DOSAGE AND CLINICAL USE

The sulfonylureas are effective, inexpensive, and easy to titrate. Sulfonylureas are

generally used as add-on therapy to patients unable to achieve BG goals on

metformin monotherapy. Like the other antidiabetic agents discussed here,

sulfonylureas may also be considered as monotherapy in patients with

contraindications to metformin therapy. The doses of the sulfonylureas are displayed

i n Table 53-26. As a general rule, one should begin with low doses and titrate

upward every 1 to 2 weeks until the desired goal is achieved. Exceeding maximal

doses is not likely to produce improvement, but may put the patient at risk for

adverse effects (see Case 53-18, Questions 5 and 6).

Thiazolidinediones

MECHANISM OF ACTION

The two available TZDs in the United States, rosiglitazone and pioglitazone, are

often referred to as insulin sensitizers. The precise molecular actions of these agents

remain to be clarified. TZDs bind to and activate a nuclear receptor (peroxisome

proliferator-activated receptor-γ [PPAR- γ]), which is expressed in many insulinsensitive tissues, including adipose (major site), skeletal muscle, and liver tissue.

184

PPAR-γ regulates transcription of genes that influence glucose and lipid metabolism.

For example, PPAR- γ stimulation increases the transcription of GLUT-4, a glucose

transporter that stimulates glucose uptake.

185 Reduced expression of GLUT-4 may

contribute to the development of insulin resistance. Pioglitazone activates PPAR-α in

addition to PPAR-y, which inhibits the transcription of tumor necrosis factor-αinduced vascular cell adhesion molecule (VCAM-1).

185 This duel effect by

pioglitazone enables HDL to be raised and triglycerides to be lowered. PPAR- α

activation is involved in actions that are anti-inflammatory and the PPAR-y and -α

activation can improve insulin sensitivity and lipid profiles.

186

TZDs either directly or indirectly sensitize adipose tissue to insulin action.

175,184

The effects may include stimulating apoptosis of large adipocytes, increasing the

number of small adipose cells, and promoting fatty acid uptake and storage in

adipose tissue. The subsequent reduction in free circulating fatty acids may spare

other insulin-sensitive tissues (e.g., liver, skeletal muscle, β-cells) from the effects of

lipotoxicity. TZDs also lower expression of tumor necrosis factor-α, a cytokine

produced by adipose tissue that may contribute to insulin resistance and fatty acid

release.

175,184 Other adipokines are likely involved, including adiponectin, resistin,

and leptin.

175 TZD interaction with adipocytes may be their primary mechanism of

action in sensitizing other tissues to insulin action. As with metformin, TZDs have

been shown to directly stimulate the AMPK pathway in liver and adipose tissue,

resulting in a lowering of glucose and free fatty acids.

164,184

Other observed effects of TZDs that may be beneficial in patients with Type 2

diabetes and metabolic syndrome include favorable effects on triglycerides,

reduction of inflammatory mediators, inhibition of vascular smooth muscle cell

proliferation, improved endothelial function, lowering microalbumin excretion, and

enhanced fi brinolysis.

175,184 Despite these apparently favorable effects on surrogate

measures of vascular disease, the data regarding TZDs and vascular risk are mixed

and controversial. In 2009, the FDA added language to the label for rosiglitazone

indicating an increased risk of angina and MI. The Food and Drug Administration and

GlaxoSmithKline instituted a Risk Evaluation and Management Strategy (REMS) to

restrict the distribution and prescribing of rosiglitazone; however, in May, 2014, the

REMS was changed to allow for the prescribing and filling of prescriptions of

rosiglitazone.

In summary, the TZDs clinically decrease insulin resistance in muscle and liver,

which enhances glucose utilization and decreases hepatic glucose output. They have

favorable effects on markers of vascular disease such as triglycerides and

inflammatory cytokines.

p. 1123

p. 1124

PHARMACOKINETICS

Rosiglitazone is completely absorbed, with peak plasma concentrations reached in

approximately 1 hour.

187 Pioglitazone has a bioavailability of 83%, with peak plasma

concentrations reached within 2 hours.

185 Food delays the time to peak concentration,

but does not alter the extent of absorption of either drug. Both TZDs are extensively

(>99%) protein bound, primarily to albumin. The plasma elimination half-life of

rosiglitazone is 3 to 4 hours.

187 Pioglitazone has a serum half-life of 3 to 7 hours, and

its metabolites have a serum half-life of 16 to 24 hours.

185 Rosiglitazone is

extensively metabolized in the liver by CYP 2C8 and to a much lesser extent by 2C9.

Its conjugated metabolites are considerably less potent than the parent drug and are

excreted two-thirds in urine and one-third in feces.

187 Pioglitazone is hepatically

metabolized, mainly by CYP 2C8 and 3A4, and to a lesser degree by CYP 1A1, to

three active metabolites; however, the main metabolites found in serum are M-III and

M-IV. Approximately 15% to 30% of the dose is recovered in the urine as

metabolites, with the remainder excreted either into the bile as unchanged drug or

into the feces as metabolites.

184,185

Because the action of the TZDs relies on gene transcription and protein

production, the onset and duration of action are unrelated to the plasma half-life. The

onset of their effect occurs in 1 to 2 weeks, although maximal effects are not usually

seen before 8 to 12 weeks. No dose adjustment is necessary in patients with renal

impairment. There is no dose adjustment needed for hepatic impairment with

pioglitazone; however, rosiglitazone should be avoided in patients with moderate-tosevere hepatic impairment.

185,187

ADVERSE EFFECTS

Hepatotoxicity

Liver failure has been very rarely reported with either rosiglitazone or pioglitazone,

although causality in most cases remains uncertain.

188–190 For both drugs, monitoring

of liver function tests (LFTs) is recommended at baseline, and then periodically

thereafter (see Contraindications and Precautions section). Many practitioners check

the LFTs every 3 to 6 months during the first year of therapy and then every 6 to 12

months thereafter.

Hematologic Effects

TZD therapy may result in small decreases in hemoglobin and hematocrit and,

infrequently, anemia.

175,184 These effects may be attributable to dilutional effects (see

below).

Weight Gain

Dose-related weight gain (2–3 kg for every 1% decrease in A1C) has been seen with

rosiglitazone and pioglitazone.

184 Weight gain is likely caused by fluid retention or fat

accumulation. The weight gain seems to be associated with an increase in peripheral

adipose tissue along with a reduction in visceral adiposity.

175,184

Vascular and Cardiovascular Effects

Increases in plasma volume and peripheral edema (4%–6%), possibly caused by

increased endothelial cell permeability, occur with the TZDs.

184 The incidence of

peripheral edema is greatly increased when TZDs are used in combination with

insulin.

The FDA added black box warnings to rosiglitazone and pioglitazone based on the

association of their use with an increased risk of developing or exacerbating HF in

patients with Type 2 diabetes.

185,187 TZDs are contraindicated for use in patients with

NYHA class III or IV HF and are not recommended in patients with acute or systemic

HF. Meta-analysis and retrospective observational studies have suggested that

rosiglitazone is associated with risk of MI,

191–193 but not for overall cardiovascular or

all-cause mortality.

193 Pioglitazone does not appear to increase the risk of MI or

mortality.

194–197 TZDs should be used with caution in patients with preexisting edema,

which may increase the risk of developing new-onset HF or exacerbating preexisting

HF (see Case 53-15).

Other Effects

Hypersensitivity reactions including rash, pruritus, urticaria, angioedema,

anaphylactic reaction, and Stevens–Johnson syndrome have been rarely reported with

rosiglitazone.

185,187 Macular edema has been rarely reported with TZDs.

185,187,198

Patients experiencing changes or worsening in vision should be referred to an

ophthalmologist for follow-up. In some cases, macular edema improved or resolved

after discontinuation of TZD therapy.

Pioglitazone has been associated with an increased risk of bladder cancer

compared to the general population and is contraindicated in patients with bladder

cancer.

199

In patients with previous history of bladder cancer, the need for use of

pioglitazone for glycemic control must outweigh the potential recurrence of bladder

cancer.

199 The FDA has released a safety communication warning that the use of

pioglitazone for longer than 1 year may increase the risk for bladder cancer.

199

Recently analyzed 10-year findings show no statistically significant increase in

bladder cancer, but a risk as previously identified cannot be ruled out. This metaanalysis identified a possible increased risk of pancreatic and prostate cancer.

Monitoring for causal effect of cancers should be continued.

199

An increased risk of distal limb bone fractures (e.g., forearm, hand, wrist, foot,

and ankle) and bone loss have been observed in women receiving TZDs.

200–203 Men

may also be at increased fracture risk, but the evidence is not as strong.

202,203 The

mechanism is thought to be reduced osteoblast differentiation as a result of increased

adipogenesis in the bone marrow.

204 The potential for fractures in older female

patients and patients on chronic steroids should be carefully considered before using

a TZD.

CONTRAINDICATIONS AND PRECAUTIONS

Type 1 diabetes: Because insulin is required for their action, TZDs should not be

used in people with type 1 diabetes.

Patients with type 2 diabetes using insulin: TZDs should be used with caution

because of the increased risk of developing edema.

Preexisting hepatic disease: Pioglitazone and rosiglitazone should not be used in

patients whose ALT is more than 2.5 times normal. TZDs should be discontinued

if the ALT is more than 3 times normal, if serum bilirubin levels begin to rise, or

if the patient complains of any symptoms that could be attributed to hepatitis (e.g.,

fatigue, nausea, vomiting, abdominal pain, and dark urine).

Symptomatic or severe (NYHA classes III and IV) HF: See previous discussion.

Myocardial ischemia (rosiglitazone only): See previous discussion.

Premenopausal anovulatory women: TZDs may cause resumption of ovulation and

menstruation in women with polycystic ovarian syndrome, placing these patients

at risk for an unwanted pregnancy.

History of hypersensitivity to TZDs.

Patients with osteoporosis or at risk for bone fractures (e.g., chronic steroid use).

Drugs metabolized by CYP 3A4: See the Drug Interactions section for further

details.

Patients with a current or previous history of bladder cancer should not use TZDs.

Macular edema: Patients should receive regular eye examinations to evaluate acute

visual changes.

DRUG INTERACTIONS

Coadministration of a TZD with other antidiabetic medications or insulin does not

alter the pharmacokinetics of either drug, but may increase the patient’s risk for

hypoglycemia. Pioglitazone

p. 1124

p. 1125

induces CYP 3A4 and may, therefore, decrease effectiveness of other drugs

metabolized by this enzyme, such as estrogens, cyclosporine, tacrolimus, and βhydroxy-β-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors.

Ketoconazole may significantly inhibit the metabolism of pioglitazone.

185 Patients

taking oral contraceptives or estrogen-replacement therapy should be informed of the

possible risk of decreased effectiveness of estrogen therapy. Rosiglitazone does not

seem to inhibit any of the major CYP enzymes.

187 Rifampin decreases the area under

the plasma concentration–time curve (AUC) for both rosiglitazone and pioglitazone,

although the clinical significance of this interaction is unknown. Pioglitazone is a

substrate of CYP2C8; therefore, interactions may occur when administered with

drugs that inhibit or induce CYP2C8.

205 The maximum dose of pioglitazone is 15 mg

once a day if given with a strong CYP2C8 inhibitor.

185 Gemfibrozil significantly

increases the AUC of both drugs. For patients receiving both a TZD and a

gemfibrozil, a dose reduction of the TZD may be warranted.

205

EFFICACY

The effects of TZDs on A1C and FPG are intermediate between those of acarbose

and the sulfonylureas or metformin.

175,184 When combined with other antidiabetic

agents in a poorly controlled type 2 diabetes patient, one can expect to see an

augmented effect on the A1C (0.9%–1.3% decrease with a sulfonylurea, 0.8%–1.0%

decrease with metformin, and 0.7%–1.0% decrease with insulin).

185,187 When added

to the therapy of a type 2 diabetes patient taking insulin, rosiglitazone and

pioglitazone can enhance glycemic control (approximately 0.6% lower with

pioglitazone) while decreasing insulin requirements; however, weight gain (>3 kg)

and increased edema will likely occur.

206

Individuals who are minimally responsive

or unresponsive to TZD therapy may include those who are not obese and have lower

levels of endogenous insulin.

Other potential benefits of the TZDs are their favorable, but variable, effects on

lipids.

184,185,187 Pioglitazone and to a lesser extent rosiglitazone may decrease

triglycerides. Both drugs may increase HDL-C levels by 10%. Rosiglitazone has

been observed to increase LDL-C by 8% to 16%, whereas pioglitazone may not

affect LDL-C. As noted, TZD therapy has been associated with weight gain, and this

may be substantial when used in combination with sulfonylureas or insulin.

DOSAGE AND CLINICAL USE

Patients who are unable to take or have failed metformin or sulfonylurea

monotherapy or who have not responded to combination therapy with other oral

antidiabetic agents are usually candidates for TZD therapy. For monotherapy or

combination therapy with a sulfonylurea, metformin, or insulin, the starting dose for

pioglitazone is 15 or 30 mg once daily with or without food. The dose can be titrated

up to a maximum of 45 mg/day.

185 The starting dose for rosiglitazone is 4 mg given

once daily or in divided doses. The dose may be increased after 8 to 12 weeks if

adequate response is not seen. The maximum daily dose is 8 mg.

187

Glucosidase Inhibitors

MECHANISM OF ACTION

T h e α-glucosidase inhibitors, acarbose

207 and miglitol,

208

reversibly inhibit

glucosidases present in the brush border of the mucosa of the small intestine. These

enzymes break down complex polysaccharides and disaccharides into glucose and

other absorbable monosaccharides. Enzyme inhibition delays carbohydrate digestion

and subsequent glucose absorption. Postprandial BG concentrations are therefore

lowered when these agents are taken with a meal containing complex carbohydrates.

PHARMACOKINETICS

Acarbose is minimally absorbed from the GI tract, with an oral bioavailability of the

parent drug of less than 2.0%.

207

It is extensively metabolized by GI amylases to

inactive metabolites. The peak plasma concentration occurs in approximately 1 hour

and the elimination half-life for acarbose is 2 hours, although there may be a longer

terminal half-life. Unlike acarbose, miglitol is absorbed. Absorption of miglitol is

saturable at higher doses (>25 mg) and peak concentration occurs in 2 to 3 hours.

208

The drug is primarily distributed in extracellular fluids and is not metabolized. After

a 25-mg dose, 95% of the drug is excreted unchanged by the kidneys within 24 hours.

ADVERSE EFFECTS

Flatulence, diarrhea, and abdominal pain are the most frequently reported adverse

effects of α-glucosidase inhibitors.

207,208

In placebo-controlled trials of acarbose,

these complaints were experienced by 74%, 31%, and 19% of subjects, respectively.

GI side effects are attributable to fermentation of unabsorbed carbohydrate in the

small intestine and can be minimized by slowly titrating the dose of either agent. GI

discomfort usually improves with continued therapy because induction of the αglucosidase enzymes occurs in the distal jejunum and terminal ileum.

In studies using doses of acarbose 300 mg/day or more, a transient increase in

serum hepatic transaminases was reported.

209 The manufacturer recommends

monitoring hepatic transaminases every 3 months for the first year of therapy and

periodically thereafter. If an elevation of serum transaminases occurs, the dose

should be decreased or discontinued if elevations persist. Because miglitol is not

metabolized, it does not seem to affect hepatic function.

CONTRAINDICATIONS AND PRECAUTIONS

Acarbose and miglitol are contraindicated with known hypersensitivity to the

medications.

207,208 Both medications are contraindicated in patients with DKA and

acarbose is contraindicated in patients with cirrhosis.

Gastrointestinal Conditions

Because of their profound GI effects (flatulence, diarrhea), acarbose and miglitol are

contraindicated in patients with malabsorption, inflammatory bowel disease, colonic

ulceration or other marked disorders of digestion or absorption, or with intestinal

obstruction.

207,208

Renal Impairment

Acarbose has not been studied in patients with severe renal impairment (SCr >2.0

mg/dL) and should not be used in these patients.

209 There is little information with

regard to safety of the use of miglitol in patients with a CrCl<25 mL/minute;

therefore, its use is contraindicated in these patients.

207,208

DRUG INTERACTIONS

Patients who use acarbose or miglitol in combination with other antidiabetic agents

may experience hypoglycemia. These reactions should be treated with dextrose

because acarbose may limit the availability of the disaccharide sucrose (table sugar).

Because acarbose and miglitol delay carbohydrate passage through the bowel, they

could influence the absorption kinetics of concomitantly administered drugs.

Conversely, because their own absorption may be diminished by digestive enzyme

preparations and charcoal, they should not be taken concomitantly with these

agents.

207–209 The bioavailability of digoxin can be reduced and may require dose

adjustment. Miglitol decreases the bioavailability of ranitidine and propranolol by

60% and 40%, respectively.

208

p. 1125

p. 1126

EFFICACY

By delaying the absorption of glucose after ingestion of complex carbohydrates and

disaccharides, the α-glucosidase inhibitors can lower postprandial plasma glucose

concentrations in patients with type 2 diabetes by 25 to 50 mg/dL.

207,208 FPG

concentrations remain unchanged or are slightly lowered (20–30 mg/dL), but this

effect may be related to decreased glucose toxicity, which improves insulin secretion

and action. Mean A1C values decline by 0.3% to 0.7%. Acarbose and miglitol have

no effect on weight or lipid profiles.

207,208

DOSAGE AND CLINICAL USE

Because of limited effects on A1C and their side effect profile, α-glucosidase

inhibitors are used infrequently and, when used, are usually given as add-on therapy

in patients who have failed monotherapy or combination therapy with other oral

antidiabetic agents. The recommended initial dose of either drug is up to 25 mg TID,

taken at the start of each meal.

207,208 The dosage of acarbose can be gradually

increased (e.g., 25 mg/meal) every 4 to 8 weeks to a maximum of 50 mg TID for

individuals weighing 60 kg or less, or 100 mg TID for individuals weighing more

than 60 kg. The dose for miglitol is titrated up after 4 to 8 weeks to a dose of 50 to

100 mg TID if needed, regardless of a patient’s weight. A maximal response is

observed at 6 months.

Incretin-Based Therapies

Incretins are insulinotropic hormones secreted from specialized neuroendocrine cells

in the small intestinal mucosa in response to carbohydrate ingestion and absorption.

13

The two hormones accounting for most incretin effects are glucose-dependent

insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1). GIP and

GLP-1 stimulate pancreatic β-cells in a glucose-dependent manner, contributing to

the early-phase insulin response. GLP-1 also inhibits pancreatic α-cells, thus

reducing glucagon secretion and hepatic glucose production. Incretin action is

efficient, but short lived. As they enter the blood vessels, incretins undergo rapid

metabolism via proteolytic cleavage by dipeptidyl peptidase-4 (DPP-4) to inactive

metabolites. Thus, only small amounts are needed to exert their effects on glucose

metabolism.

Glucagon-like Peptide-1 Agonists (GLP-1

Mimetics/Analogs)

Exenatide, extended-release exenatide, albiglutide, liraglutide, and dulaglutide are

the five available GLP-1 agonists in the United States. The formulations available

are a once-daily injection (liraglutide), twice-daily injection (exenatide), and onceweekly injections (extended-release exenatide, albiglutide and dulaglutide).

MECHANISM OF ACTION

GLP-1 mimetics and analogs have stability in the presence of DPP-4, resulting in a

longer duration of action than endogenous GLP-1. Exenatide is a synthetic form of

exendin-4, a peptide originally discovered from the saliva of the Gila monster.

210

Exendin-4 shares 50% of its amino acid sequence with GLP-1, demonstrating similar

affinity for receptor sites but a strong resistance to DPP-4. Liraglutide is a GLP-1

analog that is 97% homologous to human GLP-1, and reversibly binds to plasma

albumin owing to the C16 fatty acid side chain, thereby increasing resistance to DPP4 degradation.

211 Albiglutide has 2 tandem copies of modified human GLP-1 fused to

human albumin. The human fragment sequence has been modified to allow for

resistance to DPP-IV-mediated proteolysis, and in combination with the human

albumin moiety of the fusion protein, the half-life is extended allowing for onceweekly dosing.

212 Dulaglutide is a human GLP-1 receptor agonist that has 90%

homology to endogenous human GLP-1.

213 These agents augment early or first-phase

insulin response in response to elevated glucose concentrations (i.e., glucosedependent), moderate glucagon secretion, and decrease hepatic glucose production

without impairing the normal glucagon response to hypoglycemia. They slow gastric

emptying, thereby reducing the rate at which glucose is absorbed. In addition, they

suppress appetite, which may contribute to the prevention of weight gain and the

weight loss (1.5–5 kg) observed in patients. A promising action of these agents is

their potential to increase β-cell mass and preservation, which has been shown in

animal models.

PHARMACOKINETICS

After SC injection, exenatide reaches peak plasma concentrations in 2.1 hours.

214 The

injection site (abdomen, thigh, or upper arm) does not significantly alter its kinetics.

Both exenatide formulations are eliminated predominantly by glomerular filtration

with subsequent proteolytic degradation. The mean terminal half-life of exenatide is

2.4 hours, with levels measurable up to 10 hours, thus allowing for twice-daily

dosing. Its metabolism and elimination are dose independent. Extended-release

exenatide is released from microspheres over approximately 10 weeks after

administration. After discontinuation of therapy, minimal detectable concentrations

can be seen in approximately 10 weeks.

215

Absorption of liraglutide is delayed owing to its self-association in heptameric

aggregates within the injection depot that are too large to cross the capillary

membranes.

216 On disassociation of the heptamers at the absorption site, liraglutide is

absorbed. This delayed absorption is the primary reason for its prolonged action.

Liraglutide is highly protein bound (>98%), with a half-life of 13 hours, allowing for

once-daily dosing.

211,216 Metabolism of liraglutide occurs endogenously in a manner

similar to large proteins and there is no specific organ as its route of

elimination.

211,216

Albiglutide administered SC resulted in maximum concentrations in 3 to 5 days.

Steady state is achieved after 4 to 5 weeks of administration. Metabolism to small

peptides and amino acids occurs through a metabolic pathway involving proteolytic

enzymes in the vascular endothelium. The elimination half-life is approximately 5

days, which allows for once-weekly administration.

212

Dulaglutide achieves maximum plasma concentration in about 48 hours, and steady

state occurs between 2 to 4 weeks with once-weekly administration. There is no

statistically significant difference of exposure to dulaglutide between the site of

administration in the abdomen, upper arm, or thigh. Metabolism occurs through

general protein catabolism pathways into its component amino acids. The elimination

half-life is approximately 5 days.

213

ADVERSE EFFECTS

GI side effects are common and dose-dependent, particularly nausea, vomiting, and

diarrhea. Rates vary between agents with albiglutide appearing to have the lowest

rates in placebo-controlled trials.

212 These side effects may be lessened by starting

patients on lower doses for daily injections, ensuring correct timing and

administration of the drug, and titrating the dose slowly. Other reported side effects

have included decreased appetite and injection site reactions. Hypoglycemic risk can

be increased in patients who are also taking an oral insulin secretagogue (e.g.,

sulfonylurea) or insulin.

211–215

These agents have been rarely related to hypersensitivity reactions, acute

pancreatitis, and reduced renal function. Patients should be educated about symptoms

of acute pancreatitis, including severe abdominal pain accompanied by vomiting, and

instructed to report to their practitioner immediately. Patients in whom acute

pancreatitis is confirmed with no other probable cause should not be rechallenged

with GLP-1 agonists.

211–215

p. 1126

p. 1127

The development of antibodies against these agents is well established. In general,

the presence of antibodies does not appear to significantly affect the A1C reduction

seen with GLP-1 agonists, although some patients with high antibody titers may

experience reduced efficacy.

211–215

,

217 Patients who demonstrate adherence to therapy,

yet experience no change or worsening in glycemic control, should discontinue

therapy and be switched to alternative agents.

CONTRAINDICATIONS AND PRECAUTIONS

GLP-1 agonists are contraindicated in patients with known hypersensitivity. They

should not be used in patients with a history of pancreatitis.

211–215 They are not

recommended for use in patients with severe GI disease. Exenatide should not be

used in severe renal impairment (CrCl <30 mL/minute) or end-stage renal failure, or

in those requiring hemodialysis.

215,218 Dulaglutide and albiglutide have limited

clinical experience in patients with end-stage renal disease and should be used with

caution in these patients. If these patients experience GI adverse effects, renal

function should be closely monitored.

212,213

GLP-1 agonists are contraindicated in patients with a personal or family history of

medullary thyroid carcinoma (MTC) or in patients with multiple endocrine neoplasia

syndrome type 2 (MEN 2) due to risks in rodents. A causal relationship in humans

has not been established.

211–215 Currently, the FDA has required black box warnings

on each of the GLP-1 agonists for MTC, MEN 2, and thyroid cancer as well as a

REMS program for each medication.

DRUG INTERACTIONS

GLP-1 agonists may increase the risk for hypoglycemia when used with sulfonylureas

or insulin. Because of their mechanism of action, they may reduce the rate and extent

of absorption of orally administered drugs.

211–215 They should therefore be used with

caution in patients taking medications that require rapid GI absorption and are dosedependent on threshold concentrations for efficacy, such as antibiotics and oral

contraceptives. The manufacturers of twice-daily exenatide recommend that patients

take the affected medications at least 1 hour before exenatide administration.

214 There

have been case reports of an increased international normalized ratio, sometimes

associated with bleeding, in patients taking exenatide and warfarin. Patients should

be closely monitored, with dose adjustments to warfarin therapy made as

needed.

214,215

EFFICACY

In clinical trials, maximal doses of exenatide combined with a sulfonylurea,

metformin, a TZD, or sulfonylurea plus metformin therapy for 30 weeks reduced

FBG by 5 to 25 mg/dL, 2-hour postprandial BG by 60 to 70 mg/dL, and A1C by

0.8% to 1.0%.

214,215 Exenatide use for 80 weeks has been reported to reduce body

weight by 4 to 5 kg. In a 24-week trial comparing exenatide with extended-release

exenatide, the extended-release formulation resulted in a reduction in A1C of 1.6%

and a reduction in FBG of 25 mg/dL.

214,215 Clinical trials with liraglutide

monotherapy demonstrate decreases in FBG of 15 to 26 mg/dL and A1C of 0.8% to

1.1%, and weight loss of 2.1 to 2.5 kg.

214 When used as combination therapy,

additional A1C lowering of 1% to 1.5% can be expected.

219–221

Albiglutide as monotherapy in a 52-week trial resulted in A1C reduction of 0.7%

to 0.9% and reduction in FBG of 16 to 25 mg/dL. When used in combination therapy,

albiglutide reduced A1C by 0.6% to 0.8% and reduced FBG by 18 to 23 mg/dL.

212

Dulaglutide monotherapy resulted in a decrease in A1C of 0.7% to 0.8% and a

reduction of FBG of 26 to 29 mg/dL, as well as weight loss of 1.4 to 2.3 kg. When

dulaglutide is used in combination therapy, it results in an A1C reduction of 0.8% to

1.5%, a reduction in FBG of 16 to 41 mg/dL, and a weight loss of 0.2 to 2.7 kg.

213

DOSAGE AND CLINICAL USE

Exenatide (not the extended-release formulation) is approved for use as

monotherapy, whereas the other GLP-1 agonists are not recommended as

monotherapy per the manufacturers.

211–215 GLP-1 agonists are indicated as add-on

agents in patients with Type 2 diabetes who have been unable to reach target goals on

monotherapy with metformin or in combination therapy. Although not indicated for

weight loss, these agents may be helpful in patients with Type 2 diabetes who are

obese and struggling with obesity. Albiglutide and exenatide have been studied in

patients with Type 2 diabetes already on insulin glargine; albiglutide lowered the

A1C by 0.8%, whereas exenatide lowered the A1C by 1.7% and patients required

lower doses of insulin glargine compared with the placebo group.

212,214

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more