当前位置: 首页 > 期刊 > 《癌症医师》 > 2006年第6期 > 正文
编号:11121682
Symptoms and Treatment in Cancer Therapy-Induced Early Menopause
http://www.100md.com 《中华首席医学网》
     a Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; b Division of Biomedical Analysis, Faculty of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands

    Key Words. Hot flashes ; Pathophysiology ; Epidemiology ; Complementary therapies ; Clonidine ; Gabapentin ; Selective serotonin reuptake inhibitors ; Breast neoplasm

    ABSTRACT

    Young women with breast cancer often experience early menopause as a result of the therapy for their malignant disease. The sudden occurrence of menopause resulting from chemotherapy, oophorectomy, radiation, or gonadal dysgenesis frequently results in hot flashes that begin at a younger age and may occur at a greater frequency and intensity than hot flashes associated with natural menopause.

    Hormone therapy relieves symptoms effectively in 80%–90% of women who initiate treatment. This therapy, however, is generally contraindicated in estrogen-dependent cancers, such as breast cancer, because of the potentially increased risk for recurrence. Many agents have been investigated as potential means for alleviating hot flashes in survivors of breast cancer, such as progestagens, clonidine, gabapentin, and anti-depressants. Several complementary and alternative medicines frequently used by patients have also been studied. These include black cohosh, phytoestrogens, homeopathy, vitamin E, acupuncture, and behavior strategies.

    To support the use of one of more of these nonpharmacological or pharmacological options in the treatment of hot flashes in breast cancer patients, more evidence from well-controlled clinical trials is needed. In particular, soundly based scientific research with complementary and alternative medicine therapies is lacking. Pharmacological treatments appear to be more beneficial than nonpharmacological treatments.

    This article reviews the current literature to assess the epidemiology and diagnosis of hot flashes and the nonpharmacological and pharmacological options for the treatment of hot flashes, in breast cancer patients in particular. When specific treatment options have not been evaluated in breast cancer patients specifically, published data on the management of hot flashes with this modality in healthy postmenopausal women are described.

    INTRODUCTION

    Breast cancer is the most common life-threatening cancer diagnosis in women. Although primary local control surgery is still the mainstay of treatment for early breast cancer, it often may not cure patients, as it does not eradicate micrometastases that are present in a subset of patients. In order to decrease the risk for local and distant relapse, the addition of adjuvant local radiation therapy and/or systemic hormonal and/or chemotherapy is necessary to increase the cure rate.

    However, the treatment of early or advanced breast cancer, with the aim to improve survival or palliation, may induce potentially severe short- and long-term toxicities. One disabling side effect of cancer treatment is premature menopause. The risk for the development of early menopause with polyagent adjuvant chemotherapy has been reported to be in the range of 53%–89% [1]. The frequency of chemotherapy-related amenorrhea varies with age, the cytotoxic agents used, and the cumulative dose [2, 3].

    In premenopausal women with endocrine-responsive tumors, the additional benefit of endocrine therapies after locoregional treatment and chemotherapy has been shown in the adjuvant setting of breast cancer. Endocrine therapy results in a significant improvement in both recurrence-free survival and overall survival in patients younger than 50 years of age [4, 5]. Endocrine therapies include suppression of ovarian function by irradiation, surgery, or luteinizing hormone-releasing hormone (LHRH) or gonadotropin hormone-releasing hormone (GnRH) agonists. It is an option to combine ovarian ablation with other types of endocrine therapies, including aromatase inhibitors and selective estrogen receptor downregulators [6].

    However, these therapies for breast cancer in premenopausal women increase the risk for early menopause, with the resulting loss of childbearing capacity and symptoms such as hot flashes, genitourinary atrophy, and psychological distress [2, 7, 8]. Hot flashes are one of the symptoms that occur with considerable frequency in premenopausal breast cancer patients. Hot flashes can interfere with quality of life and the quality or duration of sleep [9, 10].

    There is a wide range of possible disruptions in day-to-day living because of the impact of hot flashes. When hot flashes disrupt quality of life or the quality of sleep in breast cancer patients, these patients should be informed about therapeutic options, and interventions should be considered in the prevention and management of hot flashes.

    PATHOPHYSIOLOGY AND EPIDEMIOLOGY OF HOT FLASHES

    Definitions

    A hot flash is a subjective sensation of heat that is associated with objective signs of cutaneous vasodilation and a subsequent drop in core temperature. Sweating, flushing, palpitations, anxiety, irritability, and even panic may accompany the hot flash, and women may also report night sweats. The frequency, duration, and intensity of hot flashes vary. The duration of hot flashes can last a few seconds to several minutes and vary from mild to intolerable. Some women will have hot flashes several times a month, whereas other women complain that symptoms occur every hour [11, 12].

    "Hot flashes," "vasomotor symptoms," "hot flushes," "night sweats," and "climacteric symptoms" are all terms used to describe the same phenomenon. The term "hot flash" is used in this article.

    Pathophysiology

    The exact pathophysiological mechanisms of the occurrence of hot flashes are unknown. The theory of the cause of hot flashes is that there is a dysfunction in the central thermoregulatory set point in the hypothalamus as a result of decreased estrogen or decreased gonadal steroid levels. Studies suggest that estrogen withdrawal leads to an imbalance in plasma levels of several neurotransmitters. Norepinephrine is the primary neurotransmitter responsible for lowering the thermoregulatory set point. Plasma levels of norepinephrine metabolites are increased. The effect of higher norepinephrine and serotonin levels is to lower the thermoregulatory set point, which allows vasodilation and hot flash sensation.

    The neurotransmitter serotonin might also have an important role in thermoregulation. Decreased blood serotonin levels and upregulation of serotonin receptors in the hypothalamus are associated with estrogen withdrawal. The thermoregulatory set point might be dependent on the balance of these plasma levels, and a change in the balance may trigger the hot flash sensation [11, 13, 14].

    Prevalence of Hot Flashes

    Hot flashes affect two thirds of postmenopausal women, and 10%–20% of all postmenopausal women find them nearly intolerable [15]. Hot flashes are the most usual complaint of perimenopausal and postmenopausal women. Various entities—such as spicy foods, alcohol and drugs, systemic disease, neurological disorders, androgen deprivation, and carcinoid tumors—could lead to flashing reactions [16, 17]. Hot flashes are very common in breast cancer survivors after treatment as well as because of some adjuvant treatment, such as tamoxifen. They are significantly more frequent and more severe in breast cancer patients than in women without a diagnosis of breast cancer [7, 18].

    The use of tamoxifen, aromatase inhibitors, or suppression of ovarian function in the adjuvant treatment of breast cancer increases the frequency and severity of hot flashes [19–21]. Hot flashes were the most common adverse event in the anastrozole (18%) and in the tamoxifen (26%) group in the Immediate Preoperative Anastrozole, Tamoxifen, or Combined with Tamoxifen (IMPACT) trial and in the Arimidex?, Tamoxifen, Alone or in Combination (ATAC) trial [22, 23].

    TREATMENT OF HOT FLASHES

    There is a diversity of treatments developed throughout the years, differing in safety, efficacy, and acceptability for alleviating hot flashes among women with a history of breast cancer. These include hormonal and nonhormonal and pharmacological and nonpharmacological treatments. Furthermore, women have also used simple strategies, such as wearing light clothes, dressing in layers, lowering the room temperature, using air conditioners, drinking cold beverages, and avoiding alcohol, spicy foods, hot drinks, and hot foods, to ameliorate their symptoms before starting complementary or pharmacological interventions [24, 25].

    Placebo Effect

    When reviewing published data on the effect of new therapies in the management of hot flashes, the placebo effect must be seriously considered. Several placebo-controlled trials showed a substantial placebo effect in intervention studies for hot flashes [26–31]. Four weeks of placebo treatment can reduce hot flash frequency and hot flash scores by about 25%. The placebo effect on hot flash scores is, in this regard, a consistent effect [32]. Therefore, when interpreting clinical data, the placebo effect must always be taken into consideration.

    Complementary Interventions

    Black Cohosh

    Black cohosh (Actaea racemosa, synonym Cimicifuga racemosa) is a plant native to eastern parts of North America. In Europe, especially in Germany, black cohosh has been used in the treatment of menopausal symptoms. The mechanism of action is uncertain. There are conflicting data on the estrogenic effect of black cohosh [33–40]. The most recent data suggest that black cohosh has no estrogenic effects and is safe in the treatment of menopausal hot flashes [35, 40, 41].

    There are differences in the results of published clinical data on the efficacy of black cohosh in the treatment of hot flashes. In an uncontrolled trial, 80% of women reported a reduction in hot flashes after 4 weeks of treatment [42]. In an open-label, randomized study of 12-months’ duration, a satisfactory reduction in the frequency and severity of hot flashes in the intervention group was observed [43]. A pilot study revealed that black cohosh reduced hot flashes significantly more than placebo [44]. In contrast, in a randomized, double-blind, placebo-controlled trial of 60 days’ duration in 69 women with a history of breast cancer, black cohosh was not significantly more effective than placebo [45]. A systematic review concluded that there is no evidence for the clinical efficacy of black cohosh in the treatment of hot flashes [46].

    Drug-related toxicities such as nausea, vomiting, headache, and dizziness have been reported. One case suggests a relationship between black cohosh and hepatotoxicity, but assessment of a direct correlation between the two was not possible [47].

    The published data on the effect of black cohosh in the treatment of hot flashes are conflicting. That black cohosh has beneficial effects on the relief of hot flashes is not supported by evidence from methodologically sound clinical trials.

    Phytoestrogens

    Phytoestrogens are plant-derived, naturally occurring estrogens and have the ability to bind and activate human estrogen receptor alpha (ER-) and human estrogen receptor beta (ER-?). Phytoestrogens have both estrogenic and antiestrogenic effects [48]. Soy products and red clover are rich sources of phytoestrogens. Phytoestrogens include isoflavones, lignans, and coumestans.

    The effect of phytoestrogens on hot flashes has been studied in several clinical studies. Two studies showed the efficacy of soy protein in the treatment of hot flashes. In a double-blind, randomized trial of 12 weeks’ duration, 40 g of protein and 76 mg of phytoestrogens per day demonstrated a significant reduction in the incidence of hot flashes compared with placebo [49]. Another double-blind, controlled trial, comparing a soy isoflavone extract of 50 mg genistin and daidzin with placebo, also showed a statistically significant reduction in hot flashes [50]. In contrast, a double-blind, placebo-controlled trial that evaluated soy phytoestrogens for the treatment of hot flashes in 177 breast cancer survivors failed to show a beneficial effect in the incidence and severity of hot flashes. Four weeks of treatment did not show a difference between the group taking soy tablets containing 50 mg of soy isoflavones in each tablet and the placebo group [51]. In a second double-blind, placebo-controlled trial of 12 weeks of treatment in postmenopausal women who were previously treated for early-stage breast cancer, a soy beverage did not alleviate hot flashes more than placebo [52]. Results of other studies also suggest that soy is not better in the reduction of hot flashes than placebo [53–55].

    Several clinical studies assessed the direct relationship between an individual’s dietary intake of soy products and the risk for the development of breast cancer. Few prospective data are available on the effects of phytoestrogens on breast cancer risk. Results do not show protective effects, with the exception of the consumption of phytoestrogens at young ages or the intake of high amounts [56]. The effect of increased phytoestrogens in breast tissue and on the endometrium is obscure. A recent trial of red clover-derived isoflavones did not show any increased mammographic breast density in 205 women, and no effects on estradiol, gonadotropins, lymphocyte tyrosine kinase activity, or menopausal symptoms were observed [57]. No randomized, controlled trials have addressed the long-term safety of phytoestrogens in patients after a diagnosis of breast cancer.

    The results of the trials on the effect of phytoestrogens on hot flashes are contradictory. Comparison of all clinical studies is difficult because of the differences in products and dosages applied. It is not clear what the long-term safety of phytoestrogens is in women after a diagnosis of breast cancer. There is no evidence to support using high doses of soy products for alleviating hot flashes in breast cancer survivors.

    Homeopathy

    There are fewer published clinical trials to evaluate the effectiveness of homeopathy in the treatment of hot flashes. A randomized, placebo-controlled study with three arms—an individualized homeopathic single remedy (a homeopathic practitioner prescribed an individualized homeopathic medication), a homeopathic combination of three medicines (amyl nitrate 3x [1:1,000 dilution], Sanguinaria canadensis 3x [1:1,000 dilution], and Lachesis 12x [1:1,000,000,000,000 dilution], and a placebo —revealed that there was no significant difference in the severity and frequency of hot flashes among the treatment arms [58]. Results from a prospective observational study suggested a significant improvement in symptom scores of estrogen withdrawal after a homeopathic approach in women with breast cancer [59].

    Evidently, well-controlled and good quality clinical trials assessing the efficacy and safety of homeopathic treatments are needed in the treatment of hot flashes in women after a diagnosis of breast cancer before homeopathic treatments can be advised. For now, homeopathic therapies appear not to be effective for alleviating hot flashes.

    Vitamin E

    A double-blind, randomized, placebo-controlled, crossover clinical trial reported a marginal statistical effect of vitamin E (800 IU/day) in the treatment of hot flashes [60]. Four weeks of daily vitamin E was compared with placebo in 120 breast cancer patients. Vitamin E was associated with one less hot flash per person per day and did not induce toxicity. A crossover analysis showed that vitamin E was associated with a minimal decrease in hot flashes. At the end of the study, patients did not prefer vitamin E use over placebo.

    The investigators of that study suggested that vitamin E at a dose of 800 IU daily can be used because it is inexpensive and nontoxic and it might result in a slightly better relief of hot flashes than placebo.

    In other clinical trials, no statistically significant difference between treatment groups and placebo groups was found for adverse events [61]. Evidence is thus limited, and before supporting vitamin E in the management of hot flashes, more clinical data are warranted. Vitamin E is not registered for this indication and should be used with caution.

    Acupuncture

    A randomized study of 45 postmenopausal women with vasomotor symptoms suggested that electroacupuncture decreased the number of hot flashes by 50% in 11 of 15 women studied [62]. Other studies also reported a reduction in hot flashes with acupuncture [63].

    A review article summarized the adverse reactions after acupuncture, such as hepatitis, subacute bacterial endocarditis, and dermatitis [64]. When acupuncture is used, sterile needles are obviously necessary for the safety of patients. Especially in women who have had axillary surgery for lymph node dissection, acupuncture must be used with care to the operated arm because of the risk for the development of lymphedema.

    Behavioral Therapies

    A comprehensive menopausal assessment (CMA) intervention program was tested in 76 breast cancer survivors with menopausal symptoms [65]. In this structured intervention program, the target symptoms were hot flashes, vaginal dryness, and stress urinary incontinence. The focus was on symptom assessment, education, counseling, and specific pharmacological and behavioral interventions. After randomization, a group of women received the usual care and another group received the interventions. Women in the usual care group were not precluded from these interventions but were not encouraged to do so. Women in the intervention group received an individualized plan of care including pharmacological and/or behavioral interventions. The symptom assessments resulted in a reduction in menopausal symptoms and an improvement in sexual functioning in the intervention group. The comprehensive menopausal assessment included so many interventions that it is not clear which intervention was essential to relieve women from menopausal symptoms. Thus, to date, it is unknown which specific component reduced the hot flashes.

    A behavioral relaxation procedure significantly reduced the frequency of hot flashes [66]. The active component of this relaxation treatment was training in slow, deep breathing. Results of another study suggested that an education and exercise program alleviated climacteric symptoms [67]. In a prospective study of 75 postmenopausal women with vasomotor symptoms, women were randomized to a physical exercise program or oral estradiol therapy. Ten women fulfilled the 12-week exercise period. In five of these women, hot flashes decreased to 28% of baseline [68].

    Results of a 10-week randomized trial including 33 postmenopausal women demonstrated that relaxation response training (including mental focusing, diaphragmatic breathing, and breath awareness) could significantly reduce hot flash intensity compared with that seen in a control group [69].

    Behavioral interventions can be effective in the treatment of hot flashes. The drawback of behavioral trials is the impossibility of using a placebo. A substantial placebo effect must be accounted for when interpreting published behavioral therapy data in the treatment of hot flashes.

    Pharmacological Interventions

    Progestagens

    Progestagens have been studied in the treatment of hot flashes. Megestrol acetate, a progestagen used in the treatment of breast cancer, decreased hot flashes by 80%, compared with a 20% decrease in hot flashes in the placebo group. The patients were women with a history of breast cancer and men with prostate cancer receiving androgen-ablation therapy. The medication was equally efficacious in men and women. One of the main side effects was menstrual withdrawal bleeding 1–2 weeks after discontinuing the megestrol acetate [27]. In a randomized trial in postmenopausal breast cancer patients, one group of patients received an i.m. depot of medroxyprogesterone acetate (500 mg on days 1, 14, and 28) and another group of patients received oral megestrol acetate at a dose of 40 mg/d. Hot flashes were reduced by 86% in the entire group of patients without a significant difference between groups. The treatment was generally well tolerated. More patients in the megestrol group experienced adverse events, which, in six women, led to early discontinuation of the treatment [70]. Reasons for interruption of treatment were skin rashes, dyspnea, gastric pain, and increased arterial blood pressure. More clinical studies showed that depomedroxyprogesterone acetate and medroxyprogesterone acetate are effective in the treatment of hot flashes [71–74].

    Transdermal progesterone cream has also been studied, andafter4weeksoftreatment,itwasassociatedwithan83% reduction in hot flashes in progesterone-treated patients and a 19% reduction in placebo-treated patients [75].

    High-dose megestrol acetate is an effective treatment in the management of breast cancer, but the theoretical concern is that low doses of progestagens may stimulate tumor growth [76, 77]. There are conflicting reports about the safety of progestagens in the treatment of hot flashes in breast cancer patients.

    Neuroendocrine Agents

    Clonidine hydrochloride. Clonidine hydrochloride is a centrally active -agonist that reduces vascular reactivity and is primarily indicated in the treatment of hypertension. Several studies suggest that oral or transdermal clonidine is effective in the treatment of hot flashes in postmenopausal women. Clonidine reduces brain norepinephrine release, raises the sweating threshold, and ameliorates hot flashes [78]. Transdermal clonidine was studied in a randomized, double-blind study in postmenopausal women with hot flashes. The reduction in hot flashes was significant in patients who received transdermal clonidine. Eighty percent of patients reported fewer hot flashes, 73% reported a decrease in severity, and 67% reported a decrease in duration [79].

    Another randomized, double-blind trial showed that clonidine reduced the hot flash frequency and severity, but this effect was clinically modest. Drug-related toxicities were mouth dryness, constipation, itchiness under the patch, and drowsiness [26].

    A randomized, double-blind, placebo-controlled trial reported that oral clonidine (0.1 mg/d) given for at least 8 weeks is effective in the treatment of tamoxifen-induced hot flashes in women with a history of breast cancer. Hot flashes decreased by about 37% after 4 weeks of treatment compared with a 20% reduction with placebo, and after 8 weeks of treatment, a reduction in hot flashes of 38% in the clonidine group, versus 24% in the placebo group, was observed [80]. The patients in the clonidine group had significantly more difficulty sleeping than patients in the placebo group.

    Clinical studies demonstrated that clonidine reduced the frequency of hot flashes, but the effect of clonidine on hot flash duration and severity was marginal [81, 82].

    Gabapentin. Gabapentin is a gamma-aminobutyric acid analogue that is used in the treatment of epilepsy or neurogenic pain.

    Two pilot studies suggested that gabapentin is effective in the treatment of hot flashes [83, 84]. A 12-week, randomized, placebo-controlled trial in postmenopausal women taking gabapentin at a dose of 900 mg/d showed a reduction of 45% in hot flash frequency and a 54% reduction in hot flash composite score (frequency and severity) [30]. Adverse events in the gabapentin group were somnolence, dizziness, and rash with or without peripheral edema. In some patients who took gabapentin, blood laboratory tests showed a decrease in serum albumin, total protein, total bilirubin, blood urea nitrogen, and platelets after 12 weeks of treatment, compared with baseline.

    In a randomized, double-blind, placebo-controlled trial in 420 women with breast cancer, one group received a placebo, one group received gabapentin at a dose of 300 mg/d, and one group received gabapentin at a dose of 900 mg/d for 8 weeks. The percentage decreases in hot flash severity score between baseline and 8 weeks were: 31% in the 300-mg gabapentin group and 46% in the 900-mg gabapentin group. Gabapentin at a dose of 300 mg/d gave a significant reduction in hot flash frequency but not a significant reduction in the severity of hot flashes. There was a significant decrease in hot flash frequency and severity at a dose of 900 mg/d of gabapentin. A major limitation of this study is that the side effects of gabapentin have not been reported [31].

    Based on these results, gabapentin can be considered effective in the management of hot flashes in postmenopausal women after a diagnosis of breast cancer. More clinical data are needed on the side effects and safety associated with the use of gabapentin in the treatment of hot flashes.

    Selective serotonin reuptake inhibitors. Selective serotonin reuptake inhibitors (SSRIs) are antidepressants. Based on the modes of action of these antidepressants and the theory of the pathophysiology of hot flashes, SSRIs have been studied in the treatment of hot flashes. Specifically, the SSRIs venlafaxine, paroxetine, and fluoxetine were evaluated in the treatment of hot flashes.

    Venlafaxine.

    Several clinical studies have been performed to evaluate the use of venlafaxine in the treatment of hot flashes. Venlafaxine affects both serotonin and norepinephrine reuptake in contrast to the SSRIs paroxetine and fluoxetine. Two pilot studies reported that hot flashes were reduced by approximately 55%–67% with venlafaxine [28, 85]. Results of a double-blind, placebo-controlled trial in 192 breast cancer patients showed a significant decrease in median hot flash scores in all venlafaxine groups compared with placebo. After 4 weeks of treatment, median hot flash scores were reduced by 27% in the placebo group, by 37% in the 37.5-mg venlafaxine extended release (XR) group, and by 61% in the 75-mg venlafaxine XR and 150-mg venlafaxine XR treated patients [29]. At the end of 4 weeks of treatment, the investigators of the study asked the participants to participate in an 8-week open-label study. Participants received 37.5–150 mg/day of venlafaxine XR. In the 37.5-mg group, hot flashes decreased by 26%. In the 75-mg and 150-mg group, hot flashes scores decreased by 60% [86].

    In a 12-week placebo-controlled trial, postmenopausal women received 37.5 mg of venlafaxine XR for 1 week followed by 75 mg of venlafaxine XR for 11 weeks. After 4 weeks, the mean scores in the placebo and venlafaxine groups were the same. After 11 weeks of treatment, venlafaxine was associated with a 51% reduction in hot flashes, compared with a 15% reduction in the placebo group [87].

    In all venlafaxine studies, venlafaxine was well tolerated. The most common adverse events included dry mouth, constipation, loss of appetite, nausea, and sleepiness. The 150-mg dose of venlafaxine XR was associated with more adverse events than the 37.5-mg and 75-mg doses of venlafaxine XR. The efficacies of 75 mg of venlafaxine XR and 150 mg of venlafaxine XR in the treatment of hot flashes were not different and were 60%. Based on these results, the advised dose of venlafaxine in the treatment of hot flashes is 75 mg/d [86].

    Sexual dysfunction is a common adverse event of anti-depressant treatment, especially when using SSRIs [88]. In the study of Loprinzi et al. [29], the venlafaxine doses of 37.5–150 mg XR per day did not reduce libido after 4 weeks of treatment.

    In conclusion, venlafaxine is effective and can be used in the treatment of hot flashes in women after a diagnosis of breast cancer. How long patients must take venlafaxine after starting this intervention and also the effectiveness of venlafaxine for durations of longer than 12 weeks are unknown. Further long-term, well-controlled clinical trials are needed to investigate the efficacy of venlafaxine in the management of hot flashes. These trials should include the side effects and, specifically, the possible interference of venlafaxine with sexual function after long-term treatment.

    We are currently conducting a randomized, double-blind, placebo-controlled trial comparing venlafaxine with clonidine and placebo.

    Fluoxetine.

    A randomized, double-blind, crossover, placebo-controlled trial in 81 breast cancer patients reported a decrease in the incidence of hot flashes by 50% using fluoxetine at a dose of 20 mg/d, versus a 36% decrease with placebo. There was no statistically significant difference in the toxicities in the two treatment arms [89]. More clinical data are needed on the efficacy of fluoxetine in the management of hot flashes.

    Paroxetine.

    Controlled-release paroxetine was evaluated in a randomized, double-blind, placebo-controlled trial in 165 postmenopausal women with hot flashes. After 6 weeks of treatment, 12.5 mg/d of paroxetine reduced hot flashes by 62% and 25 mg/d of paroxetine reduced hot flashes by 65%, whereas there was a 38% reduction in hot flashes by placebo. The most frequently reported adverse events in the controlled-release paroxetine group were headache, nausea, and insomnia. Paroxetine was well tolerated, with more adverse events in the 25-mg than in the 12.5-mg treatment group [90].

    Recently published data reported that paroxetine in doses of 10–20 mg is effective in the treatment of hot flashes in women with and without a history of breast cancer. This randomized, double-blind, crossover, placebo-controlled trial demonstrated that 10 mg of paroxetine reduced hot flash frequency and the composite score by 40.6% and 45.6%, respectively, compared with 13.7% and 13.7% reductions, respectively, with placebo. Paroxetine at a dose of 20 mg reduced hot flash frequency and the composite score by 51.7% and 56.1%, respectively, versus 26.6% and 28.8% reductions, respectively, in the placebo group. The most common adverse events were drowsiness and nausea in the 20-mg paroxetine group [91]. These trials demonstrated that paroxetine is effective in the treatment of hot flashes, but further clinical trials assessing the efficacy and safety of paroxetine in breast cancer survivors are needed.

    Safety of SSRIs.

    The SSRIs are also commonly prescribed in the treatment of hot flashes in women who take tamoxifen. SSRIs are known to inhibit cytochrome P450 (CYP)2D6, an enzyme that is important for the metabolism of many drugs, such as tamoxifen, to its active metabolite endoxifen. Some studies reported an interaction of CYP2D6 polymorphisms and CYP2D6 inhibitors. Plasma concentrations of endoxifen were lower in patients who were treated with SSRIs than in patients who did not receive treatment with SSRIs in combination with tamoxifen [92]. The weak inhibitor of CYP2D6, venlafaxine, had very little effect on plasma endoxifen concentrations, which is in contrast to the combination of paroxetine or sertraline with tamoxifen, which resulted in substantially lower plasma endoxifen concentrations. The variations in plasma endoxifen concentrations that are associated with CYP2D6 gene polymorphisms and CYP2D6 inhibitors can affect the antitumor efficacy or adverse events of tamoxifen [93].

    Further clinical trials are needed to evaluate the safety of SSRIs in combination with tamoxifen. It is advisable to inform women after a diagnosis of breast cancer of the uncertainties about the interactions of some SSRIs and tamoxifen.

    CONCLUSIONS AND RECOMMENDATIONS

    Many published results of clinical trials are available in the management of hot flashes, but there are limited well-controlled trials assessing the role of pharmacologically and nonpharmacologically based treatments of hot flashes in breast cancer patients. A limitation of most trials is the duration of the treatment and follow-up of patients. The U.S. Food and Drug Administration recommended a study period of 12 weeks for trials in the treatment of hot flashes [94]. Several trials in the management of hot flashes have a shorter duration than 12 weeks.

    Another limitation of studies in patients with hot flashes is the absence of a placebo group. Several trials have shown a significant placebo effect in the relief of hot flashes. A placebo group as a control group is thus necessary in these studies. Therefore, when interpreting clinical data, the placebo effect should always be taken into consideration.

    Only 13 trials that we found meet the criteria of a study period of 12 weeks or longer and were placebo controlled. Six studies were performed with phytoestrogens in the management of hot flashes, but the results of those studies were contradictory. One preliminary homeopathic trial showed no treatment effect on hot flashes. Four trials were performed with progestagens and all showed a reduction in the incidence and severity of hot flashes. One trial was performed with gabapentin and one trial was performed with venlafaxine in the management of hot flashes. Both showed a reduction in the incidence and severity of hot flashes in postmenopausal women. However, only two trials performed with phytoestrogens were evaluated in breast cancer patients. The other trials evaluated treatment options in the management of hot flashes in postmenopausal women without a history of breast cancer. This emphasizes the need for larger-scale and long-term prospective trials to test the efficacy and safety of selected pharmacological and nonpharmacological strategies in the management of hot flashes after the diagnosis of breast cancer and induction of early menopause. Several questions are still unanswered, including the time period that patients need pharmacological interventions after starting with the intervention in the management of hot flashes. The optimal time period of use can be based on the natural history of hot flashes, which mostly resolve within 2–3 years of menopause. Hence, one may consider tapering and discontinuation of SSRIs after about this length of time.

    The first step to control hot flashes in breast cancer patients is advice and information about simple strategies in the management of hot flashes, such as wearing light clothes, dressing in layers, lowering the room temperature, using air conditioners, drinking cold beverages, and avoiding alcohol, hot drinks, or hot food to ameliorate symptoms. For pharmacological intervention of hot flashes, venlafaxine can be used. Venlafaxine is preferable to the other SSRIs for several reasons. Most clinical data are available for venlafaxine. The safety of the other SSRIs is unclear in combination with tamoxifen, and a possibly clinically relevant reduction in endoxifen, the active metabolite of tamoxifen, may take place. Another possible treatment option is clonidine, an antihypertensive agent. Gabapentin was reported to be effective in the treatment of hot flashes; however, more information on the toxicity profile of this agent is needed before prescribing gabapentin outside clinical trials.

    DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST

    The authors indicate no potential conflicts of interest.

    REFERENCES

    Del Mastro L, Venturini M, Sertoli MR et al. Amenorrhea induced by adjuvant chemotherapy in early breast cancer patients: prognostic role and clinical implications. Breast Cancer Res Treat 1997;43:183–190.

    Goodwin PJ, Ennis M, Pritchard KI et al. Risk of menopause during the first year after breast cancer diagnosis J Clin Oncol 1999;17:2365–2370.

    Shapiro CL, Recht A. Side effects of adjuvant treatment of breast cancer. N Engl J Med 2001;344:1997–2008.

    Early Breast Cancer Trialists’ Collaborative Group. Polychemotherapy for early breast cancer: an overview of the randomised trials. Lancet 1998;352:903–942.

    Davidson NE. Ovarian ablation as adjuvant therapy for breast cancer. J Natl Cancer Inst Monogr 2001;30:67–71.

    Boccardo F, Rubagotti A, Perrotta A et al. Ovarian ablation versus goserelin with or without tamoxifen in pre- perimenopausal patients with advanced breast cancer: results of a multicentric Italian study. Ann Oncol 1994;5:337–342.

    Couzi RJ, Helzlsouer KJ, Fetting JH et al. Prevalence of menopausal symptoms among women with a history of breast cancer and attitudes toward estrogen replacement therapy. J Clin Oncol 1995;13: 2737–2744.

    Ganz PA. Impact of tamoxifen adjuvant therapy on symptoms, functioning and quality of life. J Natl Cancer Inst Monogr 2001:30:130–134.

    Savard J, Davidson JR, Ivers H et al. The association between nocturnal hot flashes and sleep in breast cancer survivors. J Pain Symptom Manage 2004;27:513–522.

    Day R, Ganz PA, Costantino JP et al. Health-related quality of life and tamoxifen in breast cancer prevention: a report from the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Clin Oncol 1999;17: 2659–2669.

    Finck G, Barton DL, Loprinzi CL et al. Definitions of hot flashes in breast cancer survivors. J Pain Symptom Manage 1998;16:327–333.

    Kronenberg F. Hot flashes: phenomenology, quality of life, and search for treatment options. Exp Gerontol 1994;29:319–336.

    Stearns V, Ullmer L, Loepez JF et al. Hot flushes. Lancet 2002;360: 1851–1861.

    Shanafelt TD, Barton DL, Adjei AA et al. Pathophysiology and treatment of hot flashes. Mayo Clin Proc 2002;77:1207–1218.

    Kronenberg F. Hot flashes: epidemiology and physiology. Ann N Y Acad Sci 1990;592:52–86; discussion 123–133.[Abstract]

    Mohyi D, Tabassi K, Simon J. Differential diagnosis of hot flashes. Maturitas 1997;27:203–214.

    Schnirer II, Yao JC, Ajani JA. Carcinoid. A comprehensive review. Acta Oncol 2003;42: 672–692.

    Fallowfield L, Fleissig A, Edwards R et al. Tamoxifen for the prevention of breast cancer: psychosocial impact on women participating in two randomized controlled trials. J Clin Oncol 2001;19:1885–1892.

    Fisher B, Costantino J, Redmond C et al. A randomized clinical trial evaluating tamoxifen in the treatment of patients with node-negative breast cancer who have estrogen-receptor-positive tumors. N Engl J Med 1989;320:479–484.[Abstract]

    Love RR, Cameron L, Connelle BL et al. Symptoms associated with tamoxifen treatment in postmenopausal women. Arch Intern Med 1991;151:1842–1847.[Abstract]

    Carpenter JS, Andrykowski MA, Cordova M et al. Hot flashes in premenopausal women treated for breast carcinoma: prevalence, severity, correlates, management, and relation to quality of life. Cancer 1998;82: 1682–1691.

    Smith IE, Dowsett M, Ebbs SR et al. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: the Immediate Preoperative Anastrozole, Tamoxifen, or Combined with Tamoxifen (IMPACT) multicenter double-blind randomized trial. J Clin Oncol 2005;23:5108–5116.

    Howell A, Cuzick J, Baum M et al. Results of the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial after completion of 5 years’ adjuvant treatment for breast cancer. Lancet 2005;365:60–62.

    Stevenson DW, Delprato DJ. Multiple component self-control program for menopausal hot flashes. J Behav Ther Exp Psychiatry 1983;14:137–140.

    Freedman RR. Biochemical, metabolic, and vascular mechanisms in menopausal hot flashes. Fertil Steril 1998;70:332–337.

    Goldberg RM, Loprinzi CL, O’Fallon JR et al. Transdermal clonidine for ameliorating tamoxifen-induced hot flashes. J Clin Oncol 1994;12:155–158.

    Loprinzi CL, Michalak JC, Quella SK et al. Megestrol acetate for the prevention of hot flashes. N Engl J Med 1994;331:347–352.

    Loprinzi CL, Pisansky TM, Fonseca R et al. Pilot evaluation of venlafaxine hydrochloride for the therapy of hot flashes in cancer survivors. J Clin Oncol 1998;16:2377–2381.

    Loprinzi CL, Kugler JW, Sloan JA et al. Venlafaxine in management of hot flashes in survivors of breast cancer: a randomised controlled trial. Lancet 2000;356:2059–2063.

    Guttuso T Jr, Kurlan R, McDermott MP et al. Gabapentin’s effects on hot flashes in postmenopausal women: a randomized controlled trial. Obstet Gynecol 2003;101:337–345.

    Pandya KJ, Morrow GR, Roscoe JA et al. Gabapentin for hot flashes in 420 women with breast cancer: a randomised double-blind placebo-controlled trial. Lancet 2005;366:818–824.

    Sloan JA, Loprinzi CL, Novotny PJ et al. Methodologic lessons learned from hot flash studies. J Clin Oncol 2001;19:4280–4290.

    Zierau O, Bodinet C, Kolba S et al. Antiestrogenic activities of Cimicifuga racemosa extracts. J Steroid Biochem Mol Biol 2002;80:125–130.

    Huntley A, Ernst E. A systematic review of the safety of black cohosh. Menopause 2003;10:58–64.

    Bodinet C, Freudenstein J. Influence of marketed herbal menopause preparations on MCF-7 cell proliferation. Menopause 2004;11:281–289.

    Mahady GB. Black cohosh (Actae/Cimicifuga racemosa) review of the clinical data for safety and efficacy in menopausal symptoms. Treat Endocrinol 2005;4:177–184.

    Einbond LS, Shimizu M, Xiao D et al. Growth inhibitory activity of extracts and purified components of black cohosh on human breast cancer cells. Breast Cancer Res Treat 2004;83:221–231.

    Hostanska K, Nisslein T, Freudenstein J et al. Cimicifuga racemosa extract inhibits proliferation of estrogen receptor-positive and negative human breast carcinoma cell lines by induction of apoptosis. Breast Cancer Res Treat 2004;84:151–160.

    Freudenstein J, Dasenbrock C, Nisslein T. Lack of promotion of estrogen-dependent mammary gland tumors in vivo by an isopropanolic Cimicifuga racemosa extract. Cancer Res 2002;62:3448–3452.

    Lupu R, Mehmi I, Atlas E et al. Black cohosh, a menopausal remedy, does not have estrogenic activity and does not promote breast cancer cell growth. Int J Oncol 2003;23:1407–1412.

    Stromeier S, Petereit F, Nahrstedt A. Phenolic esters from the rhizomes of Cimicifuga racemosa do not cause proliferation effects in MCF-7 cells. Planta Med 2005;71:495–500.

    Liske E. Therapeutic efficacy and safety of Cimicifuga racemosa for gynecologic disorders. Adv Ther 1998;15:45–53.

    Hernandez Munoz G, Pluchino S. Cimicifuga racemosa for the treatment of hot flushes in women surviving breast cancer. Maturitas 2003;44 (suppl 1):S59–S65.

    Pockaj BA, Loprinzi CL, Sloan JA et al. Pilot evaluation of black cohosh for the treatment of hot flashes in women. Cancer Invest 2004;22:515–521.

    Jacobson JS, Troxel AB, Evans J et al. Randomized trial of black cohosh for the treatment of hot flashes among women with a history of breast cancer. J Clin Oncol 2001;19:2739–2745.

    Borrelli F, Ernst E. Cimicifuga racemosa: a systematic review of its clinical efficacy. Eur J Clin Pharmacol 2002;58:235–241.

    Cohen SM, O’Connor AM, Hart J et al. Autoimmune hepatitis associated with the use of black cohosh: a case study. Menopause 2004;11:575–577.

    Harris DM, Besselink E, Henning SM et al. Phytoestrogens induce differential estrogen receptor alpha- or beta-mediated responses in transfected breast cancer cells. Exp Biol Med (Maywood) 2005;230:558–568.

    Albertazzi P, Pansini F, Bonaccorsi G et al. The effect of dietary soy supplementation on hot flushes. Obstet Gynecol 1998;91:6–11.

    Upmalis DH, Lobo R, Bradley L et al. Vasomotor symptom relief by soy isoflavone extract tablets in postmenopausal women: a multicenter, double-blind, randomized, placebo-controlled study. Menopause 2000;7:236–242.

    Quella SK, Loprinzi CL, Barton DL et al. Evaluation of soy phytoestrogens for the treatment of hot flashes in breast cancer survivors: a North Central Cancer Group trial. J Clin Oncol 2000;18:1068–1074.

    Van Patten CL, Olivotto IA, Chambers GK et al. Effect of soy phytoestrogens on hot flashes in postmenopausal women with breast cancer: a randomized, controlled clinical trial. J Clin Oncol 2002;20: 1449–1455.

    Tice JA, Ettinger B, Ensrud K et al. Phytoestrogen supplements for the treatment of hot flashes: the Isoflavone Clover Extract (ICE) study: a randomized controlled trial. JAMA 2003;9:290:207–214.

    Penotti M, Fabio E, Modena AB et al. Effect of soy-derived isoflavones on hot flushes, endometrial thickness, and the pulsatility index of the uterine and cerebral arteries. Fertil Steril 2003;79:1112–1117.

    MacGregor CA, Canney PA, Patterson G et al. A randomized double-blind controlled trial of oral soy supplements versus placebo for treatment of menopausal symptoms in patients with early breast cancer. Eur J Cancer 2005;41:708–714.

    Peeters PH, Keinan-Boker L, van der Schouw YT et al. Phytoestrogens and breast cancer risk. Review of the epidemiological evidence. Breast Cancer Res Treat 2003;77:171–183.

    Atkinson C, Warren RM, Sala E et al. Red-clover-derived isoflavones and mammographic breast density: a double blind, randomized, placebo-controlled trial [ISRCTN42940165]. Breast Cancer Res 2004;6: R170–R179.

    Jacobs J, Herman P, Heron K et al. Homeopathy for menopausal symptoms in breast cancer survivors: a preliminary randomized controlled trial. J Altern Complement Med 2005;11:21–27.

    Thompson EA, Reilly D. The homeopathic approach to the treatment of symptoms of oestrogen withdrawal in breast cancer patients. A prospective observational study. Homeopathy 2003;92:131–134.

    Barton DL, Loprinzi CL, Quella SK et al. Prospective evaluation of vitamin E for hot flashes in breast cancer survivors. J Clin Oncol 1998;16: 495–500.

    Meagher EA. Treatment of atherosclerosis in the new millennium: is there a role for vitamin E? Prev Cardiol 2003;6:85–90.

    Wyon Y, Wijma K, Nedstrand E et al. A comparison of acupuncture and oral estradiol treatment of vasomotor symptoms in postmenopausal women. Climacteric 2004;7:153–164.

    Nedstrand E, Wijma K, Wyon Y et al. Vasomotor symptoms decrease in women with breast cancer randomized to treatment with applied relaxation or electro-acupuncture: a preliminary study. Climacteric 2005;8:243–250.

    Ernst E, White A. Life-threatening adverse reactions after acupuncture? A systematic review. Pain 1997;71:123–126.

    Ganz PA, Greendale GA, Petersen L et al. Managing menopausal symptoms in breast cancer survivors: results of a randomized controlled trial. J Natl Cancer Inst 2000;1054–1064.

    Freedman RR, Woodward S. Behavioral treatment of menopausal hot flushes: evaluation by ambulatory monitoring. Am J Obstet Gynecol 1992;167:436–439.

    Ueda M. A 12-week structured education and exercise program improved climacteric symptoms in middle-aged women. J Physiol Anthropol Appl Human Sci 2004;23:143–148.

    Lindh-Astrand L, Nedstrand E, Wyon Y et al. Vasomotor symptoms and quality of life previously sedentary postmenopausal women randomised to physical activity or estrogen therapy. Maturitas 2004;48: 97–105.

    Irvin JH, Domar AD, Clark C et al. The effects of relaxation response training on menopausal symptoms. J Psychosom Obstet Gynecol 1996;17:202–207.

    BertelliG,VenturiniM,DelMastroLetal.Intramusculardepotmedroxy-progesterone versus oral megestrol for the control of postmenopausal hot flashes in breast cancer patients: a randomized study. Ann Oncol 2002;13:883–888.

    Bullock JL, Massey FM, Gambrell RD Jr. Use of medroxyprogesterone acetate to prevent menopausal symptoms. Obstet Gynecol 1975;46: 165–168.[Abstract]

    Schiff I, Tulchinsky D, Cramer D et al. Oral medoxyprogesterone in the treatment of postmenopausal symptoms. JAMA 1980;244:1443–1445.[Abstract]

    Morrison JC, Martin DC, Blair RA et al. The use of medroxyprogesterone acetate for relief of climacteric symptoms. Am J Obstet Gynecol 1980;138:99–104.

    Quella SK, Loprinzi CL, Sloan JA et al. Long term use of megestrol acetate by cancer survivors for the treatment of hot flashes. Cancer 1998;82:1784–1788.

    Leonetti HB, Longo S, Anasti JN. Transdermal progesterone cream for vasomotor symptoms and postmenopausal bone loss. Obstet Gynecol 1999;94:225–228.

    Ross RK, Paganini-Hill A, Wan PC et al. Effect of hormone replacement therapy on breast cancer risk: estrogen versus estrogen plus progestin. J Natl Cancer Inst 2000;92:328–332.

    von Schoultz E, Rutqvist LE. Menopausal hormone therapy after breast cancer: the Stockholm randomized trial. J Natl Cancer Inst 2005;97: 533–535.

    Freedman RR. Physiology of hot flashes. Am J Hum Biol 2001;13: 453–464.

    Nagamani M, Kelver ME, Smith ER. Treatment of menopausal hot flashes with transdermal administration of clonidine. Am J Obstet Gynecol 1987;156:561–566.

    Pandya KJ, Raubertas RF, Flynn PJ et al. Oral clonidine in postmenopausal patients with breast cancer experiencing tamoxifen-induced hot flashes: a University of Rochester Cancer Center Community Clinical Oncology Program study. Ann Intern Med 2000;132:788–793.

    Clayden JR, Bell JW, Pollard P. Menopausal flushing: double-blind trial of a nonhormonal medication. Br Med J 1974;1:409–412.

    Laufer LR, Erlik Y, Meldrum DR et al. Effect of clonidine on hot flashes in postmenopausal women. Obstet Gynecol 1982;60:583–586.[Abstract]

    Loprinzi L, Barton DL, Sloan JA et al. Pilot evaluation of gabapentin for treating hot flashes. Mayo Clin Proc 2002;77:1159–1163.

    Pandya KJ, Thummala AR, Griggs JJ et al. Pilot study using gabapentin for tamoxifen-induced hot flashes in women with breast cancer. Breast Cancer Res Treat 2004;83:87–89.

    Quella SK, Loprinzi CL, Sloan J et al. Pilot evaluation of venlafaxine for the treatment of hot flashes in men undergoing androgen ablation therapy for prostate cancer. J Urol 1999;162:98–102.

    Barton D, La Vasseur B, Loprinzi C et al. Venlafaxine for the control of hot flashes: results of a longitudinal continuation study. Oncol Nurs Forum 2002;29:33–40.

    Evans ML, Pritts E, Vittinghoff E et al. Management of postmenopausal hot flushes with venlafaxine hydrochloride: a randomized, controlled trial. Obstet Gynecol 2005;105:161–166.

    Gregorian RS, Golden KA, Bahce A et al. Antidepressant-induced sexual dysfunction. Ann Pharmacother 2002;36:1577–1589.

    Loprinzi CL, Sloan JA, Perez EA et al. Phase III evaluation of fluoxetine for treatment of hot flashes. J Clin Oncol 2002;20:1578–1583.

    Stearns V, Beebe KL, Iyengar M et al. Paroxetine controlled release in the treatment of menopausal hot flashes: a randomized controlled trial. JAMA 2003;289:2827–2834.

    Stearns V, Slack R, Greep N et al. Paroxetine is an effective treatment for hot flashes: results from a prospective randomized clinical trial. J Clin Oncol 2005;23:6919–6930.

    Stearns V, Johnson MD, Rae JM et al. Active tamoxifen metabolite plasma concentrations after coadministration of tamoxifen and the selective serotonin reuptake inhibitor paroxetine. J Natl Cancer Inst 2003;95: 1758–1764.

    Jin Y, Desta Z, Stearns V et al. CYP2D6 genotype, antidepressant use, and tamoxifen metabolism during adjuvant breast cancer treatment. J Natl Cancer Inst 2005;97:30–39.

    U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER). Guidance for Industry: Estrogen and Estrogen/Progestin Drug Products to Treat Vasomotor Symptoms and Vulvar and Vaginal Atrophy Symptoms — Recommendations for Clinical Evaluation. Available at http://www.fda.gov/cder/guidance/5412dft.doc. Accessed January 2003.(Annelies H. Boekhouta, Jo)