Randomized comparison of adjuvant aromatase inhibitor (AI) exemestane (E) plus ovarian function suppression (OFS) vs tamoxifen (T) plus OFS in premenopausal women with hormone receptor-positive (HR+) early breast cancer (BC) Joint analysis of IBCSG TEXT and SOFT trials

SUMMARY: Breast cancer is the most common cancer among women in the US and about 1 in 8 women (12%) will develop invasive breast cancer during their lifetime. Approximately, 233,000 new cases of invasive breast cancer will be diagnosed in 2014 and 40,000 women will die of the disease. Approximately 75% of patients with breast cancer are hormone receptor positive (Estrogen Receptor/Progesterone Receptor positive) and this is a predictor of response to endocrine therapy. In premenopausal woman, the ovary is the main source of estrogen production, whereas in postmenopausal women, the primary source of estrogen is the Aromatase enzyme mediated conversion of androstenedione and testosterone to estrone and estradiol in extragonadal/peripheral tissues. Presently available therapies include Tamoxifen and other Selective ER Modulators, which modulate ER alpha activity, Aromatase Inhibitors and Ovarian ablation that decrease estrogen production and FASLODEX® (Fulvestrant) that down regulates Estrogen Receptor. Aromatase Inhibitors (AI’s) are often prescribed, due to their superiority over Tamoxifen, for postmenopausal women with Hormone Receptor positive breast tumors, in adjuvant as well as metastatic settings. AI’s however, are not effective in premenopausal women, as these individuals derive their estrogen mainly from ovaries and not extragonadal tissues. The 2000 Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) overview, as well as subsequent studies comparing adjuvant ovarian ablation/suppression with adjuvant chemotherapy in premenopausal women with hormone positive breast tumors, have demonstrated similar magnitude of benefit. The TEXT (Tamoxifen and Exemestane Trial) and SOFT (Suppression of Ovarian Function Trial) are two phase III randomized trials, conducted at the same time and included premenopausal women (average age was 43 years) with hormone receptor positive early breast cancer. In the joint analysis of these two trials in which 5738 women were enrolled, the authors set out to answer 2 important questions – whether adjuvant AI improves outcomes in this patient group when their Ovarian Function is suppressed and whether there is any benefit with Ovarian Function suppression in premenopausal women suitable for adjuvant Tamoxifen. TEXT randomized patients (N=2672) within 3 months of surgery to 5 years of AROMASIN® (Exemestane) plus Ovarian Function Suppression (OFS) or 5 years of Tamoxifen plus OFS. The SOFT study randomized patients (N=3066) to 5 years of AROMASIN® plus OFS or 5 years of Tamoxifen plus OFS or 5 years of Tamoxifen alone. OFS choices included oophorectomy, ovarian irradiation or 5 years of TRELSTAR® (Triptorelin), a GnRH (Gonadotropin Releasing Hormone) agonist. The primary endpoint of these two studies was Disease Free Survival (DFS). In this joint analysis the outcomes for 4690 women randomized to receive AROMASIN® plus OFS or Tamoxifen plus OFS for 5 years, were analyzed. The 5 year Disease Free Survival was 91.1% in the AROMASIN® plus OFS group and 87.3% in the Tamoxifen plus OFS group (HR=0.72, P<0.0002). Compared to patients receiving Tamoxifen plus OFS, AROMASIN® plus OFS reduced the relative risk of premenopausal women developing a subsequent invasive breast cancer by 28% and the relative risk of breast cancer recurrence by 34%. The authors concluded that this largest joint analysis, evaluating adjuvant AI therapy with OFS in premenopausal women with Hormone receptor positive breast cancer, has demonstrated that 5 years of highly effective adjuvant endocrine therapy without chemotherapy can result in excellent outcomes. Further, AROMASIN® may be better than tamoxifen, when given with Ovarian Function Suppression. Pagani O, Regan MM, Walley B, et al. J Clin Oncol 32:5s, 2014 (suppl; abstr LBA1)

Clinical validity of circulating tumour cells in patients with metastatic breast cancer a pooled analysis of individual patient data

SUMMARY: Circulating tumor cells (CTCs) are epithelial cells that are shed into the circulation from a primary or metastatic tumor. After being shed, CTCs can remain in the circulation or undergo apoptosis. Evaluation of CTCs during the course of disease has prognostic value. Because of the very low concentrations of CTCs (1 CTC in the background of millions of normal hematopoietic cells) in the peripheral blood, different technologies have been developed that will allow enrichment and detection of these CTCs. One such technology is the CellSearch® system which is the first FDA-approved test for CTC assessment, in the peripheral blood of Metastatic Breast Cancer (MBC) patients. This automated system is able to enrich the peripheral blood sample with CTCs and the cells then are fluorescently stained for CytoKeratins (CK8,18 and 19), Common Leukocyte Antigen (CD45) and a nuclear dye (DAPI). CTCs are identified when they are CK positive, CD45 negative and DAPI positive. In essence, CTC assessment is a real time, peripheral blood evaluation (“Liquid Biopsy”) in MBC patients. The authors in this study conducted a pooled analysis to assess the clinical validity of CTCs in patients with MBC, as previously published studies reported contradictory results and were unable to ascertain whether enumeration of CTCs had better prognostic value than the traditional clinical and pathological features of the tumor and serum tumor markers. Data was gathered on 1944 patients with MBC, who had participated in clinical trials at 17 centers between 2003 and 2012. Participants in these studies were starting a new line of therapy (predominantly chemotherapy), and these studies had CTC quantification using CellSearch® platform, before start of new treatment (baseline), data for Progression Free Survival, Overall Survival or both. Using accepted statistical methodologies, the authors noted the following findings-

1) At baseline prior to starting treatment, 47% (N=911) patients had 5 or more CTCs per 7•5 mL of peripheral blood, suggesting more aggressive disease and this was associated with decreased Progression Free Survival (P<0•0001) and Overall Survival (P<0•0001), compared with patients with less than 5 CTCs per 7•5 mL at baseline.

2) At 3-5 weeks after start of treatment (1-2 cycles of treatment), when adjusted for baseline CTC count, 5 or more CTCs per 7•5 mL of peripheral blood was associated with shortened Progression Free Survival (P<0•0001) and Overall Survival (P<0•0001), suggesting that these patients were treatment resistant.

3) An early decrease in the CTC at week 3-5, from a high baseline of 5 or more CTCs per 7•5 mL to less than 5 CTCs per 7.5 ml was associated with significantly longer Progression Free Survival and Overall Survival.

4) Enumeration of CTCs was a better predictor of prognosis than mucin based serum biomarkers such as CEA and CA15-3.

The prognostic value of high CTC count at baseline and at 3-5 weeks of treatment, on Progression Free Survival and Overall Survival was maintained in all subgroups tested, regardless of breast cancer subtypes and type of therapy patients received. Based on this pooled analysis, with the largest assessment of CTC enumeration in MBC patients, the authors concluded that CTC count can prognosticate Progression Free Survival and Overall Survival early in the treatment course, allowing customized care. Further, CTC enumeration, unlike serum tumor markers, correlates with clinical and pathological characteristics. Bidard F, Peeters DJ, Fehm T, et al. The Lancet Oncology 2014;15:406-414

AFINITOR® overcomes Endocrine Resistance in Breast Cancer

AFINITOR® can overcome endocrine resistance in patients with Metastatic Breast Cancer. This was demonstrated in the BOLERO-2 trial in which patients who had progressed on non-steroidal Aromatase Inhibitors, when treated with a combination of steroidal Aromatase Inhibitor AROMASIN® (Exemestane) and AFINITOR® (Everolimus), had significantly improved Progression Free Survival and Clinical Benefit. AFINITOR® is a mTOR inhibitor and mTOR pathway has been implicated as one of the mechanisms for endocrine resistance in Breast cancer. A recent BOLERO-2 trial update, was published in the Breast Cancer Research and Treatment 2013.

Everolimus plus exemestane as first-line therapy in HR+, HER2− advanced breast cancer in BOLERO-2

SUMMARY:Breast cancer is the most common cancer among women in the US and about 1 in 8 women (12%) will develop invasive breast cancer during their lifetime. Approximately, 233,000 new cases of invasive breast cancer will be diagnosed in 2014 and 40,000 women will die of the disease. Approximately 75% of patients with breast cancer are hormone receptor positive and 15%-20% of breast cancers overexpress HER2/neu oncogene. As such a significant number of breast cancers are Hormone Receptor (HR) positive and HER2 negative. Aromatase Inhibitors (AI’s) are often prescribed, due to their superiority over Tamoxifen, for post menopausal women with HR+ and HER2-negative breast tumors, both in adjuvant as well as metastatic settings. These patients will eventually develop progressive disease on endocrine therapy with AI’s, attributed to endocrine resistance. The average Progression Free Survival for these patients is 4-6 months when treated with other hormonal interventions including higher doses of FASLODEX® (Fulvestrant). Further, FASLODEX® has not been shown to be superior to steroidal or non steroidal AI’s. The mechanism of endocrine resistance has been attributed to cross talk between Estrogen Receptor signaling and PI3K/AKT/mTOR pathway. The PI3K/AKT/mTOR is a complex pathway, essential for cell proliferation, survival and apoptosis (programmed cell death). This pathway is of interest because of its increased activity in malignant cells as a result of amplification or mutation of the genes associated with PI3-kinase (phosphatidylinositol-3 kinase) and AKT, as well as loss of function of PTEN. PTEN normally prevents activation of AKT and its downstream pathways. Therefore, elevated ER signaling and breast cancer progression has been associated with activation of this mTOR (mammalian Target Of Rapamycin) pathway. Everolimus (AFINITOR®) is a mTOR inhibitor that has been shown to inhibit ER signaling and restore sensitivity to anti-estrogen therapies. With this preclinical knowledge, the Breast cancer trial of OraL EveROlimus-2 (BOLERO-2), which is a randomized, multicenter phase III trial was conducted to evaluate the benefit of combining steroidal AI, AROMASIN® (Exemestane) and an mTOR inhibitor AFINITOR®, for treatment of postmenopausal patients with HR+ and HER2 negative advanced breast cancer, who had either recurrent or progressive disease after non steroidal AI’s. Seven hundred and twenty four (N=724) patients were randomly assigned in a 2:1 ratio to receive either a combination of a steroidal AI, AROMASIN® at 25 mg PO QD and AFINITOR® at 10 mg PO QD (N=485) or AROMASIN® along with a placebo (N=239). Both treatment groups were well balanced and patients were stratified according to sensitivity to previous hormonal therapy and presence of visceral metastases. The primary endpoint for this study was Progression Free Survival (PFS) and secondary endpoints included overall Response Rate (RR), Clinical Benefit Rate (CBR defined as complete response + partial response + stable disease for at least 6 months), Overall Survival, Quality of Life, changes in bone marker levels and patient safety. In this study, close to 60% of the patients had visceral disease and approximately 80% of the patients had prior therapies for metastatic disease, with only 20% receiving the study drugs as their first therapy for metastatic disease. The combination of AFINITOR® and AROMASIN® significantly prolonged PFS compared to AROMASIN® alone (11 vs 4.1 months, HR=0.38, P<0.0001) and the Clinical Benefit Rate was 51% in the combination group and 26% in the AROMASIN® alone group (P<0.0001). The combination of AFINITOR® and AROMASIN® benefitted all subgroups of patients including those who had disease recurrence during or after neoadjuvant/adjuvant non steroidal AI therapy, those with visceral and bone metastases, as well as those who had prior chemotherapy. The most common adverse events in all age groups were rash, stomatitis, fatigue, diarrhea and nausea and these toxicities were manageable. The authors concluded that AFINITOR® given along with AROMASIN® in the study population can decrease the risk of disease progression by 60% and can double the Clinical Benefit Rate, compared to AROMASIN® alone, even in patients 65 years of age or older, thereby delaying the need for chemotherapy. Beck JT, Hortobagyi GN, Campone M, et al. Breast Cancer Res Treat. 2014; 143: 459–467

Yoga Can Improve Sense of Well Being in Breast Cancer Survivors

It appears that Yoga can substantially reduce Inflammation, Fatigue and improve Vitality in breast cancer survivors. This was substantiated in a randomized trial, which enrolled breast cancer patients following local intervention and adjuvant chemotherapy. The authors were also able to measure and demonstrate a drop in the cytokines associated with inflammation, such as Interleukin-6 (IL-6), Interleukin-1beta (IL-1b) and Tumor Necrosis Factor-alfa (TNFa), with Yoga intervention. These interesting and intriguing findings were published in the Journal of Clinical Oncology. A summary of this study is available to review at www.oncoprescribe.com.

Yoga’s Impact on Inflammation, Mood, and Fatigue in Breast Cancer Survivors A Randomized Controlled Trial

SUMMARY: Breast cancer is the most common cancer among women in the US and about 1 in 8 women (12%), will develop invasive breast cancer during their lifetime. Approximately, 233,000 new cases of invasive breast cancer will be diagnosed in 2014 and 40,000 women will die of the disease. According to the SEER database, the 5 year survival of women with early stage breast cancer is well over 90%. This is mainly because of early detection and effective therapies. A third of these cancer survivors however are likely to be physically inactive due to fatigue, general deconditioning and effects of cancer treatment. There is a reduction in their cardiorespiratory fitness. Chronic inflammation has been implicated in decreased physical functioning, disability and mortality, even in those who are in remission. Regular exercise has been shown to reduce inflammation and fatigue. The authors in this study evaluated the impact of Yoga, which provides graded exercise, on inflammation, mood and fatigue. Yoga by definition is a physical, mental, and spiritual practice and hatha Yoga which is practiced in the Western countries, consists of physical and mental strength building exercises and postures. In this randomized controlled study, 200 breast cancer survivors between ages 27 and 76 years, were assigned to either the Yoga group (N=100) or control/no Yoga group (N=100). Both groups were well balanced with no significant difference between the groups in measures of activity, fatigue, body mass index or depressive symptoms. Enrolled patients were stratified by cancer stage and whether radiation therapy was given or not. Participants had Stage 0 – Stage IIIa breast cancer and had completed cancer treatment within the past three years and were at least 2 months post surgery, adjuvant chemotherapy or radiation treatment. Patients in the Yoga group participated in a 90 minute Yoga class, twice a week for 12 weeks (3 months). The protocol included the measurement of cytokines associated with inflammation and they included Interleukin-6 (IL-6), Interleukin-1beta (IL-1b) and Tumor Necrosis Factor-alfa (TNFa). In addition, other validated instruments were utilized to measure Fatigue and Vitality. Immediately following 12 weeks of Yoga intervention, there was a significant improvement in the Vitality score in the Yoga group (P=0.01) and at 3 months post intervention, the Vitality score was even higher (P=0.01). Fatigue was significantly lower 3 months post intervention, in the Yoga group (P=0.002). At 3 months post intervention, all inflammatory cytokines were lower as well, in the Yoga group – IL-6 (P=0.027), TNFa (P=0.027) and IL-1b (P=0.037). More frequent Yoga practice resulted in greater benefits, with improved sleep and decreased inflammation. It should be noted that sleep problems are 2-3 times more common in cancer survivors and close to two thirds of the cancer survivors experience insomnia. The authors concluded that this is the first and largest randomized controlled study that has demonstrated that practice of Yoga or similar such activities, can substantially reduce inflammation and Fatigue and improve Vitality in breast cancer survivors. Kiecolt-Glaser JK, Bennett JM, Andridge R, et al. J Clin Oncol 2014;32:1040-1049

Chemotherapy for isolated locoregional recurrence of breast cancer (CALOR) a randomised trial

SUMMARY:It has been well recognized that patients with Breast Cancer with Isolated LocoRegional Recurrences (ILRR), without evidence of distant metastasis, are at a high risk of developing subsequent distant metastasis and may have poor outcomes, in spite of surgical resection of the ILRR. The role of systemic chemotherapy following surgery in this patient population has remained unclear although hormonal intervention in ER positive patients demonstrated disease free survival benefit. To address this question, three cooperative groups, The International Breast Cancer Study Group (IBCSG), Breast International Group (BIG) and NSABP collaborated and conducted this study to find out whether adjuvant chemotherapy improves the outcome of patients with ILRR following surgical resection. In this multicenter trial, 162 patients were randomly assigned to receive chemotherapy (N=85) or no chemotherapy (N=77). Eligible patients had histologically proven first ILRR resected and radiotherapy was recommended for all patients, but was mandated for those with microscopically involved surgical margins. All patients with ER or PR positive recurrent tumors received endocrine therapy and patients randomized to the chemotherapy group received at least two standard cytotoxic drugs (investigators choice) for 3-6 months. Both treatment groups were well balanced, and hormone receptor status was positive in 68% of the patients belonging to each group. The primary endpoint was Disease Free Survival and secondary endpoints included Overall Survival. The five year Disease Free Survival in the adjuvant chemotherapy group was 69% compared with 57% in the no chemotherapy group (HR=0.59, P=0.046). The five year overall survival was also significantly longer in the chemotherapy group compared to the no chemotherapy group (88% vs 76%, HR=0.41, P=0.024). With regards to the chemotherapy benefit based on hormone receptor status, patients with hormone receptor negative ILRR, had a longer Disease Free Survival with adjuvant chemotherapy (P=0.046). The authors concluded that adjuvant chemotherapy significantly improves Disease Free and Overall Survival in Breast Cancer patients with ILRR. This benefit is even more so, in those with ER negative ILRR. Adjuvant chemotherapy should therefore be considered in this patient population. Aebi S, Gelber S, Anderson SJ, et al. The Lancet Oncology 2014;15:156-163

Rapid Increase in Breast Magnetic Resonance Imaging Use Trends From 2000 to 2011

SUMMARY: Breast cancer is the most common cancer among women in the United States and 1 in 8 women will develop invasive breast cancer during their lifetime. Screening mammography complemented by breast self exam and clinical breast exam has resulted in early detection of breast cancer and successful outcomes. Even though mammography is a sensitive screening test, a small percentage of breast cancers may not show up on mammograms but may be palpable on examination by the patient or the clinician. Further, mammograms are less likely to find breast tumors in younger women with dense breast tissue. A breast Magnetic Resonance Imaging (MRI) is more sensitive than mammography although the specificity of a breast MRI is lower, resulting in a higher rate of false-positive findings and potentially unnecessary biopsies. Microcalcifications in the breast can be missed by a breast MRI. Taking these factors into consideration, appropriate utilization of breast MRI becomes relevant. The American Cancer Society (ACS) recommends an annual MRI as an adjunct to screening mammogram and clinical breast exam in certain groups with increased risk of breast cancer (See Figures). The authors conducted this study to determine the utilization of breast (MRI) in community settings. In this retrospective study, the authors reviewed data on 10,518 women from a not-for-profit health plan, and these women had at least one breast MRI between January 2000 and December 2011. The appropriateness of a breast MRI was determined using a prediction model obtained from electronic medical records on a subset of patients. Between 2000 and 2009, there was a 20-fold increase in the use of breast MRI from 6.5 per 10,000 women in the year 2000 to 130.7 per 10,000 in 2009. This increase then declined and stabilized to 104.8 per 10,000 by 2011. With regards to indications for a breast MRI, 51.7% had a family history of breast cancer, 30.1% had a personal history of breast cancer and 3.5% of women had a documented genetic mutation. It is interesting to note that of those who received screening or surveillance breast MRI’s, only 21% fulfilled the American Cancer Society (ACS) criteria for a breast MRI. Conversely, fewer participants (48.4%), with documented deleterious genetic mutations, received breast MRI screening. The authors concluded that breast MRI was over utilized in those who did not fit the ACS criteria and was under utilized in those with documented genetic mutations. It is their opinion that routine breast MRI screening is not recommended for a new breast cancer diagnosis or for breast cancer surveillance and should only be considered for the group of individuals who have the most benefit. Breast MRI is performed preferably between days 7-15 of menstrual cycle for premenopausal women, using a dedicated breast coil, with the ability to perform a biopsy under MRI guidance by experienced radiologists, during the same visit. Stout NK, Nekhlyudov L, Li L, et al. JAMA Intern Med. 2014;174:114-121.

Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere) a randomised multicentre, open-label, phase 2 trial

SUMMARY: The FDA on September 30, 2013 granted accelerated approval to PERJETA® (Pertuzumab) for use in combination with HERCEPTIN® (Trastuzumab) and other chemotherapy for the neoadjuvant (preoperative) treatment of patients with HER2-positive, locally advanced, inflammatory, or early stage breast cancer (either greater than 2 cm in diameter or with positive lymph nodes), as part of a complete treatment regimen for early breast cancer. Following surgery, patients should continue to receive HERCEPTIN® to complete one year of treatment. The HER family of receptors consist of HER1, HER2, HER3 and HER4. These proteins are transmembrane tyrosine kinases and are involved in normal cell growth and differentiation. HER1 is also known as Epidermal Growth Factor Receptor or EGFR. These receptors are activated following ligand binding, receptor pairing or dimerization and phosphorylation. This dimerization (receptor pairing) occurs often within the HER family of receptors. This has been no ligand identified for HER2 receptor, although it is able to form homo and heterodimers with other members of the HER family readily. Dimerization of HER2 and HER3 is believed to produce the strongest mitogenic signaling and activates two important pathways that regulate cell survival and growth – Mitogen Activated Protein Kinase (MAPK) pathway and PhosphoInositide 3-Kinase (PI3K) pathway. For this reason inhibiting HER2 dimerization appears to be an important step in the treatment of cancer. Overexpression of HER2 in breast cancer has been associated with higher risk for relapse as well as overall survival. Approximately 20 percent of breast cancers are HER2-positive. HERCEPTIN® is a humanized monoclonal antibody targeting HER2. It binds to the extracellular subdomain IV of the receptor and disrupts ligand independent HER2 downstream cell signaling pathways. PERJETA® is a recombinant, humanized, monoclonal antibody that binds to the HER2 subdomain II and blocks ligand dependent HER2 heterodimerization with other HER receptors, ie. HER3, HER1 and HER4. Thus HERCEPTIN® along with PERJETA® provide a more comprehensive blockade of HER2 driven signaling pathways. The accelerated approval of PERJETA® for the neoadjuvant treatment of breast cancer was based on a randomized, multicenter, open-label, phase II trial, in which 417 patients with HER2-positive, operable, locally advanced or inflammatory breast cancer (T2-4d), were randomly assigned to receive preoperative therapy with either HERCEPTIN® plus TAXOTERE® (Docetaxel), PERJETA® plus HERCEPTIN® and TAXOTERE®, PERJETA® plus HERCEPTIN® or PERJETA® plus TAXOTERE®. Patients in the three drug group received preoperative therapy with PERJETA®, HERCEPTIN® and TAXOTERE® every 3 weeks for a total of 4 cycles and following surgery, all patients received 3 cycles of Fluorouracil, ELLENCE® (Epirubicin), and CYTOXAN® (Cyclophosphamide) – (FEC) IV every 3 weeks and HERCEPTIN® was continued every 3 weeks for a total of one year of therapy. The primary endpoint was pathological Complete Response (pCR) rate defined as the absence of invasive cancer in the breast. The FDA definition of pCR is the absence of invasive cancer in the breast and lymph nodes. All treatment groups were well balanced. Seven percent of patients had inflammatory breast cancer, 32% had locally advanced cancer and 70% had clinically node-positive breast cancer. Forty-seven percent of the patients had hormone receptor-positive disease. The FDA defined pCR rates were 39.3% in the PERJETA® plus HERCEPTIN® and TAXOTERE® group and 21.5% in the HERCEPTIN® plus TAXOTERE® group (P=0.0063). Of Interest, the pCR rates in the three drug group were lower in patients with hormone receptor positive tumors compared to patients with hormone receptor negative tumors. The most common adverse events in the three drug group were alopecia, diarrhea, nausea and neutropenia. Other significant side effects included decreased cardiac function, infusion-related reactions, hypersensitivity reactions and anaphylaxis. Based on clinical studies, for the neoadjuvant treatment of breast cancer, PERJETA® should be administered every 3 weeks for 3 to 6 cycles as part of one of the following treatment regimens for early breast cancer. • Four preoperative cycles of PERJETA® in combination with HERCEPTIN® and TAXOTERE® followed by 3 postoperative cycles of Fluorouracil, ELLENCE® and CYTOXAN® (FEC). • Three preoperative cycles of FEC alone followed by 3 preoperative cycles of PERJETA® in combination with TAXOTERE® and HERCEPTIN®. • Six preoperative cycles of PERJETA® in combination with TAXOTERE®, Carboplatin, and HERCEPTIN® (TCH). Following surgery, patients should continue to receive HERCEPTIN® to complete 1 year of treatment. The accelerated approval by the FDA was based solely on the improved pCR rate with the three drug combination with no demonstrable improvement in event-free survival or overall survival. A confirmatory phase III trial is underway, with results expected in 2016. Gianni L, Pienkowski T, Im YH, et al. Lancet Oncol. 2012;13:25-32

Effects of bisphosphonate treatment on recurrence and cause-specific mortality in women with early breast cancer a meta-analysis of individual patient data from randomised trials

SUMMARY: Bisphosphonates are presently indicated for the treatment of osteoporosis. The approved bisphosphonates in the U.S. include FOSAMAX® (Alendronate), BONIVA® (Ibandronate), ACTONEL® (Risedronate) and RECLAST® (Zoledronic acid). Several trials conducted over the past 2 decades have suggested that bisphosphonates may have anti proliferative effect in patients with breast cancer. Data from several Women's Health Initiative (WHI) studies involving more than 150,000 healthy postmenopausal women, of whom 2216 used oral bisphosphonates, revealed that women taking bisphosphonates for osteoporosis had a 32% reduction in invasive breast cancer. The AZURE investigators conducted a study to determine whether the addition of RECLAST® (Zoledronic acid) to standard adjuvant therapy would improve disease outcomes in patients with early-stage breast cancer. They noted that in the subset analysis, the addition of RECLAST® significantly improved disease free survival and overall survival in postmenopausal patients, independent of estrogen receptor status, tumor stage, and lymph node involvement (N Engl J Med 2011;365:1396-1405). With this background, the authors belonging to the Early Breast Cancer Trialists’ Collaborative Group (EBCTCG), conducted a meta-analysis and reviewed data from 15 years of bisphosphonate trials, which included 36 trials of adjuvant bisphosphonates in breast cancer and involved over 17,000 pre and postmenopausal women. RECLAST® (Zoledronic acid) and Clodronate were the most common bisphosphonates used in these trials. The primary outcomes analyzed were time to distant recurrence, local recurrence, new second primary breast cancer (ipsilateral or contralateral), time to first distant recurrence (ignoring any previous locoregional or contralateral recurrences), and breast cancer mortality. Planned subset analyses included site of recurrence, site of first distant metastasis (bone vs other), menopausal status (pre, peri and post) type of bisphosphonate (aminobisphosphonates such as RECLAST® or Clodronate) and drug schedule of bisphosphonate therapy (for bone protection vs advanced cancer). Adjuvant bisphosphonates resulted in a 34% reduction in the risk of bone recurrence (P = 0.00001) and a 17% reduction in the risk of breast cancer death (P =0.004). This benefit was seen regardless of estrogen receptor status, nodal status or whether chemotherapy was used or not. Bisphosphonates had no significant impact on non-breast cancer related deaths, contralateral breast cancer or loco-regional recurrence. In this meta-analysis, all these benefits were only seen in postmenopausal women and premenopausal women had no benefit on any disease outcomes with bisphosphonates. The authors emphasized that low estrogen environment as is seen in postmenopausal women, or women rendered menopausal by suppression of ovarian function is a prerequisite for adjuvant bisphosphonate activity. Based on this large meta-analysis, the authors recommended the use of RECLAST® once every six months or oral Clodronate, where available. Because of paucity of data, they do not recommend the use of weekly dose of oral bisphosphonates, often used to prevent osteoporosis, to achieve these benefits. Coleman R, Gnant M, Paterson A, et al. San Antonio Breast Cancer Symposium 2013; San Antonio, TX. Abstract S4-07.