evening meals (clinicaltrial reg. no. NCT01804842, clinicaltrials.gov). Active treatment arms of 1,000and 2,000 mg Met XR administeredonce daily in the evening per prescribinginformation (15) were included for refer-ence. The 2,000 mg Met XR dose was ti-trated over 3 weeks; no other treatmentswere titrated.In study 2, the primary end point was

the change in FPG level from baseline toweek 4. Secondary end points includedchanges in HbA1c and FPG levels frombaseline to week 12 and changes inFPG from baseline to weeks 4, 8, and12. The baseline-corrected AUC of theFPG concentration-time curve at steadystate (AUC4–12wk) was also calculated foreach evaluable subject to integrate theFPG data collected through 12 weeksinto a single value, with week 4 chosenas the first value because 2,000 mg MetXR was titrated over the first 3 weeks.Fasting (premorning dose) PK andplasma lactate concentrations werealso measured.Changes in FPG level from baseline to

subsequent study visits were assessedusing an ANCOVAmodel with treatmentand baseline HbA1c levels (,8% and$8%) as factors and baseline FPG levelas a covariate. For the change in HbA1clevel, an ANCOVAmodel with treatmentas a factor and baseline HbA1c as a co-variate was used. Notable departuresfrom the Gaussian assumption were de-tected for the change in FPG level for allactive treatment groups. Therefore, themain analyses used the Kruskal-Wallistest for comparisons to placebo andthe Hodges-Lehmann method for CIsaround the median differences fromplacebo. Analyses of HbA1c used para-metric methods, as the departure fromthe Gaussian assumption for these didnot require alternative methods to beused.All analyses for the primary end point

were conducted using the intent-to-treat

(ITT) population (randomized subjectswho received at least one dose of thestudy drug) and the evaluable popula-tions (evaluable populations for weeks4 and 12 consisted of subjects who com-pleted the corresponding treatmentperiod without any major protocol vio-lations and with nonmissing FPG data atbaseline and the corresponding endpoint). A sample size of 40 subjectsper treatment group provided ;80%power to detect a statistically signifi-cant difference in week 4 FPG valuesbetween at least one Met DR treatmentgroup and the placebo group. The cal-culation was based on a two-sided twosample t test and a = 0.05 with balancedrandomization, a placebo-subtractedchange from baseline equal to220 mg/dL,a common SD of 30mg/dL, and amissingdata rate of 10%.

RESULTS

Metformin DR PK (Study 1)Study 1 randomized 20 subjects, and allsubjects received at least one dose ofthe study treatment. Most subjectswere male (70%), white (65%), and notHispanic or Latino (85%). The mean agewas 32.2 years, the mean weight was89.0 kg, and the mean BMI was 29.6kg/m2. One subject did not completeall four treatments because of an ad-verse event (AE) unrelated to studymedication (vessel puncture site hema-toma). All other subjects completed thestudy protocol procedures and were in-cluded in the evaluable population.

Mean plasma concentration-timeprofiles of metformin following singledaily dose administration (up to twodoses) were markedly lower for MetDR than Met IR and Met XR (Fig. 1A). Adelay in absorption with Met DR wasobserved; the median time prior to thefirst nonzero concentration ranged from6 to 7 h after the first (evening) doses ofMet DR compared with,1 h for Met IRandMet XR. The time to reach peak con-centrations was greater after the even-ing dose than the subsequent morningdose for both Met DR (10 and 6–7 h,respectively) and Met IR (5 and 3 h, re-spectively). Peak concentrations of MetDR twice daily were higher followingevening doses than morning doses(1,000 mg b.i.d.: 880 vs 604 ng/mL;500 mg b.i.d.: 538 vs 476 ng/mL). Collec-tively, these data suggest a diurnal effectin rate and extent of absorption that is

consistent with slowed transit during theevening and sleeping hours.

The relative bioavailability and expo-sure resulting from single daily doses ofMet DR twice daily versus Met IR twicedaily and Met XR once daily are shownin Fig. 1B. The mean maximal drug con-centration after dosing was higher forMet XR once daily (1,688 ng/mL) andMet IR twice daily (1,328 ng/mL) thanfor 1,000 and 500 mgMet DR b.i.d. doses(905 and 607 ng/mL). The mean AUCfrom time of dosing to the last measur-able concentration was also higher forthe Met XR once-daily dose (16,990 ng3h/mL) and the Met IR twice-daily dose(18,710 ng 3 h/mL) than for the 1,000and 500 mg Met DR b.i.d. dose (9,010and 6,160 ng3 h/mL). The rate and extentof exposure (AUC from time of dosing tothe last measurable concentration andmaximal drug concentration after dosing)from the 1,000 mg Met DR b.i.d. dosewere ;52% and 33% lower, respec-tively, than with the same dose of MetIR dose, and ;48% and 47% lower, re-spectively, than with 2,000 mg Met XRq.d. dose. The rate and extent of expo-sure from 500 mg Met DR b.i.d. were;68% and 55% lower, respectively,than with the 1,000 mg Met IR b.i.d.dose, and;65% and 64% lower, respec-tively, thanwith the 2,000mgMet XR q.d.dose. The reduction in plasma exposurewith both doses of Met DR was statisti-cally significant compared with Met IRand Met XR (P , 0.0001). The PK ofMet DRwas not dose proportional, whichis consistent with the known increasedbioavailability at lower doses. As ex-pected, the comparison of Met XR toMet IR demonstrated bioequivalencebased on total exposure.

Metformin DR Dose Response (Study 2)Study 2 randomly assigned 240 subjects(39–41 per group) to six treatmentgroups. Twenty-eight (11.7%) subjectsdiscontinued from the study early withdiscontinuation rates of 14.6%, 7.7%,7.5%, 12.5%, 10.0%, and 17.5%, respec-tively, for placebo, 600 mg Met DR, 800mg Met DR, 1,000 mg Met DR, 1,000 mgMet XR, and 2,000 mg Met XR doses(Supplemental Fig. 1) Themost commonreasons for discontinuation were lost tofollow-up (2.9%), protocol violation(2.5%), and loss of glucose control(2.5%). There were no statistically sig-nificant differences in demographics

200 Metformin Effects in the Gut Diabetes Care Volume 39, February 2016

Page 4: The Primary Glucose-Lowering - care.diabetesjournals.org · 90% power to detect a difference in the area under the curve (AUC) of at least 25% between 1,000 mg b.i.d. Met DR and 2,000

between treatment groups. Subjects ex-hibited relatively good glycemic control atbaseline (Table 1). All other characteris-tics related to diabetes were generallysimilar across treatment arms. Twenty-five subjects (10.4%) and 44 subjects(18.3%) were excluded from the week 4and week 12 evaluable populations,

respectively, prior to unblinding for notcompleting the treatment period withoutmajor protocol violations or for havingmissing values. The percentage of subjectsincluded in the week 12 evaluable popula-tion for each treatment group ranged from73.2% for the placebo group to 92.5% forthe 800 mg Met DR group.

All active treatment groups had im-provements in FPG level comparedwith placebo at week 4 (Fig. 2A). Therewere dose-dependent reductions in me-dian FPG level; the reduction producedby the 800 mg Met DR dose was statis-tically significant compared with that byplacebo. Median reductions for 1,000and 2,000 mg Met XR at week 4 werestatistically significant. The administra-tion of 1,000 mg Met DR resulted in a50% greater median reduction in plasmaglucose levels than 1,000 mg Met XRand ;72% of the 2,000 mg Met XR ef-fect. The baseline-corrected AUC4–12wkfor FPG (Fig. 2B) also decreased in a dose-dependent fashion (4.0, 296.0, 2108.0,2156.0,298.0, and2215.0mg/dL *week,respectively, for placebo, 600, 800, and1,000 mg Met DR and 1,000 and 2,000mg Met XR doses) with statistically sig-nificant reductions for all Met DR andMet XR groups compared with placebo(all P , 0.05). A dose response in FPGlevels was evident for both Met DR andMet XR with a left-shifted profile forMet DR compared with Met XR of;40%.

LS mean (SE) changes in HbA1c levelfrom baseline were negligible for all MetDR treatments and for 1,000 mg Met XRtreatment, while placebo increased theHbA1c level by 0.45% (0.14). Not surpris-ingly, the administration of 2,000 mgMet XR resulted in an LS mean (SE) re-duction of 0.21 (0.13) from baseline,since that dose was higher than themean dose of metformin that subjectswere receiving prior to the washout(1,438mg/day). The placebo-subtractedLS mean (SE) changes from baseline inHbA1c level at 12 weeks were 20.48%(0.19), 20.45% (0.18), and 20.35%(0.19) for 600, 800, and 1,000 mg MetDR, respectively, and20.45% (0.19) and20.67% (0.19) for 1,000 and 2,000 mgMet XR, respectively. All differenceswere statistically different from placebo(P , 0.05), with the exception of 1,000mg Met DR (P = 0.061).

Steady-state metformin concentra-tions were achieved by week 2 for allMet DR groups and the 1,000 mg MetXR group and byweek 4 for the 2,000mgMet XR group, which required dosetitration through week 3 (Fig. 2C).

Safety and TolerabilityConsistent with the Glucophage/Gluco-phage XR prescribing information (15),

Figure 1—Plasmametformin concentrations and bioavailability after administration of a singledaily dose (study 1). A: Mean (SD) plasma metformin concentrations by treatment and timepoint. Evaluable population (N = 19). Treatments were administered at t = 0 h (8:00 P.M.) and att = 12 h (8:00 A.M.) except for Met XR, which was administered as a single dose at t = 0 (blackarrows). Meals were administered at t = 20.42, 2.08, 11.5, 18.08, and 24.08 h relative to thefirst dose (dotted vertical lines). B: Relative bioavailability and exposure of single daily doses ofMet DR b.i.d. vs. Met IR b.i.d. and Met XR q.d. Evaluable population (N = 19). Data are ex-pressed as the percentage geometric LS mean ratio and the corresponding 90% upper confi-dence limit. ***P , 0.0001 vs. Met IR or Met XR. t, last quantifiable concentration followingdose administration.

care.diabetesjournals.org Buse and Associates 201

Page 5: The Primary Glucose-Lowering - care.diabetesjournals.org · 90% power to detect a difference in the area under the curve (AUC) of at least 25% between 1,000 mg b.i.d. Met DR and 2,000

the most common treatment-emergentAEs (TEAEs) were gastrointestinal in na-ture. In study 1, the most commonlyreported TEAEs in any treatment groupincluded diarrhea, nausea, vomiting,and headache. Most TEAEs were as-sessed as being unrelated to study treat-ment and were mild in intensity; therewere no deaths.In study 2, all active treatments were

well tolerated, and TEAEs were consis-tent with Glucophage/Glucophage XRprescribing information (15). However,the incidence of gastrointestinal TEAEswas relatively low (compared with pre-scribing information) in all active treat-ment groups, with gastrointestinalTEAEs reported by 7.3%, 12.8%, 17.5%,15.0%, 17.5%, and 12.5% of subjects, re-spectively, receiving placebo, 600mgMetDR, 800 mg Met DR, 1,000 mg Met DR,1,000 mg Met XR, and 2,000 mg Met XR.This suggests that the population was tol-erant to metformin, which is consistentwith 88%of subjects having receivedmet-formin prior to study enrollment andwashout.As metformin accumulation can re-

sult in increased lactate production,which, in turn, increases the risk of therare but serious metabolic complicationof lactic acidosis, the effects of Met DRon plasma lactate levels were also eval-uated in study 2. Mean lactic acid valueswere within normal ranges throughoutthe study, but were elevated from base-line by 0.31–0.33 mmol/L (median

0.19–0.29 mmol/L) for Met XR com-pared with 0.09–0.12 mmol/L (median20.22 to 0.22 mmol/L) for Met DR and0.16 mmol/L (median 0.17 mmol/L) forplacebo (Fig. 3). The lack of change frombaseline in lactate levels for the Met DRgroups most likely reflects lower met-formin exposure. One subject treatedwith 2,000 mg Met XR experiencedmoderate blood lactate increases for16 days (up to 5.9 mmol/L) withoutother AEs or changes to the treatmentregimen.

CONCLUSIONS

Metformin is the oldest and most com-monly prescribed oral glucose-loweringmedication in the world and isconsidered a first-line therapy for pa-tients in whom T2DM is newly diag-nosed (16). Nevertheless, there is noconsensus on its primary site of action,although it is generally agreed to havepleiotropic effects. The fact that metfor-min is not metabolized in vivo and is;50% bioavailable (11) allows for al-most equal exposures in the gut andplasma with typical dosing; however,until recently, most studies have fo-cused on systemically based mecha-nisms only. The liver has been themain focus of study, owing to decreasedhepatic glucose output with metformin(17) and observations of metformin ac-cumulation in the liver at concentrations;10 times greater than those in plasma(18,19). However, metformin also

accumulates in the intestine at concen-trations 300 times greater than inplasma (12). Thus, the gut is a major res-ervoir for metformin exposure and is po-tentially responsible for much of itsglucose-lowering effects, including en-hanced secretion of GLP-1 and peptideYY, which in turn affects systemic mecha-nisms including reducing hepatic glucoseproduction through glucagon suppressionand enhanced glucose-dependent insulinsecretion (15,20–23).

While the effects of metformin on in-creasing GLP-1 secretion have beenknown for some time (24–27), its signif-icance is debated. Interestingly, the in-crease in plasma GLP-1 levels resultingfrom metformin administration is simi-lar to that of a DPP-4i (20) and thuscould explain much of the glucose-lowering effect of metformin. In addi-tion, unlike a DPP-4i that reduces GLP-1degradation, metformin increases GLP-1 secretion and thus can significantly in-crease concentrations local to the L cell,which may in turn enhance neural signal-ing in the gut and portal vein to rapidlyregulate glycemic control (28–30).

The current study demonstrates thatmetformin primarily restricted to thegut effectively lowers plasma glucoselevels. The observation that low dosesof Met DR appear to be more effectivethan similar doses of the more bioavail-able Met XR suggests that the gut con-tribution to glucose lowering maybe more important than systemic

Table 1—Baseline characteristics of subjects with T2DM in the 12-week study (study 2)

PlaceboqAM

(n = 41)

Met DR 600mg qAM(n = 39)

Met DR 800mg qAM(n = 40)

Met DR 1,000mg qAM(n = 40)

Met XR 1,000mg qPM(n = 40)

Met XR 2,000mg qPM(n = 40)

All(N = 240)

Age, years 51 6 10 54 6 8 53 6 10 52 6 9 51 6 10 52 6 10 52 6 9

Male sex, % 46 46 32 65 45 47 47

White, black, other,a % 68/27/5 77/23/0 58/33/10 75/23/3 60/35/5 68/30/3 68/28/4

BMI, kg/m2 33.6 6 5.3 33.1 6 5.7 33.5 6 5.9 33.3 6 5.6 32.8 6 5.3 33.7 6 5.2 33.36 5.4

Duration of T2DM, years 7.0 5.9 6.0 5.9 6.6 7.0 6.4

HbA1c, % 7.4 6 1.0 7.5 6 0.9 7.3 6 0.9 7.4 6 1.0 7.4 6 1.0 7.4 6 1.0 7.4 6 0.9

FPG at screening, mg/dL 147 6 37 148 6 43 142 6 35 141 6 30 139 6 34 149 6 41 144 6 37

FPG at baseline, mg/dL 177 6 55 180 6 50 163 6 40 172 6 44 166 6 50 180 6 58 173 6 50

FPG, change from screening to baseline, mg/dL 31 6 45 33 6 44 21 6 26 31 6 34 27 6 44 31 6 34 29 6 38

Patients receiving therapy withMetb prior tostudy entry (% IR/% XR) 93 (87/13) 80 (84/16) 95 (84/16) 90(92/8) 85 (82/18) 88 (80/20) 88 (85/15)

Mean dose of prior Met,b mg (median) 1,308(1,000)

1,532(1,500)

1,446(1,500)

1,492 (1,600) 1,325(1,000)

1,541 (2,000) 1,438(1,500)

Patients with mild renal impairment,c % 34 26 40 33 43 35 35

Data are reported asmean6 SD or percentage of subjects for the ITT population (N = 240), unless otherwise indicated. Other, Asian, Pacific Islander/Native Hawaiian, American Indian/Alaska Native, or other; qAM, once daily in the morning; qPM, once daily in the evening. aPercentages maynot add up to 100 because of rounding. bIncludes Met IR and Met XR. cMild renal impairment defined as eGFR $60 to ,90 mL/min/1.73 m2.

202 Metformin Effects in the Gut Diabetes Care Volume 39, February 2016

Page 6: The Primary Glucose-Lowering - care.diabetesjournals.org · 90% power to detect a difference in the area under the curve (AUC) of at least 25% between 1,000 mg b.i.d. Met DR and 2,000

mechanisms. The apparent increase inpotency was most evident when com-paring the 600 mg Met DR dose to the1,000 mg Met XR dose (Fig. 2A and B).The fact that the administration of 1,000mg Met DR lowered fasting glucose lev-els by ;50% more than the same doseof 1,000 mg Met XR provides furthersupport of increased potency with MetDR. This leftward shift of the dose-response curve is all the more strikinggiven the ;50% reduction in plasmametformin exposure observed in study1 (Fig. 1) and the .75% reduction (Fig.2C) in fasting metformin concentrationsin study 2 at equally effective doses ofMet DR (600 mg) and Met XR (1,000mg). These data indicate that the gutis the primary site of action for theglucose-lowering effect of metforminand that plasma exposure is less impor-tant, at least at these therapeutic doses.

From a mechanistic perspective, alimitation of the current study is that ahigher Met DR dose was not included.Since the highest dose ofMetDR (1,000mg)achieved 70% of the glycemic controlof 2,000 mg Met XR, it is not possibleto determine whether all of the glucose-lowering effect of metformin can beexplained by lower-bowel gut-basedmechanisms. It is conceivable that thefull metformin effect requires a certainthreshold of upper-bowel metformin ex-posure and/or plasma exposure alongwith the lower-bowel exposure. The du-odenum has a low density of gut hor-mone-secreting L cells, and it has beenproposed that the rapid appearance ofGLP-1 following a meal is a result of acomplex integration of proximal and dis-tal neural and hormonal signaling (31).However, by virtue of its enteric coating,Met DR limits both proximal gut expo-sure and plasma exposure, so it is notpossible to quantitate their potential in-dividual contributions in these studies.Our data support the idea that the distalintestine is responsible for at least 70%of the maximal glucose-lowering effectof metformin, and future studies usinghigher doses may indicate an evengreater contribution.

Importantly, our data are not in con-flict with those from a recent report byMadiraju et al. (4) indicating that intra-venous metformin administration inrats reduces the conversion of lactateand glycerol to glucose, thus decreasinghepatic gluconeogenesis. Their finding is

Figure 2—Change in FPG and fasting metformin concentrations in the 12-week study (study 2).A: Median change in FPG level at week 4. Week 4 evaluable population (N = 215). P value fromKruskal-Wallis test; 95% CI based on Hodges-Lehmann estimation of the median difference vs.placebo; baseline is defined as the median measurement at day 1. B: Median change in and FPGAUC4–12wk (mg/dL * week). Week 12 evaluable population (N = 196). *P , 0.05 vs. placebo forpairwise comparison without adjustment. C: Median fasting plasma metformin concentrations.Week 12 evaluable population (N = 196).

care.diabetesjournals.org Buse and Associates 203

Page 7: The Primary Glucose-Lowering - care.diabetesjournals.org · 90% power to detect a difference in the area under the curve (AUC) of at least 25% between 1,000 mg b.i.d. Met DR and 2,000

entirely consistent with multiple obser-vations in animals and humans (includingour data) that high plasma concentrationsof the guanide/biguanide class increaseplasma lactate levels (14,32–34). Whiletheir study used metformin dosesequivalent to the clinically therapeuticrange (20 and 50 mg/kg), they were ad-ministered intravenously or intraperito-neally, which would result in muchhigher peak metformin plasma concen-trations and overall exposures thanwould be observed with oral adminis-tration. Thus, while gut-based mecha-nisms appear to account for the majorityof the glucose-lowering effect of met-formin at therapeutic doses, the inhibi-tion of the redox shuttle enzymemitochondrial glycerophosphate dehy-drogenase may have important glucose-lowering actions at higher metforminplasma exposures.Our data show an increase in plasma

lactate concentrations with Met XRtreatments compared with placebothat was not observed with any of theMet DR groups. Conditions that increasemetformin plasma exposure (renal im-pairment, hepatic insufficiency, orstates of circulatory dysfunction) can in-crease the risk of metformin-associatedlactic acidosis (MALA), a rare but life-threatening condition (15). With typicalmetformin use, the incidence of MALAis very low (,10 cases per 100,000patient-years) (15,35,36). MALA events

that are reported are usually associatedwith an elevated metformin dose orplasma exposure and an intercurrentevent that further disrupts lactate pro-duction or clearance, such as sepsis, re-duced tissue perfusion, anoxia, orimpaired hepatic metabolism (15,22,36–39). Optimization of the presystemicgut-restricted metformin mechanismsof action may yield a significant treat-ment advantage by lowering the risk ofMALA, particularly in at-risk popula-tions. Of note, simply reducing thedose of currently available metforminformulations to reduce the risk ofMALA is not a viable approach becauselow doses do not provide optimal glyce-mic control (13,40). Since the bioavail-ability of metformin increases withdecreasing dose, the lower bowel is ex-posed to disproportionately less metfor-min when doses of #1,000 mg areadministered (11), likely reducing oreliminating the glucose-lowering contri-bution of the gut-based mechanisms.

In summary, the delivery of metfor-min to the lower bowel with Met DRresulted in a glucose-lowering efficacycomparable to that with Met XR, butwith lower doses and significantly lowersystemic exposure. These data providesubstantial evidence that currently pre-scribed metformin doses work pre-dominantly in the gut and that thecontribution of systemic metformin issmall. Based on its gut-restricted

properties, Met DR may allow for themetformin treatment of patients withrenal impairment without the risk oflactic acidosis associated with metfor-min accumulation.

Acknowledgments. The authors thank SonjaBilles, PhD (August Scientific), for medicalwriting support, and Thomas Bicsak, PhD(Elcelyx Therapeutics), for manuscript reviewand revision. The authors also thank thepatients, investigators, and their staff for theirparticipation.Duality of Interest. This study was commis-sionedand fundedbyElcelyxTherapeutics. J.B.B.is a consultant at and holds stock in PhaseBioPharmaceuticals, under contract with the Uni-versity of North Carolina, from which he derivesno direct financial benefit, and is a consultantor investigator for Andromeda, AstraZeneca,Boehringer Ingelheim GmbH, Bristol-MyersSquibb, Elcelyx Therapeutics, Eli Lilly and Com-pany, GI Dynamics, GlaxoSmithKline, HalozymeTherapeutics,F.Hoffmann-LaRoche,IntarciaTher-apeutics, Johnson&Johnson, Lexicon, LipoScience,Medtronic,Merck,Metabolon,Metavention, NovoNordisk A/S, Orexigen Therapeutics, Inc., OsirisTherapeutics, Pfizer, PhaseBio Pharmaceuticals,Quest Diagnostics, Rhythm Pharmaceuticals,Sanofi, Spherix, Takeda, ToleRx, and TransTechPharma. R.A.D. is on the advisory boards ofAstraZeneca, Novo Nordisk, Janssen, Lexicon,and Boehringer Ingelheim GmbH; has receivedresearch support from Bristol-Myers Squibb,Boehringer Ingelheim GmbH, Takeda, andAstraZeneca; and is on speaker’s bureaus forNovo Nordisk and AstraZeneca. J.R. is a memberof advisory boards of and has received honorariaor consulting fees from Merck, Sanofi, NovoNordisk, Eli Lilly and Company, MannKind,GlaxoSmithKline, Takeda, Daiichi Sankyo, Novartis,Roche, Boehringer Ingelheim GmbH, Janssen,Lexicon, and Intarcia and has received researchgrants from Merck, Pfizer, Sanofi, Novo Nordisk,Eli Lilly and Company, GlaxoSmithKline, Takeda,Novartis, AstraZeneca, Janssen, Daiichi Sankyo,MannKind, Bristol-Myers Squibb, BoehringerIngelheim GmbH, Lexicon, and Intarcia. T.K. isan employee at Zafgen and a consultant atElcelyx Therapeutics. C.B. and S.S. are employeesof Elcelyx Therapeutics. A.B. and M.F. areemployees of and hold stock in Elcelyx Therapeu-tics. No other potential conflicts of interest rele-vant to this article were reported.Author Contributions. J.B.B. contributed todata acquisition, analysis, or interpretation andwrote the manuscript. R.A.D., J.R., A.B., and M.F.participated in conduct or design of the work andcontributed to data acquisition, analysis, or in-terpretation and wrote the manuscript. T.K. andC.B. participated in the conduct or design of thework and contributed todata acquisition, analysis,or interpretation. S.S. participated in the conductor design of the work. All authors contributed tothe revision of the manuscript, and all authorsreviewed and approved the final version. M.F. isthe guarantor of this work and, as such, had fullaccess to all the data in the study and takesresponsibility for the integrity of the data andthe accuracy of the data analysis.

Figure 3—Change from baseline to week 12 in fasting lactate (study 2). Data are reported as themean (SE) for the ITT population (N = 240). Normal lactate range is 0.5–2.2 mmol/L.

204 Metformin Effects in the Gut Diabetes Care Volume 39, February 2016

Page 8: The Primary Glucose-Lowering - care.diabetesjournals.org · 90% power to detect a difference in the area under the curve (AUC) of at least 25% between 1,000 mg b.i.d. Met DR and 2,000

Prior Presentation. Parts of these studies werepresented in abstract form at the 50th AnnualMeeting of the European Association for theStudy of Diabetes, Vienna, Austria, 15–19 Sep-tember 2014, and at the 75th Scientific Sessionsof the American Diabetes Association, Boston,MA, 5–9 June 2015.

References1. Pernicova I, Korbonits M. Metformin–modeof action and clinical implications for diabetesand cancer. Nat Rev Endocrinol 2014;10:143–1562. Miller RA, Chu Q, Xie J, Foretz M, Viollet B,Birnbaum MJ. Biguanides suppress hepatic glu-cagon signalling by decreasing production of cy-clic AMP. Nature 2013;494:256–2603. Zhou G, Myers R, Li Y, et al. Role of AMP-activated protein kinase in mechanism of met-formin action. J Clin Invest 2001;108:1167–11744. Madiraju AK, Erion DM, Rahimi Y, et al. Met-formin suppresses gluconeogenesis by inhibitingmitochondrial glycerophosphate dehydrogenase.Nature 2014;510:542–5465. Stepensky D, Friedman M, Raz I, Hoffman A.Pharmacokinetic-pharmacodynamic analysis ofthe glucose-lowering effect of metformin in di-abetic rats reveals first-pass pharmacodynamiceffect. Drug Metab Dispos 2002;30:861–8686. Bonora E, Cigolini M, Bosello O, et al. Lack ofeffect of intravenous metformin on plasma con-centrations of glucose, insulin, C-peptide, gluca-gon and growth hormone in non-diabeticsubjects. Curr Med Res Opin 1984;9:47–517. Napolitano A, Miller S, Nicholls AW, et al.Novel gut-based pharmacology of metforminin patients with type 2 diabetes mellitus. PLoSOne 2014;9:e1007788. ShinNR, Lee JC, LeeHY, et al. An increase in theAkkermansia spp. population induced by metfor-min treatment improves glucose homeostasis indiet-induced obese mice. Gut 2014;63:727–7359. Vidon N, Chaussade S, Noel M, Franchisseur C,Huchet B, Bernier JJ. Metformin in the digestivetract. Diabetes Res Clin Pract 1988;4:223–22910. Tucker GT, Casey C, Phillips PJ, Connor H,Ward JD, Woods HF. Metformin kinetics inhealthy subjects and in patients with diabetesmellitus. Br J Clin Pharmacol 1981;12:235–24611. Graham GG, Punt J, Arora M, et al. Clinicalpharmacokinetics of metformin. Clin Pharmaco-kinet 2011;50:81–9812. Bailey CJ, Wilcock C, Scarpello JH. Metforminand the intestine. Diabetologia 2008;51:1552–155313. Garber AJ, Duncan TG, Goodman AM, MillsDJ, Rohlf JL. Efficacy of metformin in type II

diabetes: results of a double-blind, placebo-controlled, dose-response trial. Am J Med 1997;103:491–49714. Hong Y, Rohatagi S, Habtemariam B,WalkerJR, Schwartz SL, Mager DE. Population expo-sure-response modeling of metformin in pa-tients with type 2 diabetes mellitus. J ClinPharmacol 2008;48:696–70715. Glucophage (Metformin Hydrochloride) Tab-lets; Glucophage XR (Metformin Hydrochloride)Extended-Release Tablets [package insert].Princeton, NJ, Bristol-Myers Squibb Company,200916. Inzucchi SE, Bergenstal RM, Buse JB, et al.;American Diabetes Association (ADA); Euro-pean Association for the Study of Diabetes(EASD). Management of hyperglycemia in type2 diabetes: a patient-centered approach: posi-tion statement of the American Diabetes Asso-ciation (ADA) and the European Association forthe Study of Diabetes (EASD). Diabetes Care2012;35:1364–137917. Cusi K, Consoli A, DeFronzo RA. Metaboliceffects of metformin on glucose and lactate me-tabolism in noninsulin-dependent diabetes melli-tus. J Clin Endocrinol Metab 1996;81:4059–406718. Wilcock C, Bailey CJ. Reconsideration of in-hibitory effect of metformin on intestinal glucoseabsorption. J Pharm Pharmacol 1991;43:120–12119. Beckmann R. Absorption, distribution in theorganism and elimination of metformin. Diabe-tologia 1969;5:318–32420. Migoya EM, Bergeron R, Miller JL, et al. Di-peptidyl peptidase-4 inhibitors administered incombination with metformin result in an additiveincrease in the plasma concentration of activeGLP-1. Clin Pharmacol Ther 2010;88:801–80821. Hundal RS, Krssak M, Dufour S, et al. Mech-anism by which metformin reduces glucose pro-duction in type 2 diabetes. Diabetes 2000;49:2063–206922. Bailey CJ, Turner RC. Metformin. N Engl JMed 1996;334:574–57923. Habib AM, Richards P, Rogers GJ, ReimannF, Gribble FM. Co-localisation and secretion ofglucagon-like peptide 1 and peptide YY fromprimary cultured human L cells. Diabetologia2013;56:1413–141624. Mannucci E, Ognibene A, Cremasco F, et al.Effect of metformin on glucagon-like peptide 1(GLP-1) and leptin levels in obese nondiabeticsubjects. Diabetes Care 2001;24:489–49425. Mannucci E, Tesi F, Bardini G, et al. Effects ofmetformin on glucagon-like peptide-1 levels inobese patients with and without type 2 diabetes.Diabetes Nutr Metab 2004;17:336–342

26. Molloy AM, Ardill J, Tomkin GH. The ef-fect of metformin treatment on gastric acidsecretion and gastrointestinal hormone lev-els in normal subjects. Diabetologia 1980;19:93–9627. Yasuda N, Inoue T, Nagakura T, et al. En-hanced secretion of glucagon-like peptide 1 bybiguanide compounds. Biochem Biophys ResCommun 2002;298:779–78428. Larsen PJ, Holst JJ. Glucagon-related pep-tide 1 (GLP-1): hormone and neurotransmitter.Regul Pept 2005;128:97–10729. Donath MY, Burcelin R. GLP-1 effects onislets: hormonal, neuronal, or paracrine? Diabe-tes Care 2013;36(Suppl. 2):S145–S14830. Bohorquez DV, Shahid RA, Erdmann A, et al.Neuroepithelial circuit formed by innervation ofsensory enteroendocrine cells. J Clin Invest2015;125:782–78631. Nauck MA. Incretin-based therapies fortype 2 diabetes mellitus: properties, functions,and clinical implications. Am J Med 2011;124(Suppl.):S3–S1832. Lalau JD, Lacroix C, Compagnon P, et al. Roleof metformin accumulation in metformin-associated lactic acidosis. Diabetes Care 1995;18:779–78433. Seidowsky A, Nseir S, Houdret N, Fourrier F.Metformin-associated lactic acidosis: a prognos-tic and therapeutic study. Crit Care Med 2009;37:2191–219634. Al-Abri SA, Hayashi S, Thoren KL, Olson KR.Metformin overdose-induced hypoglycemia inthe absence of other antidiabetic drugs. ClinToxicol (Phila) 2013;51:444–44735. Stang M, Wysowski DK, Butler-Jones D. In-cidence of lactic acidosis in metformin users.Diabetes Care 1999;22:925–92736. Misbin RI, Green L, Stadel BV, Gueriguian JL,Gubbi A, Fleming GA. Lactic acidosis in patientswith diabetes treated with metformin. N Engl JMed 1998;338:265–26637. Peters N, Jay N, Barraud D, et al. Metfor-min-associated lactic acidosis in an intensivecare unit. Crit Care 2008;12:R14938. Almirall J, Briculle M, Gonzalez-ClementeJM. Metformin-associated lactic acidosis intype 2 diabetes mellitus: incidence and presen-tation in common clinical practice. Nephrol DialTransplant 2008;23:2436–243839. Lucis OJ. The status of metformin in Can-ada. Can Med Assoc J 1983;128:24–2640. Jones GC, Sainsbury CA. Comment on “Ajustification for less restrictive guidelines onthe use of metformin in stable chronic renalfailure.” Diabet Med 2015;32:287

care.diabetesjournals.org Buse and Associates 205