Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Intern Med ; 254(1): 85-94, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12823645

ABSTRACT

OBJECTIVES: To study the long-term effects of raloxifene, a potential designer oestrogen, and oestrogen monotherapy on endothelial function in healthy postmenopausal women. DESIGN: A 2-year double-blind, randomized and placebo-controlled study in an Academic Medical Center. Fifty-six hysterectomized but otherwise healthy postmenopausal women randomly received raloxifene hydrochloride 60 mg day-1 (n = 15) or 150 mg day-1 (n = 13), conjugated equine oestrogen (CEE) 0.625 mg day-1 (n = 15), or placebo (n = 13). MAIN OUTCOME MEASURES: Endothelial function as estimated from brachial artery flow-mediated, endothelium-dependent vasodilation and nitroglycerine-induced endothelium-independent vasodilation, and plasma levels of the endothelium-derived regulatory proteins, von Willebrand factor (vWF) and endothelin (ET). RESULTS: Raloxifene 60 mg did not significantly affect endothelial function. As compared with placebo, at 6 months of therapy, raloxifene 150 mg and CEE were associated with a mean increase in vWF of 25.5% point (95% CI 3.6-47.3) and 26.6% point (95% CI 6.9-46.3), respectively. At 24 months of therapy, raloxifene 150 mg was associated with a mean decrease in ET of 0.96 pg mL-1 (95% CI -1.57 to -0.36). Raloxifene nor CEE significantly affected endothelium-dependent and/or -independent vasodilation. CONCLUSIONS: Our results suggest that long-term therapy with raloxifene or oral CEE does not affect endothelium-dependent vasodilation in healthy postmenopausal women. Raloxifene 150 mg day-1 might have both positive and negative effects on endothelium. The clinical significance of these findings remains to be investigated.


Subject(s)
Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Vascular Diseases/drug therapy , Analysis of Variance , Brachial Artery , Designer Drugs/administration & dosage , Designer Drugs/pharmacology , Double-Blind Method , E-Selectin/blood , Endothelium, Vascular/drug effects , Female , Humans , Middle Aged , Postmenopause , Raloxifene Hydrochloride/administration & dosage , Selective Estrogen Receptor Modulators/administration & dosage , Vascular Diseases/physiopathology , Vasodilation/drug effects , von Willebrand Factor/analysis
2.
Thromb Haemost ; 84(6): 968-72, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11154142

ABSTRACT

Estrogen therapy may increase the risk of arterial thromboembolism, at least in the short term. In a randomized, double-blind and placebo-controlled study in 25 healthy postmenopausal women (52.5 +/- 2.8 years), we therefore examined the short-term effect of unopposed estrogen on the fasting and fat-load-stimulated plasma levels of total factor VII versus active factor VII. Plasma total factor VII was measured by use of a chromogenic assay; plasma active FVII by a recently developed method using truncated tissue factor. As compared to placebo, 8 weeks of oral 17beta-estradiol (2 mg daily) increased the mean fasting and postprandial plasma levels of total factor VII by 17 and 21% points, respectively (both P < 0.01 ), but did not affect the fasting and/or postprandial plasma levels of active factor VII (mean change both 0.05 ng/mL; P > 0.35). Furthermore, the change in the fasting level of total factor VII after therapy was not associated with the change in the fasting level of active factor VII (r = 0.27; P = 0.21). These findings argue against the idea that elevated levels of total factor VII underlie an increased risk of arterial thromboembolism in postmenopausal women using unopposed estrogen replacement.


Subject(s)
Estrogens/pharmacology , Factor VII/drug effects , Factor VIIa/drug effects , Double-Blind Method , Estrogens/administration & dosage , Factor VII/metabolism , Factor VIIa/metabolism , Fasting , Female , Hormone Replacement Therapy , Humans , Middle Aged , Postmenopause , Postprandial Period , Prospective Studies , White People
3.
Arterioscler Thromb Vasc Biol ; 19(12): 2993-3000, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10591680

ABSTRACT

Currently raloxifene, a selective estrogen receptor modulator, is being investigated as a potential alternative for postmenopausal hormone replacement to prevent osteoporosis and cardiovascular disease. We compared the 2-year effects of raloxifene on a wide range of cardiovascular risk factors with those of placebo and conjugated equine estrogens (CEEs). Analyses were based on 56 hysterectomized but otherwise healthy postmenopausal women aged 54. 8+/-3.5 (mean+/-SD) years who entered this double-blind study and who were randomly assigned to raloxifene hydrochloride 60 mg/d (n=15) or 150 mg/d (n=13), placebo (n=13), or CEEs 0.625 mg/d (n=15). At baseline and after 6, 12, and 24 months of treatment, we assessed serum lipids, blood pressure, glucose metabolism, C-reactive protein, and various hemostatic parameters. Compared with placebo, both raloxifene and CEEs lowered the level of low density lipoprotein cholesterol by 0.53 to 0.79 mmol/L (all P<0.04) and lowered, at 24 months, the level of fibrinogen by 0.71 to 0.86 g/L (all P<0.05). The effects of raloxifene and CEEs did not differ significantly. In contrast to raloxifene, from 6 months on CEEs increased high density lipoprotein cholesterol by 0.25 to 0.29 mmol/L and reduced plasminogen activator inhibitor-1 antigen by 30.6 to 48.6 ng/mL (all P<0.02 versus both placebo and raloxifene). CEEs transiently increased C-reactive protein by 1.0 mg/L at 6 months (P<0.05 versus placebo) and prothrombin-derived fragment F1+2 by 0. 79 nmol/L at 12 months (P<0.001 versus placebo). Finally, from 12 months on, CEEs increased triglycerides by 0.33 to 0.56 mmol/L (all P<0.05 versus both placebo and raloxifene). Our findings suggest that in healthy postmenopausal women, raloxifene and estrogen monotherapy have similar beneficial effects on low density lipoprotein cholesterol and fibrinogen levels. These treatments differ, however, in their effects on high density lipoprotein cholesterol, triglycerides, and plasminogen activator inhibitor-1 and possibly in their effects on prothrombin fragment F1+2 and C-reactive protein.


Subject(s)
Coronary Disease/prevention & control , Estrogen Antagonists/administration & dosage , Estrogens/administration & dosage , Hormone Replacement Therapy , Postmenopause , Raloxifene Hydrochloride/administration & dosage , Biomarkers , Blood Coagulation/drug effects , Blood Pressure , C-Reactive Protein/metabolism , Carbohydrate Metabolism , Coronary Disease/drug therapy , Coronary Disease/epidemiology , Double-Blind Method , Female , Fibrinolysis/drug effects , Humans , Hysterectomy , Middle Aged , Placebos , Risk Factors
4.
Clin Endocrinol (Oxf) ; 50(3): 387-92, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10435066

ABSTRACT

OBJECTIVE: Impaired intestinal calcium absorption in postmenopausal women is often indirectly linked to decreased serum 1,25(OH)2D or to intestinal resistance to its action rather than directly to low circulating oestrogen levels following the menopause. The purpose of this clinical study was to investigate the short-term effect of oral 17 beta-oestradiol on intestinal calcium absorption, with strontium as a marker. DESIGN AND PATIENTS: Twenty-five healthy postmenopausal women participated in this randomised double blind placebo controlled clinical trial. Twelve women received oestradiol therapy (2 mg/day) and thirteen placebo for 2 months. Fractional strontium absorption (Fc240) was assessed at baseline and after 2 months of oestradiol/placebo therapy. RESULTS: Intestinal strontium absorption (Fc240) was unchanged after treatment with 17 beta-oestradiol (10.1 +/- 5.0 vs. 10.2 +/- 3.8(%)). Serum total calcitriol (1,25(OH)2D) was unchanged after treatment with placebo (88 +/- 22 vs. 79 +/- 21 (pmol/l)) but increased after treatment with oestradiol (88 +/- 30 vs. 116 +/- 33 (pmol/l); P < 0.005). Serum vitamin D binding protein (DBP) increased after oestradiol but not after placebo treatment. The free serum 1,25(OH)2D index was calculated. This index did not change after oestrogen therapy (1.6 +/- 0.5 vs. 1.8 +/- 0.5). CONCLUSION: In healthy postmenopausal women, short-term suppletion with exogenous oral oestrogen did not influence intestinal calcium absorption as measured by the strontium absorption test.


Subject(s)
Calcium/pharmacokinetics , Estradiol/administration & dosage , Intestinal Absorption/drug effects , Postmenopause/blood , Strontium/pharmacokinetics , Administration, Oral , Biomarkers/blood , Calcitriol/blood , Double-Blind Method , Estradiol/pharmacology , Estrogen Replacement Therapy , Female , Humans , Middle Aged , Vitamin D-Binding Protein/blood
5.
Clin Chem ; 45(8 Pt 1): 1200-5, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10430785

ABSTRACT

BACKGROUND: Hyperhomocysteinemia is an independent cardiovascular risk factor, possibly through the induction of endothelial dysfunction. The postmenopausal state is associated with increased plasma homocysteine. We examined whether increased homocysteine is associated with impaired endothelial function. METHODS: Sixty-three hysterectomized but otherwise healthy postmenopausal women (54.8 +/- 3.5 years) participated in this study. Fasting total plasma homocysteine (tHcy) was measured as free plus protein-bound homocysteine. Endothelial function was assessed by measuring plasma concentrations of the endothelium-derived proteins endothelin (ET), von Willebrand factor (vWF), and plasminogen activator inhibitor type 1 (PAI-1) as well as brachial artery flow-mediated, endothelium-dependent vasodilatation (FMD). RESULTS: Plasma tHcy was 9.6 +/- 2.5 micromol/L. After adjustment for possible confounders, a 1 micromol/L increase in tHcy was associated with an increase in ET of 0.08 ng/L (P = 0.045) and an increase in vWF of 4.2% (P = 0.05). No statistically significant association was present between tHcy and PAI-1 or FMD. CONCLUSIONS: Increased fasting homocysteine in postmenopausal women may impair some aspects of endothelial function. It is of clinical interest to study whether homocysteine lowering can improve endothelial function and thus cardiovascular morbidity and mortality in postmenopausal women.


Subject(s)
Endothelins/blood , Endothelium, Vascular/physiology , Homocysteine/blood , Postmenopause/blood , Vasodilation/physiology , von Willebrand Factor/metabolism , Adult , Brachial Artery/physiology , Female , Humans , Hysterectomy , Middle Aged , Muscle, Smooth, Vascular/physiology , Plasminogen Activator Inhibitor 1/blood
6.
Menopause ; 6(2): 134-7, 1999.
Article in English | MEDLINE | ID: mdl-10374220

ABSTRACT

OBJECTIVE: Long-term postmenopausal estrogen replacement therapy lowers the risk of osteoporotic fractures and coronary artery disease but increases the risk of endometrial cancer and probably breast cancer. Raloxifene, a nonsteroidal estrogen receptor ligand, seems to have a tissue-specific antiestrogenic action on endometrium and breast and the desired estrogenic action on bone and lipid metabolism. The purpose of this study was to investigate the effects of 24-month treatment with orally administered raloxifene in two doses (60 mg and 150 mg daily) and conjugated equine estrogens in a standard oral dose (0.625 mg daily) on serum lipoprotein(a) [Lp(a)], an independent risk factor for coronary artery disease, in healthy postmenopausal women who had undergone hysterectomy. DESIGN: A randomized, double-blind, placebo-controlled study was performed with 56 women. RESULTS: In the placebo group serum Lp(a) levels did not change throughout the study. After 6 months, serum Lp(a) was significantly reduced versus baseline in the raloxifene 150 (-17%; p = 0.003) and conjugated equine estrogens (-26%; p = 0.003) groups, but this reduction was significantly different from placebo only in the conjugated equine estrogens group. At 12 and 24 months, serum Lp(a) levels were significantly lowered versus baseline in all active treatment groups. However, these reductions were significantly different from placebo only in the raloxifene 150 and conjugated equine estrogens groups. After 24 months, serum Lp(a) was reduced versus baseline with 30% (p = 0.001) in the raloxifene 150 group and 35% (p = 0.001) in the conjugated equine estrogens group. CONCLUSIONS: Long term raloxifene treatment significantly lowers serum Lp(a) levels in postmenopausal women and thus might reduce the risk of coronary artery disease.


Subject(s)
Estrogen Antagonists/administration & dosage , Estrogen Replacement Therapy/methods , Estrogens, Conjugated (USP)/therapeutic use , Lipoprotein(a)/blood , Piperidines/administration & dosage , Postmenopause/drug effects , Administration, Oral , Analysis of Variance , Cardiovascular Diseases/prevention & control , Double-Blind Method , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Hysterectomy , Middle Aged , Postmenopause/blood , Raloxifene Hydrochloride , Treatment Outcome
7.
Ann N Y Acad Sci ; 873: 99-104, 1999 Apr 20.
Article in English | MEDLINE | ID: mdl-10372156

ABSTRACT

An approach to determine intra- and extracellular conduction on the basis of Bode analysis is presented. Estimation of the ratio between intra- and extracellular conduction could be performed by phase measurement only, midrange in the bandwidth of interest. An important feature is that the relation between intra- and extracellular conduction can be continuously monitored by phase measurement and no curve fitting whatsoever is required. Based on a two-frequency measurement determining Re at 4 kHz and phi max at 64 kHz, it proved possible to estimate extracellular volume (ECV) in 23 patients. Reference values on ECV were determined by sodium bromide. The results show a good correlation (r = 0.90) with the reference method. The average error of ECV estimation was -3.6% (SD 8.4).


Subject(s)
Body Composition , Extracellular Space/physiology , Calcium/metabolism , Electric Conductivity , Electric Impedance , Estradiol/pharmacology , Female , Humans , Intestinal Absorption/physiology , Models, Biological , Postmenopause
8.
Fertil Steril ; 70(6): 1085-9, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9848300

ABSTRACT

OBJECTIVE: To investigate the long-term effects of raloxifene on fasting plasma homocysteine levels in postmenopausal women compared with conjugated equine estrogen (CEE). DESIGN: Randomized, double-blind, placebo-controlled study. SETTING: Outpatient department of a university hospital. PATIENT(S): Fifty-two hysterectomized, healthy postmenopausal women. INTERVENTION(S): Oral raloxifene in two dosages (60 mg/d [n=13] and 150 mg/d [n=13]), oral CEE (0.625 mg/d [n=13], and placebo (n=13) were given for 24 months. MAIN OUTCOME MEASURE(S): Fasting plasma homocysteine concentrations. RESULT(S): Plasma homocysteine levels were not altered in the placebo group. After 12 months, a significant reduction versus baseline in the mean plasma homocysteine level (-16%) was found only in the raloxifene 150-mg group. The mean change in plasma homocysteine levels within this group also was significantly different from the changes versus baseline found in the placebo group (+2%) and the raloxifene 60-mg group (-2%), but not different from those found in the CEE group (-8%). After 24 months, plasma homocysteine levels were decreased significantly in the raloxifene 150-mg and CEE groups compared with both baseline (-13% and -10%, respectively) and placebo values (-15% and -11%, respectively). No significant change in plasma homocysteine levels was observed in the raloxifene 60-mg group. CONCLUSION(S): Raloxifene has a favorable, dose-related effect on plasma homocysteine levels in postmenopausal women.


Subject(s)
Estrogen Replacement Therapy , Estrogens, Conjugated (USP)/therapeutic use , Homocysteine/blood , Piperidines/therapeutic use , Postmenopause/blood , Receptors, Estrogen/drug effects , Administration, Oral , Animals , Double-Blind Method , Fasting/blood , Female , Horses , Humans , Hysterectomy , Middle Aged , Placebos , Raloxifene Hydrochloride , Reference Values
9.
Physiol Meas ; 19(4): 517-26, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9863677

ABSTRACT

In order to determine body fluid shifts between the intra- and extra-cellular spaces, multifrequency impedance measurement is performed. According to the Cole-Cole extrapolation, lumped values of intra- and extra-cellular conduction can be estimated which are commonly expressed in resistances Ri and Re respectively. For this purpose the magnitude and phase of the impedance under study are determined at a number of frequencies in the range between 5 kHz and 1 MHz. An approach to determine intra- and extra-cellular conduction on the basis of Bode analysis is presented in this article. On this basis, estimation of the ratio between intra- and extra-cellular conduction could be performed by phase measurement only, midrange in the bandwidth of interest. An important feature is that the relation between intra- and extra-cellular conduction can be continuously monitored by phase measurement and no curve fitting whatsoever is required. Based on a two frequency measurement determining Re at 4 kHz and phi(max) at 64 kHz it proved possible to estimate extra-cellular volume (ECV) more accurately compared with the estimation based on extrapolation according to the Cole-Cole model in 26 patients. Reference values of ECV were determined by sodium bromide. The results show a correlation of 0.90 with the reference method. The average error of ECV estimation was -3.6% (SD 8.4), whereas the Cole-Cole extrapolation showed an error of 13.2% (SD 9.5). An important feature of the proposed approach is that the relation between intra- and extra-cellular conduction can be continuously monitored by phase measurement and no curve fitting whatsoever is required.


Subject(s)
Electric Impedance , Extracellular Space/physiology , Intracellular Fluid/physiology , Humans , Models, Biological , Plethysmography, Impedance , Reference Values
10.
Maturitas ; 28(2): 153-62, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9522323

ABSTRACT

In this study we examined whether the effect of continuously combined hormone replacement therapy (HRT) on bone metabolism is influenced by dydrogesterone dose, smoking and initial degree of bone turnover. In a double-blind randomized study, 123 healthy postmenopausal women (mean age 51.7 years; range 30-61 years) received 17 beta-estradiol, 2 mg orally per day, continuously combined with either 2.5, 5, 10 or 15 mg of dydrogesterone daily. At baseline and at 3 and 6 months of therapy, bone formation was assessed by determining total alkaline phosphatase (TAP), bone-derived alkaline phosphatase (BAP), and the carboxy-terminal propeptide of collagen type I (PICP) in serum; bone resorption was assessed by the calcium/creatinine (Ca/Creat) and hydroxyproline/creatinine (Hp/Creat) ratio in 2-h fasting urine, and the serum carboxy-terminal pyridinolyne cross-linked telopeptide of collagen type I (ICTP). Dydrogesterone dose did not influence the effect of HRT on any of the bone markers. Combining the data of the four treatment groups, the decrease in each marker, compared to baseline values, was significant. However, in non-smokers, compared to smokers, after 6 months of therapy the decline in BAP and TAP was significantly more pronounced and the plasma estradiol level was significantly higher. For each bonemarker at baseline, women in the highest quartile, compared to women in the lowest quartile, showed a significantly stronger decrease in this marker in response to HRT. We conclude that dydrogesterone dose does not modify the effectiveness of replacement therapy. However, smoking and a low bone turnover at baseline may diminish its beneficial effect on bone.


Subject(s)
Dydrogesterone/therapeutic use , Estradiol/therapeutic use , Estrogen Replacement Therapy/methods , Postmenopause/drug effects , Progesterone Congeners/therapeutic use , Smoking/physiopathology , Adult , Alkaline Phosphatase/blood , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Biomarkers/blood , Biomarkers/urine , Calcium/metabolism , Calcium/urine , Cohort Studies , Collagen/blood , Collagen/drug effects , Collagen/metabolism , Collagen Type I , Creatinine/metabolism , Creatinine/urine , Dose-Response Relationship, Drug , Double-Blind Method , Dydrogesterone/administration & dosage , Estradiol/administration & dosage , Female , Humans , Hydroxyproline/drug effects , Hydroxyproline/metabolism , Hydroxyproline/urine , Middle Aged , Peptide Fragments/blood , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Peptides/blood , Peptides/drug effects , Peptides/metabolism , Postmenopause/blood , Postmenopause/urine , Procollagen/blood , Procollagen/drug effects , Procollagen/metabolism , Progesterone Congeners/administration & dosage , Smoking/blood , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...