Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Breast Cancer Res Treat ; 167(1): 107-116, 2018 01.
Article in English | MEDLINE | ID: mdl-28918548

ABSTRACT

BACKGROUND: Methylated gene markers have shown promise in predicting breast cancer outcomes and treatment response. We evaluated whether baseline and changes in tissue and serum methylation levels would predict pathological complete response (pCR) in patients with HER2-negative early breast cancer undergoing preoperative chemotherapy. METHODS: The TBCRC008 trial investigated pCR following 12 weeks of preoperative carboplatin and albumin-bound paclitaxel + vorinostat/placebo (n = 62). We measured methylation of a 10-gene panel by quantitative multiplex methylation-specific polymerase chain reaction and expressed results as cumulative methylation index (CMI). We evaluated association between CMI level [baseline, day 15 (D15), and change] and pCR using univariate and multivariable logistic regression models controlling for treatment and hormone receptor (HR) status, and performed exploratory subgroup analyses. RESULTS: In univariate analysis, one log unit increase in tissue CMI levels at D15 was associated with 40% lower chance of obtaining pCR (odds ratio, OR 0.60, 95% CI 0.37-0.97; p = 0.037). Subgroup analyses suggested a significant association between tissue D15 CMI levels and pCR in vorinostat-treated [OR 0.44 (0.20, 0.93), p = 0.03], but not placebo-treated patients. CONCLUSION: In this study investigating the predictive roles of tissue and serum CMI levels in patients with early breast cancer for the first time, we demonstrate that high D15 tissue CMI levels may predict poor response. Larger studies and improved analytical procedures to detect methylated gene markers in early stage breast cancer are needed. TBCRC008 is registered on ClinicalTrials.gov (NCT00616967).


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/drug therapy , DNA Methylation/drug effects , Hydroxamic Acids/administration & dosage , Adult , Aged , Breast Neoplasms/blood , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA, Neoplasm/adverse effects , DNA, Neoplasm/blood , Female , Gene Expression Regulation, Neoplastic , Humans , Hydroxamic Acids/adverse effects , Middle Aged , Neoplasm Proteins/genetics , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Preoperative Period , Vorinostat
2.
Breast Cancer Res Treat ; 146(3): 557-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25001612

ABSTRACT

Nearly half of patients with advanced triple negative breast cancer (TNBC) develop brain metastases (BM) and most will also have uncontrolled extracranial disease. This study evaluated the safety and efficacy of iniparib, a small molecule anti-cancer agent that alters reactive oxygen species tumor metabolism and penetrates the blood brain barrier, with the topoisomerase I inhibitor irinotecan in patients with TNBC-BM. Eligible patients had TNBC with new or progressive BM and received irinotecan and iniparib every 3 weeks. Time to progression (TTP) was the primary end point; secondary endpoints were response rate (RR), clinical benefit rate (CBR), overall survival (OS), toxicity, and health-related quality of life. Correlative endpoints included molecular subtyping and gene expression studies on pre-treatment archival tissues, and determination of germline BRCA1/2 status. Thirty-seven patients began treatment; 34 were evaluable for efficacy. Five of 24 patients were known to carry a BRCA germline mutation (4 BRCA1, 1 BRCA2). Median TTP was 2.14 months and median OS was 7.8 months. Intracranial RR was 12 %, while intracranial CBR was 27 %. Treatment was well-tolerated; the most common grade 3/4 adverse events were neutropenia and fatigue. Grade 3/4 diarrhea was rare (3 %). Intrinsic subtyping revealed 19 of 21 tumors (79 %) were basal-like, and intracranial response was associated with high expression of proliferation-related genes. This study suggests a modest benefit of irinotecan plus iniparib in progressive TNBC-BM. More importantly, this trial design is feasible and lays the foundation for additional studies for this treatment-refractory disease.


Subject(s)
Benzamides/administration & dosage , Brain Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Triple Negative Breast Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzamides/adverse effects , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Camptothecin/administration & dosage , Camptothecin/adverse effects , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Irinotecan , Middle Aged , Neoplasm Staging , Receptor, ErbB-2/genetics , Survival Analysis , Triple Negative Breast Neoplasms/pathology , Vascular Endothelial Growth Factor A/biosynthesis
3.
Curr Treat Options Oncol ; 15(1): 55-62, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24338340

ABSTRACT

OPINION STATEMENT: Estrogen-dependent growth of some breast cancers was a key observation, which led to the development of tamoxifen and aromatase inhibitors (AIs). Tamoxifen and AIs have different modes of action and side-effect profiles. Based on evidence, both in laboratory models and clinical trials, longer duration of hormone suppression therapy is beneficial. The most important factor deciding their use is "menopausal status." Sometimes, defining menopause might be challenging in clinical practice. Measuring serum follicle stimulating hormone (FSH) and estradiol levels are helpful when in doubt. Tamoxifen should be offered to those women with normal FSH and estradiol levels even with cessation of menstruation. Once menopause is defined, it is relatively clear to decide about the endocrine therapy. Premenopausal women should be treated with tamoxifen and postmenopausal women with AIs. Perimenopausal women should be treated with tamoxifen initially and later switched to AIs once they become postmenopausal. With current recent evidence, premenopausal women should be treated with 10 years of tamoxifen. Current evidence also supports 5 years of an AI alone or 5 years of tamoxifen followed by 5 years of an AI; studies evaluating longer duration of AI treatment are in progress (Figure 1). Compliance with long-term use of these adjuvant endocrine therapies depends on screening for and management of side effects. Patients taking tamoxifen should be clinically screened for thromboembolism and for endometrial cancer if abnormal bleeding occurs. Patients on AI should pay careful attention to management of other chronic health disorders. They also should be screened for optimal bone health. Management of vasomotor symptoms also helps with adherence to long-term treatment for both tamoxifen and AIs.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Drug Substitution , Female , Humans , Neoplasm Staging , Practice Guidelines as Topic , Risk Factors , Tamoxifen/administration & dosage , Tamoxifen/therapeutic use
4.
J Clin Oncol ; 30(26): 3242-9, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22649152

ABSTRACT

PURPOSE: Neoadjuvant chemotherapy for breast cancer provides critical information about tumor response; how best to leverage this for predicting recurrence-free survival (RFS) is not established. The I-SPY 1 TRIAL (Investigation of Serial Studies to Predict Your Therapeutic Response With Imaging and Molecular Analysis) was a multicenter breast cancer study integrating clinical, imaging, and genomic data to evaluate pathologic response, RFS, and their relationship and predictability based on tumor biomarkers. PATIENTS AND METHODS: Eligible patients had tumors ≥ 3 cm and received neoadjuvant chemotherapy. We determined associations between pathologic complete response (pCR; defined as the absence of invasive cancer in breast and nodes) and RFS, overall and within receptor subsets. RESULTS: In 221 evaluable patients (median tumor size, 6.0 cm; median age, 49 years; 91% classified as poor risk on the basis of the 70-gene prognosis profile), 41% were hormone receptor (HR) negative, and 31% were human epidermal growth factor receptor 2 (HER2) positive. For 190 patients treated without neoadjuvant trastuzumab, pCR was highest for HR-negative/HER2-positive patients (45%) and lowest for HR-positive/HER2-negative patients (9%). Achieving pCR predicted favorable RFS. For 172 patients treated without trastuzumab, the hazard ratio for RFS of pCR versus no pCR was 0.29 (95% CI, 0.07 to 0.82). pCR was more predictive of RFS by multivariate analysis when subtype was taken into account, and point estimates of hazard ratios within the HR-positive/HER2-negative (hazard ratio, 0.00; 95% CI, 0.00 to 0.93), HR-negative/HER2-negative (hazard ratio, 0.25; 95% CI, 0.04 to 0.97), and HER2-positive (hazard ratio, 0.14; 95% CI, 0.01 to 1.0) subtypes are lower. Ki67 further improved the prediction of pCR within subsets. CONCLUSION: In this biologically high-risk group, pCR differs by receptor subset. pCR is more highly predictive of RFS within every established receptor subset than overall, demonstrating that the extent of outcome advantage conferred by pCR is specific to tumor biology.


Subject(s)
Biomarkers/analysis , Breast Neoplasms/drug therapy , Treatment Outcome , Breast Neoplasms/metabolism , Diagnostic Imaging , Disease-Free Survival , Humans , Middle Aged , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Recurrence
5.
Breast Cancer Res Treat ; 132(3): 1049-62, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22198468

ABSTRACT

Neoadjuvant chemotherapy for breast cancer allows individual tumor response to be assessed depending on molecular subtype, and to judge the impact of response to therapy on recurrence-free survival (RFS). The multicenter I-SPY 1 TRIAL evaluated patients with ≥ 3 cm tumors by using early imaging and molecular signatures, with outcomes of pathologic complete response (pCR) and RFS. The current analysis was performed using data from patients who had molecular profiles and did not receive trastuzumab. The various molecular classifiers tested were highly correlated. Categorization of breast cancer by molecular signatures enhanced the ability of pCR to predict improvement in RFS compared to the population as a whole. In multivariate analysis, the molecular signatures that added to the ability of HR and HER2 receptors, clinical stage, and pCR in predicting RFS included 70-gene signature, wound healing signature, p53 mutation signature, and PAM50 risk of recurrence. The low risk signatures were associated with significantly better prognosis, and also identified additional patients with a good prognosis within the no pCR group, primarily in the hormone receptor positive, HER-2 negative subgroup. The I-SPY 1 population is enriched for tumors with a poor prognosis but is still heterogeneous in terms of rates of pCR and RFS. The ability of pCR to predict RFS is better by subset than it is for the whole group. Molecular markers improve prediction of RFS by identifying additional patients with excellent prognosis within the no pCR group.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Adult , Aged , Anthracyclines/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Clinical Trials as Topic , Disease-Free Survival , Female , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Middle Aged , Multicenter Studies as Topic , Multivariate Analysis , Neoplasm, Residual , Proportional Hazards Models , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Taxoids/administration & dosage , Trastuzumab
6.
Clin Breast Cancer ; 10(4): 275-80, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20705559

ABSTRACT

INTRODUCTION: Tumor content or expression of vascular endothelial growth factor (VEGF) is associated with impaired efficacy of antiestrogen adjuvant therapy. We designed a pilot study to assess the feasibility and short-term efficacy of neoadjuvant letrozole and bevacizumab (anti-VEGF) in postmenopausal women with stage II and III estrogen receptor/progesterone receptor-positive breast cancer. PATIENTS AND METHODS: Patients were treated with a neoadjuvant regimen of letrozole orally 2.5 mg/day and bevacizumab intravenously 15 mg/kg every 3 weeks for a total of 24 weeks before the surgical treatment of their breast cancer. Patients were followed for toxicity at 3-week intervals, and tumor assessment (a physical examination and ultrasound) was performed at 6-week intervals. Positron emission tomography (PET) scans were performed before therapy and 6 weeks after the initiation of therapy. RESULTS: Twenty-five evaluable patients were treated. The regimen was well-tolerated, except in 2 patients who were taken off the study for difficulties controlling their hypertension. An objective clinical response occurred in 17 of 25 patients (68%), including 16% complete responses (CRs) and 52% partial responses. The 4 patients with clinical CRs manifested pathologic CRs in their breasts (16%), although 1 patient had residual tumor cells in her axillary nodes. Eight of 25 patients (32%) attained stage 0 or 1 status. The PET scan response at 6 weeks correlated with clinical CRs and breast pathologic CRs at 24 weeks (P < .0036). CONCLUSION: Combination neoadjuvant therapy with letrozole and bevacizumab was well-tolerated and resulted in impressive clinical and pathologic responses. The Translational Breast Cancer Research Consortium has an ongoing randomized phase II trial of this regimen in this patient population.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Neoadjuvant Therapy/methods , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Bevacizumab , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Letrozole , Middle Aged , Neoplasm Staging , Nitriles/administration & dosage , Nitriles/adverse effects , Pilot Projects , Positron-Emission Tomography , Postmenopause , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/genetics , Triazoles/administration & dosage , Triazoles/adverse effects
8.
J Clin Oncol ; 21(6): 976-83, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12637460

ABSTRACT

PURPOSE: This study was designed to determine whether increasing the dose of doxorubicin in or adding paclitaxel to a standard adjuvant chemotherapy regimen for breast cancer patients would prolong time to recurrence and survival. PATIENTS AND METHODS: After surgical treatment, 3,121 women with operable breast cancer and involved lymph nodes were randomly assigned to receive a combination of cyclophosphamide (C), 600 mg/m(2), with one of three doses of doxorubicin (A), 60, 75, or 90 mg/m(2), for four cycles followed by either no further therapy or four cycles of paclitaxel at 175 mg/m(2). Tamoxifen was given to 94% of patients with hormone receptor-positive tumors. RESULTS: There was no evidence of a doxorubicin dose effect. At 5 years, disease-free survival was 69%, 66%, and 67% for patients randomly assigned to 60, 75, and 90 mg/m(2), respectively. The hazard reductions from adding paclitaxel to CA were 17% for recurrence (adjusted Wald chi(2) P =.0023; unadjusted Wilcoxon P =.0011) and 18% for death (adjusted P =.0064; unadjusted P =.0098). At 5 years, the disease-free survival (+/- SE) was 65% (+/- 1) and 70% (+/- 1), and overall survival was 77% (+/- 1) and 80% (+/- 1) after CA alone or CA plus paclitaxel, respectively. The effects of adding paclitaxel were not significantly different in subsets defined by the protocol, but in an unplanned subset analysis, the hazard ratio of CA plus paclitaxel versus CA alone was 0.72 (95% confidence interval, 0.59 to 0.86) for those with estrogen receptor-negative tumors and only 0.91 (95% confidence interval, 0.78 to 1.07) for patients with estrogen receptor-positive tumors, almost all of whom received adjuvant tamoxifen. The additional toxicity from adding four cycles of paclitaxel was generally modest. CONCLUSION: The addition of four cycles of paclitaxel after the completion of a standard course of CA improves the disease-free and overall survival of patients with early breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Paclitaxel/administration & dosage , Adult , Aged , Antibiotics, Antineoplastic/administration & dosage , Disease-Free Survival , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Female , Humans , Lymphatic Metastasis , Middle Aged , Proportional Hazards Models , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...