Climara Pro

Name: Climara Pro

Warnings

Included as part of the PRECAUTIONS section.

Overdose

Overdosage of estrogen plus progestin may cause nausea, vomiting, breast tenderness, abdominal pain, drowsiness and fatigue, and withdrawal bleeding may occur in women. Treatment of overdose consists of discontinuation of Climara Pro therapy with institution of appropriate symptomatic care.

Climara Pro Overview

Climara Pro is a prescription medication used after menopause to reduce moderate to severe hot flashes and help reduce your chances of getting osteoporosis (thin weak bones).

Climara Pro contains two hormones, estradiol and levonorgestrel, and belongs to a group of drugs called estrogen and progestin combinations. Climara Pro works as a hormone replacement to relieve issues caused by hormonal changes. 

Climara Pro comes in a patch form and is usually applied to the skin once weekly. 

Common side effects of Climara Pro include reactions at the application site, vaginal bleeding, and breast pain. 

Dosage Forms and Strengths

Climara Pro (estradiol/levonorgestrel transdermal system) 0.045 mg/day estradiol and 0.015 mg/day levonorgestrel – each 22 cm2 system contains 4.4 mg of estradiol and 1.39 mg of levonorgestrel.

Adverse Reactions

The following serious adverse reactions are discussed elsewhere in the labeling:

• Cardiovascular Disorders [see Boxed Warning, Warnings and Precautions (5.1)] • Malignant Neoplasms [see Boxed Warning, Warnings and Precautions (5.2)]

Clinical Trials Experience

Because clinical trials are conducted under widely varying conditions, adverse reaction rates observed in the clinical trials of a drug cannot be directly compared to rates in the clinical trials of another drug and may not reflect the rates observed in practice.

The data described below is from a one-year, prospective, multicenter, double blind, double dummy, randomized, controlled trial investigating the effect of three different dosage combinations of E2/LNG versus E2 alone on the development of endometrial hyperplasia. All women were postmenopausal, had a serum estradiol level of less than 20 pg/mL, and the sample included both symptomatic and asymptomatic women. The data below includes all adverse reactions reported at a frequency of >3% in the E2/LNG 0.045 / 0.015 group (the approved dosage for Climara Pro, N=212) and the E2 alone group (N=204).

Table 1: All Treatment Emergent Reactions Regardless of Relationship Reported at a Frequency of >3% with Climara Pro in the 1-year Endometrial Hyperplasia Study
* N = total number of subjects in a treatment group; n = number of subjects with event.

Body System

  Adverse Reaction

Climara Pro
0.045 / 0.015

E2

N* = 212

N = 204

Body as a Whole

  Abdominal pain

9 (4.2)

11 (5.4)

  Accidental injury

7 (3.3)

6 (2.9)

  Back pain

13 (6.1)

12 (5.9)

  Flu syndrome

10 (4.7)

13 (6.4)

  Infection

7 (3.3)

10 (4.9)

  Pain

11 (5.2)

13 (6.4)

Cardiovascular System

  Hypertension

7 (3.3)

9 (4.4)

Digestive System

  Flatulence

8 (3.8)

11 (5.4)

Metabolic and Nutritional

  Edema

8 (3.8)

5 (2.5)

  Weight gain

6 (2.8)

10 (4.9)

Musculoskeletal System

  Arthralgia

9 (4.2)

10 (4.9)

Nervous System

  Depression

12 (5.7)

7 (3.4)

  Headache

11 (5.2)

14 (6.9)

Respiratory System

  Bronchitis

9 (4.2)

7 (3.4)

  Sinusitis

8 (3.8)

12 (5.9)

  Upper respiratory infection

28 (13.2)

26 (12.7)

Skin and Appendages

  Application site reaction

86 (40.6)

69 (33.8)

  Breast pain

40 (18.9)

20 (9.8)

  Rash

5 (2.4)

10 (4.9)

Urogenital System

  Urinary Tract Infection

7 (3.3)

8 (3.9)

  Vaginal Bleeding

78 (36.8)

44 (21.6)

  Vaginitis

4 (1.9)

6 (2.9)

Irritation potential of Climara Pro was assessed in a 3-week irritation study. The study compared the irritation of a Climara Pro placebo patch (22 cm2) to a placebo (25 cm2). Visual assessments of irritation were made on Day 7 of each wear period, approximately 30 minutes after patch removal using a 7-point scale (0 = no evidence of irritation; 1 = minimal erythema, barely perceptible; 2 = definite erythema, readily visible, or minimal edema, or minimal papular response; 3–7 = erythema and papules, edema, vesicles, strong extensive reaction).

The mean irritation scores were 0.13 (week 1), 0.12 (week 2), and 0.06 (week 3) for the Climara Pro placebo. The mean scores for the Climara placebo were 0.2 (week 1), 0.26 (week 2), 0.12 (week 3). There were no irritation scores greater than 2 at any timepoint in any subject.

In controlled clinical trials, withdrawals due to application site reactions occurred in 6 (2.1 percent) of subjects in the 12-week symptom study and in 71 (8.5 percent) of subjects in the 1-year endometrial protection study.

Postmarketing Experience

The following adverse reactions have been identified during post-approval use of the Climara Pro transdermal system. Because these reactions are reported voluntarily from a population of uncertain size, it is not always possible to reliably estimate their frequency or establish a causal relationship to drug exposure.

Genitourinary System

Changes in bleeding patterns

Gastrointestinal

Abdominal distension,* abdominal pain,* nausea

Skin

Alopecia, night sweats, pruritus,* Rash,* hot flush*

Central Nervous System

Dizziness, headache, insomnia

Miscellaneous

Application site reaction,* weight increased, anaphylactic reaction

* Combined two or more similar ARs

Overdosage

Overdosage of estrogen plus progestin may cause nausea, vomiting, breast tenderness, abdominal pain, drowsiness and fatigue, and withdrawal bleeding may occur in women. Treatment of overdose consists of discontinuation of Climara Pro therapy with institution of appropriate symptomatic care.

Climara Pro Description

Climara Pro (estradiol/levonorgestrel transdermal system) is an adhesive-based matrix transdermal patch designed to release both estradiol and levonorgestrel, a progestational agent, continuously upon application to intact skin. The 22 cm2 Climara Pro system contains 4.4 mg estradiol and 1.39 mg levonorgestrel and provides a nominal delivery rate (mg per day) of 0.045 estradiol and 0.015 levonorgestrel.

Estradiol USP has a molecular weight of 272.39 and the molecular formula is C18H24O2.

Levonorgestrel USP has a molecular weight of 312.4 and a molecular formula of C21H28O2.

The structural formulas for estradiol and levonorgestrel are:

The Climara Pro transdermal system comprises 3 layers. Proceeding from the visible surface towards the surface attached to the skin, these layers are:

• A translucent polyethylene backing film. • An acrylate adhesive matrix containing estradiol and levonorgestrel. • A protective liner of either siliconized or fluoropolymer coated polyester film. The protective liner is attached to the adhesive surface and must be removed before the system can be used.

The active components of the transdermal system are estradiol and levonorgestrel. The remaining components of the transdermal system (acrylate copolymer adhesive and polyvinylpyrrolidone/vinyl acetate copolymer) are pharmacologically inactive.

Climara Pro - Clinical Pharmacology

Mechanism of Action

Endogenous estrogens are largely responsible for the development and maintenance of the female reproductive system and secondary sexual characteristics. Although circulating estrogens exist in a dynamic equilibrium of metabolic interconversions, estradiol is the principal intracellular human estrogen and is substantially more potent than its metabolites, estrone and estriol, at the receptor level.

The primary source of estrogen in normally cycling adult women is the ovarian follicle, which secretes 70 to 500 mcg of estradiol daily, depending on the phase of the menstrual cycle. After menopause, most endogenous estrogen is produced by conversion of androstenedione, which is secreted by the adrenal cortex, to estrone in the peripheral tissues. Thus, estrone and the sulfate conjugated form, estrone sulfate, are the most abundant circulating estrogens in postmenopausal women.

Estrogens act through binding to nuclear receptors in estrogen-responsive tissues. To date, two estrogen receptors have been identified. These vary in proportion from tissue to tissue.

Circulating estrogens modulate the pituitary secretion of the gonadotropins, luteinizing hormone (LH) and FSH, through a negative feedback mechanism. Estrogens act to reduce the elevated levels of these hormones seen in postmenopausal women.

Levonorgestrel inhibits gonadotropin production resulting in retardation of follicular growth and inhibition of ovulation.

Studies to assess the potency of progestins using estrogen-primed postmenopausal endometrial biochemistry and morphologic features have shown that levonorgestrel counteracts the proliferative effects of estrogens on the endometrium.

Pharmacodynamics

There are no pharmacodynamic data for Climara Pro.

Pharmacokinetics

Absorption

Transdermal administration of Climara Pro produces mean maximum estradiol concentrations in serum in about 2 to 2.5 days. Estradiol concentrations equivalent to the normal ranges observed at the early follicular phase in premenopausal women are achieved within 12–24 hours after the first application.

In one study, steady state estradiol concentrations in serum were measured during week 4 in 44 healthy, postmenopausal women during four consecutive Climara Pro applications of two formulations (0.045 mg estradiol/0.03 mg levonorgestrel and 0.045 mg estradiol/0.015 mg levonorgestrel) to the abdomen (each dose was applied for four 7-day periods). Both formulations were bioequivalent in terms of estradiol and estrone Cmax and AUC parameters. A summary of Climara Pro single and multiple applications estradiol, estrone and levonorgestrel pharmacokinetic parameters is shown in Table 2.

Table 2: Summary of Mean Pharmacokinetic Parameters

Summary of Mean (± SD) Pharmacokinetic Parameters Following a Single Application
of Climara Pro in 24 Healthy Postmenopausal Women

Parameter

Units

Estradiol

Estrone

Levonorgestrel

Single application
Week 1 Data

Cave

Pg/mL

37.7 ± 10.4

41 ± 15

136 ± 52.7

Cmax

Pg/mL

54.3 ± 18.9

43.9 ± 14.9

138 ± 51.8

Tmax

Hours

42

84

90

Cmin

Pg/mL

27.2 ± 7.66

32.6 ± 14.3

110 ± 41.7

AUC

Pg.h/mL

6340 ± 1740

6890 ± 2520

22900 ± 8860

Summary of Mean (± SD) Pharmacokinetic Parameters (Week 4) Following Four Consecutive Weekly Applications of Climara Pro in 44 Healthy Postmenopausal Women

Multiple application
Week 4 Data

Cave

Pg/mL

35.7 ± 11.4

45.5 ± 62.6

166 ± 97.8

Cmax

Pg/mL

50.7 ± 28.6

81.6 ± 252

194 ± 111

Tmax

Hours

36

48

48

Cmin

Pg/mL

33.8 ± 28.7

72.5 ± 253

153 ± 69.6

AUC

Pg.h/mL

6002 ± 1919

7642 ± 10518

27948 ± 16426

All mean parameters are arithmetic means except Tmax which is expressed as the median.

At steady state, Climara Pro maintains during the application period an average serum estradiol concentration of 35.7 pg/mL as depicted in Figure 1.

Figure 1: Mean Estradiol Concentration Profile (Week 4) Following Four Consecutive Weekly Applications of Climara Pro

Following the application of the Climara Pro transdermal system, levonorgestrel concentrations are maximum in about 2.5 days. At steady state, Climara Pro maintains during the application period an average serum levonorgestrel concentration of 166 pg/mL as depicted in Figure 2. The mean levonorgestrel pharmacokinetic parameters of Climara Pro are summarized in Table 2.

Figure 2: Mean Levonorgestrel Concentration Profile (Week 4) Following Four Consecutive Weekly Applications of Climara Pro

Adhesion

A study of the adhesion potential of Climara Pro was conducted in 104 healthy women of 45–75 years of age. Each woman applied a placebo patch, containing only the Climara Pro adhesive without active ingredient, to the upper outer abdominal areas weekly for three weeks. The adhesion assessment was done visually on Days 2, 4, 5, 6 and 7 of each of the three weeks using a four-point scale. The mean scores ranked in the highest category possible on the 0 to 4 scale demonstrating clinically acceptable adhesion performance

Excretion

Estradiol, estrone, and estriol are excreted in the urine along with glucuronide and sulfate conjugates. Following patch removal, serum estradiol concentrations decline with a mean (± SD) terminal half-life of 3± 0.67 hours.

Levonorgestrel and its metabolites are primarily excreted in the urine. Mean (± SD) terminal half-life for levonorgestrel was determined to be 28 ± 6.4 hours.

Metabolism

Exogenous estrogens are metabolized in the same manner as endogenous estrogens. Circulating estrogens exist in a dynamic equilibrium of metabolic interconversions. These transformations take place mainly in the liver. Estradiol is converted reversibly to estrone, and both can be converted to estriol, which is a major urinary metabolite. Estrogens also undergo enterohepatic recirculation via sulfate and glucuronide conjugation in the liver, biliary secretion of conjugates into the intestine, and hydrolysis in the intestine followed by reabsorption. In postmenopausal women, a significant proportion of the circulating estrogens exist as sulfate conjugates, especially estrone sulfate, which serves as a circulating reservoir for the formation of more active estrogens.

The most important metabolic pathway for levonorgestrel occurs in the reduction of the Δ4- and the 3-oxo-group as well as hydroxylations at positions 2α, 1β, and 16β, followed by conjugation. Most of the metabolites that circulate in the blood are sulfates of 3α, 5β-tetrahydro-levonorgestrel, while excretion occurs predominantly in the form of glucuronides. Some of the parent levonorgestrel also circulates as the 17β-sulfate. In-vitro studies on the biotransformation of levonorgestrel in human skin did not indicate any significant metabolism of levonorgestrel during skin penetration.

Distribution

The distribution of exogenous estrogens is similar to that of endogenous estrogens. Estrogens are widely distributed in the body and are generally found in higher concentrations in the sex hormone target organs. Estrogens circulate in the blood largely bound to SHBG and albumin.

Levonorgestrel in serum is bound to both SHBG and albumin. Following four consecutive weekly applications of Climara Pro mean (± SD) SHBG concentrations declined from a predose value of 47.5 (25.8) to 41.2 (22.4) nmol/L at week 4.

Clinical Studies

Effects on Vasomotor Symptoms

The efficacy of 0.045 mg estradiol/0.03 mg levonorgestrel administered weekly versus placebo in the relief of moderate to severe vasomotor symptoms in postmenopausal women was studied in one 12-week clinical trial (n=183, average age 52.1 ± 4.93, 82 percent Caucasian). The 0.045 mg estradiol/0.03 mg levonorgestrel dosage strength was shown to be statistically better than placebo at weeks 4 and 12 for relief of both the number and severity of moderate to severe hot flushes. See Tables 3 and 4. Climara Pro and the 0.045 mg estradiol/0.03 mg levonorgestrel dosage strength are bioequivalent in terms of estradiol delivery [See Clinical Pharmacology (12.3)].

Table 3: Summary of Mean Daily Number of Moderate to Severe Hot Flushes-ITT*
* Intent-to-Treat population † A subject was included at baseline only if the subject had a post-baseline mean score. The post-baseline mean score required 3 days in one week. ‡ n = Number of subjects in a treatment group in a cycle; number of subjects varied from cycle to cycle due to missing data. § SD = standard deviation ¶ p-value for comparison to placebo, adjusted by the method of Bonferroni # p <0.025

Baseline†

Week 4

Week 8

Week 12

Placebo

n‡

88

82

73

69

Mean (SD)§

10.8

(5.803)

6.13

(4.311)

5.35

(4.095)

5.59

(4.93)

Mean Change

from baseline (SD)

NA

-4.23

(4.374)

-4.8

(4.448)

-4.55

(5.407)

0.045/.03

n‡

92

88

80

73

Mean (SD)§

10.13

(3.945)

2.69

(4.455)

1.22

(2.804)

1.06

(3.187)

Mean Change

from baseline (SD)§

NA

-7.4

(4.715)

-8.68

(4.146)

-8.82

(4.336)

p-Value ¶

NA

<0.001#

NA

<0.001#

Table 4: Summary of Mean Severity of Moderate to Severe Hot Flushes-ITT*
* ITT= Intent-to-Treat population † A subject was included at baseline only if the subject had at least 1 post-baseline value. ‡ SD= standard deviation Severity scores are: 1 = Mild, 2 = Moderate, 3 = Severe. Mean severity of hot flushes by day is [(2X number of moderate hot flushes) + (3X number of severe hot flushes)] / total number of moderate to severe hot flushes on that day. If no moderate to severe hot flush was indicated, the mean severity was 0. § p-value for comparison to placebo, adjusted by the method of Bonferroni ¶ p <0.025

Baseline†

Week 4

(day 7)

Week 8

(day 7)

Week 12

(day 7)

Placebo

n‡

89

76

68

57

Mean (SD)d

2.42

(0.282)

1.99

(0.875)

1.93

(0.955)

1.8

(1.034)

Mean Change

from baseline

(SD)‡

NA

-0.4

(0.865)

-0.48

(0.922)

-0.57

(1.044)

0.045/.03

n‡

92

83

72

55

Mean (SD)‡

2.48

(0.295)

1.1

(1.191)

0.82

(1.226)

0.44

(0.96)

Mean Change

from baseline

(SD)‡

NA

-1.4

(1.164)

-1.67

(1.245)

-2.06

(1.005)

p-value§

NA

<0.001¶

NA

<0.001¶

Effects on the Endometrium

In a 1-year clinical trial of 412 postmenopausal women (with intact uteri) treated with a continuous regimen of Climara Pro or with an continuous estradiol-only transdermal system, results of evaluable endometrial biopsies show that no hyperplasia was seen with Climara Pro. Table 5 below summarizes these results (Intent-to-Treat populations).

Table 5: Incidence of Endometrial Hyperplasia during Continuous Combined Treatment with Climara Pro, ITT*
* ITT = Intent-to-Treat population † n = number of intent-to-treat subjects. ‡ Defined as at least 180 days of treatment. § Defined as ≥ 323 days of treatment. ¶ Includes hyperplasia occurring at any time after initiation of treatment as a proportion of patients with biopsies at 1 year. # p < 0.0167 p-value for comparison to unopposed estradiol dose using the Fisher Exact test. P-values were adjusted by the method of Bonferroni.

Climara Pro

E2 0.045 mg / LNG 0.015 mg

Estradiol

E2 0.045 mg

n† = 210

na = 202

No. of Patients with Biopsies at >6 months‡

124

139

No. of Patients with Biopsies at 1 year§

102

110

No. (%) of Patients with Hyperplasia¶

0 (0%)#

19 (17.3%)

95% Confidence Interval

0-3.55%

9.75–24.79%

Effects on Uterine Bleeding or Spotting

The effects of Climara Pro on uterine bleeding or spotting, as recorded using an interactive voice response system, were evaluated in one 12-month clinical trial. Results are shown in Figure 3.

Figure 3: Cumulative Proportion of Subjects at Each Cycle with No Bleeding/Spotting Through the End of Cycle 13 Last Observation Carried Forward

• Percent based upon the number of subjects with data • Last non-missing cycle carried forward through cycle 13 • Bleeding associated with endometrial biopsies not included

Effects on Bone Mineral Density

The effects on bone mineral density (BMD) were studied in a randomized, double-blind, placebo-controlled clinical trial of transdermal systems (patches) containing only estradiol (E2). The patients were postmenopausal women with hysterectomies, 40–83 years of age (mean=51.4 years), and 77.3 percent Caucasian. Patients received calcium supplements if they appeared deficient on a questionnaire. Vitamin D supplements were not given.

A total of 154 patients were randomized in a 2:2:3 ratio to weekly application of 22 cm2 patches containing 2.2 mg E2, 4.4 mg E2, or placebo, for 728 days of continuous treatment (26 28-day cycles). Only the results for the estradiol dose in Climara Pro (4.4 mg E2) and for placebo are presented.

Statistically significant increases in the primary efficacy variable, BMD of the lumbar spine (A-P view, L2-L4), were seen for 4.4 mg E2 compared to placebo (see Table 5 and Figure 4). BMD was also measured at the hip (total, non-dominant side) and radius (midshaft, non-dominant side) with statistically significant treatment effects only observed for the hip (see Table 6).

Table 6: Mean Bone Mineral Density (Standard Deviation)*
* Intent-to-treat population with on-treatment efficacy data † E2=estradiol; LOCF= Last Observation Carried Forward

4.4 mg E2†

Placebo

Total Lumbar Spine

Baseline (g/cm2)

n=36

1.1 (0.2)

n=46

1.1 (0.2)

% Change from baseline LOCF

+1.7% (4.4)

-2.9% (3.8)

P-value compared to placebo

<0.0001

Total Hip

Baseline (g/cm2)

n=36

0.97 (0.1)

n=48

0.94 (0.1)

% Change from baseline LOCF

+1.3% (4.2)

-0.9% (5.2)

P-value compared to placebo

0.05

Figure 4: Percent Change From Baseline in Bone Mineral Density (g/cm2) of Lumbar Spine (A-P View, L2–L4) by Treatment Group and Cycle (Mean ± SE)*

* Data in the figure is for 21 patients on 4.4 mg E2 and 27 placebo patients who completed the study; approximately 44 percent of randomized patients.

Women's Health Initiative Studies

The WHI enrolled approximately 27,000 predominantly healthy postmenopausal women in two substudies to assess the risks and benefits of daily oral CE (0.625 mg)-alone or in combination with MPA (2.5 mg) compared to placebo in the prevention of certain chronic diseases. The primary endpoint was the incidence of CHD (defined as nonfatal MI, silent MI and CHD death), with invasive breast cancer as the primary adverse outcome. A "global index" included the earliest occurrence of CHD, invasive breast cancer, stroke, PE, endometrial cancer (only in the CE plus MPA substudy), colorectal cancer, hip fracture, or death due to other cause. These substudies did not evaluate the effects of CE plus MPA or CE-alone on menopausal symptoms.

WHI Estrogen Plus Progestin Substudy

The WHI estrogen plus progestin substudy was stopped early. According to the predefined stopping rule, after an average follow-up of 5.6 years of treatment, the increased risk of invasive breast cancer and cardiovascular events exceeded the specified benefits included in the "global index". The absolute excess risk of events included in the “global index” was 19 per 10,000 women-years.

For those outcomes included in the WHI “global index” that reached statistical significance after 5.6 years of follow-up, the absolute excess risks per 10,000 women-years in the group treated with CE plus MPA were 7 more CHD events, 8 more strokes, 10 more PEs, and 8 more invasive breast cancers, while the absolute risk reductions per 10,000 women-years were 6 fewer colorectal cancers and 5 fewer hip fractures.

Results of the CE plus MPA substudy, which included 16,608 women (average 63 years of age, range 50 to 79; 83.9 percent White, 6.8 percent Black, 5.4 percent Hispanic, 3.9 percent Other) are presented in Table 7. These results reflect centrally adjudicated data after an average follow-up of 5.6 years.

Table 7: Relative and Absolute Risk Seen in the Estrogen Plus Progestin Substudy of WHI at an Average of 5.6 Years*†
* Adapted from numerous WHI publications. WHI publications can be viewed at www.nhlbi.nih.gov/whi. † Results are based on centrally adjudicated data. ‡ Nominal confidence intervals unadjusted for multiple looks and multiple comparisons § Not included in “global index.” ¶ Includes metastatic and non-metastatic breast cancer, with the exception of in situ breast cancer. # All deaths, except from breast or colorectal cancer, definite or probable CHD, PE or cerebrovascular disease. Þ A subset of the events was combined in a "global index", defined as the earliest occurrence of CHD events, invasive breast cancer, stroke, pulmonary embolism, colorectal cancer, hip fracture, or death due to other causes.

Event

Relative Risk
CE/MPA vs. Placebo
(95% nCI‡)

CE/MPA
n = 8,506 *

Placebo

n = 8,102

Absolute Risk per 10,000 Women-Years

CHD events
Non-fatal MI
CHD death

1.23 (0.99-1.53)
1.28 (1.00-1.63)
1.10 (0.70-1.75)

41

34

31

25

8

8

All strokes

1.31 (1.03-1.68)

33

25

Ischemic stroke

1.44 (1.09-1.90)

26

18

Deep vein thrombosis§

1.95 (1.43-2.67)

26

13

Pulmonary embolism

2.13 (1.45-3.11)

18

8

Invasive breast cancer¶

1.24 (1.01-1.54)

41

33

Colorectal cancer

0.61 (0.42-0.87)

10

16

Endometrial cancer§

0.81 (0.48-1.36)

6

7

Cervical cancer§

1.44 (0.47-4.42)

2

1

Hip fracture‡

0.67 (0.47-0.96)

11

16

Vertebral fractures§

0.65 (0.46-0.92)

11

17

Lower arm/wrist fractures§

0.71 (0.59-0.85)

44

62

Total fractures§

0.76 (0.69-0.83)

152

199

Overall Mortality#

1.00 (0.83-1.19)

52

52

Global IndexÞ

1.13 (1.02-1.25)

184

165

Timing of the initiation of estrogen plus progestin therapy relative to the start of menopause may affect the overall risk benefit profile. The WHI estrogen plus progestin substudy stratified by age showed in women 50 to 59 years of age, a non-significant trend toward reduced risk for overall mortality [hazard ratio (HR) 0.69 (95 percent CI, 0.44–1.07)].

WHI Estrogen-Alone Substudy

The WHI estrogen-alone substudy was stopped early because an increased risk of stroke was observed, and it was deemed that no further information would be obtained regarding the risks and benefits of estrogen-alone in predetermined primary endpoints.

Results of the estrogen-alone substudy, which included 10,739 women (average age of 63 years, range 50 to 79; 75.3 percent White, 15.1 percent Black, 6.1 percent Hispanic, 3.6 percent Other), after an average follow-up of 7.1 years are presented in Table 8.

Table 8: Relative and Absolute Risk Seen in the Estrogen-Alone Substudy of WHI*
* Nominal confidence intervals unadjusted for multiple looks and multiple comparisons. † Results are based on centrally adjudicated data for an average follow-up of 7.1 years. ‡ Not included in “global index”. § Results are based on an average follow-up of 6.8 years. ¶ All deaths, except from breast or colorectal cancer, definite/probable CHD, PE or cerebrovascular disease. # A subset of the events was combined in a "global index", defined as the earliest occurrence of CHD events, invasive breast cancer, stroke, pulmonary embolism, colorectal cancer, hip fracture, or death due to other causes.

Event

Relative Risk
CE vs. Placebo
(95% nCI*)

CE
n = 5,310

Placebo
n = 5,429

Absolute Risk per 10,000 Women-Years

CHD events†

0.95 (0.78-1.16)

54

57

  Non-fatal MI†

0.91 (0.73-1.14)

40

43

  CHD death†

1.01 (0.71-1.43)

16

16

All strokes†

1.33 (1.05-1.68)

45

33

Ischemic stroke†

1.55 (1.19-2.01)

38

25

Deep vein thrombosis†, ‡

1.47 (1.06-2.06)

23

15

Pulmonary embolism†

1.37 (0.90-2.07)

14

10

Invasive breast cancer†

0.80 (0.62-1.04)

28

34

Colorectal cancer†

1.08 (0.75-1.55)

17

16

Hip fracture†

0.65 (0.45-0.94)

12

19

Vertebral fractures†,‡

0.64 (0.44-0.93)

11

18

Lower arm/wrist fractures†, ‡

0.58 (0.47-0.72)

35

59

Total fractures†, ‡

0.71 (0.64-0.80)

144

197

Death due to other causes§, ¶

1.08 (0.88-1.32)

53

50

Overall Mortality†, ‡

1.04 (0.88-1.22)

79

75

Global Index#

1.02 (0.92-1.13)

206

201

For those outcomes included in the WHI "global index" that reached statistical significance, the absolute excess risk per 10,000 women-years in the group treated with CE- alone was 12 more strokes, while the absolute risk reduction per 10,000 women-years was 7 fewer hip fractures.9 The absolute excess risk of events included in the "global index" was a non-significant 5 events per 10,000 women-years. There was no difference between the groups in terms of all-cause mortality.

No overall difference for primary CHD events (nonfatal MI, silent MI and CHD death) and invasive breast cancer incidence in women receiving CE-alone compared with placebo was reported in final centrally adjudicated results from the estrogen-alone substudy, after an average follow-up of 7.1 years.

Centrally adjudicated results for stroke events from the estrogen-alone substudy, after an average follow-up of 7.1 years, reported no significant difference in distribution of stroke subtype or severity, including fatal strokes, in women receiving CE-alone compared to placebo. Estrogen-alone increased the risk for ischemic stroke, and this excess risk was present in all subgroups of women examined.10

Timing of the initiation of estrogen-alone therapy relative to the start of menopause may affect the overall risk benefit profile. The WHI estrogen-alone substudy stratified by age showed in women 50 to 59 years of age, a non-significant trend toward reduced risk for CHD [HR 0.63 (95 percent CI, 0.36-1.09)] and overall mortality [HR 0.71 (95 percent CI, 0.46-1.11)].

Women's Health Initiative Memory Study

The WHIMS estrogen plus progestin ancillary study of WHI enrolled 4,532 predominantly healthy postmenopausal women 65 to 79 years of age (47 percent were 65 to 69 years of age, 35 percent were 70 to 74 years of age, and 18 percent were 75 years of age and older) to evaluate the effects of daily CE (0.625 mg) plus MPA (2.5 mg) on the incidence of probable dementia (primary outcome) compared to placebo.

After an average follow-up of 4 years, the relative risk of probable dementia for CE plus MPA versus placebo was 2.05 (95 percent CI, 1.21-3.48). The absolute risk of probable dementia for CE plus MPA was 45 versus 22 cases per 10,000 women-years. Probable dementia as defined in this study included Alzheimer’s disease (AD), vascular dementia (VaD) and mixed type (having features of both AD and VaD). The most common classification of probable dementia in the treatment group and the placebo group was AD. Since the ancillary study was conducted in women 65 to 79 years of age, it is unknown whether these findings apply to younger postmenopausal women [see Warnings and Precautions (5.3), and Use in Specific Populations (8.5)].

The WHIMS estrogen-alone ancillary study of WHI enrolled 2,947 predominantly healthy hysterectomized postmenopausal women 65 years of age and older (45 percent were age 65 to 69 years of age, 36 percent were 70 to 74 years of age, and 19 percent were 75 years of age and older) to evaluate the effects of daily CE (0.625 mg)-alone on the incidence of probable dementia (primary outcome) compared to placebo.

After an average follow-up of 5.2 years, the relative risk of probable dementia for CE-alone versus placebo was 1.49 (95 percent CI, 0.83-2.66). The absolute risk of probable dementia for CE-alone versus placebo was 37 versus 25 cases per 10,000 women-years. Probable dementia as defined in this study included Alzheimer’s disease (AD), vascular dementia (VaD) and mixed type (having features of both AD and VaD). The most common classification of probable dementia in the treatment group and the placebo groups was AD. Since the ancillary study was conducted in women 65 to 79 years of age, it is unknown whether these findings apply to younger postmenopausal women [see Warnings and Precautions (5.3), and Use in Specific Populations (8.5)].

When data from the two populations were pooled as planned in the WHIMS protocol, the reported overall relative risk for probable dementia was 1.76 (95 percent CI, 1.19-2.60). Differences between groups became apparent in the first year of treatment. It is unknown whether these findings apply to younger postmenopausal women [see Warnings and Precautions (5.3), and Use in Specific Populations (8.5)].

Package/label principal display panel

Climara Pro 
estradiol and levonorgestrel patch
Product Information
Product Type HUMAN PRESCRIPTION DRUG LABEL Item Code (Source) NDC:50419-491
Route of Administration TRANSDERMAL DEA Schedule     
Active Ingredient/Active Moiety
Ingredient Name Basis of Strength Strength
ESTRADIOL (ESTRADIOL) ESTRADIOL 0.045 mg  in 1 d
LEVONORGESTREL (LEVONORGESTREL) LEVONORGESTREL 0.015 mg  in 1 d
Packaging
# Item Code Package Description
1 NDC:50419-491-04 4 PATCH in 1 CARTON
1 7 d in 1 PATCH
2 NDC:50419-491-73 1 PATCH in 1 CARTON
2 7 d in 1 PATCH
Marketing Information
Marketing Category Application Number or Monograph Citation Marketing Start Date Marketing End Date
NDA NDA021258 11/21/2003
Labeler - Bayer HealthCare Pharmaceuticals Inc. (005436809)
Establishment
Name Address ID/FEI Operations
3M Pharmaceuticals 128688199 MANUFACTURE(50419-491)
Revised: 08/2017   Bayer HealthCare Pharmaceuticals Inc.
(web3)