Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
JAMA Oncol ; 5(1): 74-82, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30242306

ABSTRACT

Importance: Atezolizumab (anti-programmed cell death ligand 1 [PD-L1]) is well tolerated and clinically active in multiple cancer types. Its safety and clinical activity in metastatic triple-negative breast cancer (mTNBC) has not been reported. Objective: To evaluate the safety, clinical activity, and biomarkers associated with the use of single-agent atezolizumab in patients with mTNBC. Design, Setting, and Participants: Women with mTNBC (defined by investigator assessment) were enrolled between January 2013 and February 2016 in a multicohort open-label, phase 1 study at US and European academic medical centers. Median follow-up was 25.3 months (range, 0.4-45.6 months). Eligible patients regardless of line of therapy had measurable disease by Response Evaluation Criteria in Solid Tumors, version 1.1; Eastern Cooperative Oncology Group performance status of 0 to 1; and a representative tumor sample for assessment of immune cell (IC) PD-L1 expression. Interventions: Atezolizumab was given intravenously every 3 weeks until unacceptable toxic effects or loss of clinical benefit. Main Outcomes and Measures: Primary outcome was safety and tolerability. Activity and exploratory outcomes included objective response rate (ORR), duration of response, progression-free survival (PFS), and overall survival (OS). Outcomes were assessed in all patients and in key patient subgroups. Results: Among 116 evaluable patients (median age, 53 years [range, 29-82 years]), treatment-related adverse events occurred in 73 (63%); 58 (79%) were grade 1 to 2. Most adverse events occurred within the first treatment year. The ORRs were numerically higher in first-line (5 of 21 [24%]) than in second-line or greater patients (6 of 94 [6%]). Median duration of response was 21 months (range, 3 to ≥38 months). Median PFS was 1.4 (95% CI, 1.3-1.6) months by RECIST and 1.9 (95% CI, 1.4-2.5) months by irRC. In first-line patients, median OS was 17.6 months (95% CI, 10.2 months to not estimable). Patients with PD-L1 expression of at least 1% tumor-infiltrating ICs had higher ORRs and longer OS (12% [11 of 91]; 10.1 [95% CI, 7.0-13.8] months, respectively) than those with less than 1% ICs (0 of 21; 6.0 [95% CI, 2.6-12.6] months, respectively). High levels of ICs (>10%) were independently associated with higher ORRs and longer OS. Conclusions and Relevance: Single-agent atezolizumab was well tolerated and provided durable clinical benefit in patients with mTNBC with stable or responding disease and in earlier lines of treatment. Trial Registration: ClinicalTrials.gov identifier: NCT01375842.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , B7-H1 Antigen/immunology , Disease Progression , Europe , Female , Humans , Middle Aged , Neoplasm Metastasis , Progression-Free Survival , Time Factors , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , United States
2.
Oncologist ; 20(1): 37-44, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25492923

ABSTRACT

PURPOSE: This study evaluated age-related changes in pharmacokinetic and pharmacodynamic parameters of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) in patients with metastatic breast cancer. METHODS: Forty patients received nab-paclitaxel (100 mg/m(2) weekly for 3 weeks followed by a 1-week break) as first- or second-line chemotherapy. Blood samples were collected for analysis, and response was assessed every two cycles. Planned statistical analyses included linear regression to examine the relationship between age and pharmacokinetic variables (ln clearance [CL] and ln area under the curve [AUC]) and two-sided two-sample t tests to evaluate age differences in pharmacodynamic variables. The association between chemotherapy toxicity risk scores and pharmacokinetic and pharmacodynamic variables including grade ≥ 3 toxicity were examined post hoc. RESULTS: Of 40 patients enrolled, 39 (98%) were evaluable (mean age: 60 years; range: 30-81 years). A partial response was achieved in 31%, and 38% had stable disease. There was a borderline positive association between age and 24-hour ln AUC (slope = 0.011; SE = 0.006; p = .055). Grade 3 toxicity was experienced by 26% (8% hematologic, 18% nonhematologic). There were no differences in age based on the presence of grade 3 toxicity (p = .75), dose reductions (p = .38), or dose omissions (p = .15). A significant association was noted between chemotherapy toxicity risk score category and presence of grade 3 toxicity (toxicity rate by risk score category: low, 5 of 30 patients; medium, 3 of 6 patients; high, 2 of 3 patients; p = .041). CONCLUSION: A borderline significant relationship exists between age and 24-hour AUC, but no differences were noted for pharmacodynamic variables (grade 3 toxicity, dose reductions, or dose omissions) based on age. There is an association between toxicity risk score and grade ≥ 3 chemotherapy toxicity and pharmacokinetic variables. The treatment is well tolerated across all age groups.


Subject(s)
Breast Neoplasms/drug therapy , Nanoparticles/administration & dosage , Paclitaxel/administration & dosage , Adult , Aged , Aged, 80 and over , Albumins/administration & dosage , Albumins/adverse effects , Albumins/chemistry , Breast Neoplasms/blood , Breast Neoplasms/pathology , Female , Humans , Middle Aged , Nanoparticles/adverse effects , Nanoparticles/chemistry , Neoplasm Metastasis , Paclitaxel/adverse effects , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics
3.
Clin Breast Cancer ; 14(2): 94-100, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24370210

ABSTRACT

PURPOSE: We investigated the efficacy and toxicity of sorafenib, a multikinase inhibitor of vascular endothelial growth factor receptor tyrosine kinase, in combination with vinorelbine therapy in a phase I/II trial in patients with metastatic breast cancer. PATIENTS AND METHODS: We enrolled 11 patients in the phase I portion to determine the maximum tolerated dose (MTD) of the combination, followed by 35 extra patients treated at the MTD in phase II. The median age of patients was 54 years old (range, 31-72 years old). Tumors were estrogen receptor and progesterone receptor (ER/PR) positive in 54% (22/54) of patients, and triple negative (ER(-), PR(-), HER2(-)) in 41% (17/54) of patients. Of all patients, 22% received sorafenib and vinorelbine as first-line therapy, 37% as second-line therapy, and 41% as third-line therapy. RESULTS: In total, 41 patients were treated at the MTD (6 during phase I; 35 in phase II). The observed 44% 4-month progression-free survival rate was similar to the estimated historical rate of 43% with vinorelbine treatment. The combination was tolerated with expected toxicities. Patients treated at the MTD who had received prior bevacizumab treatment received a median of 1.5 cycles (range, 1-10 cycles) compared with a median of 5 cycles (range, 2-12 cycles) for patients without prior bevacizumab treatment. CONCLUSION: Further evaluation of vinorelbine and sorafenib in bevacizumab-naive patients may be of interest if specific biomarkers guiding patient selection can be identified.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/secondary , Adult , Aged , Breast Neoplasms/mortality , Female , Follow-Up Studies , Humans , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Prognosis , Sorafenib , Survival Rate , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinorelbine
4.
J Natl Compr Canc Netw ; 11(10): 1193-8, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24142820

ABSTRACT

Delays between presentation and treatment could have a significant effect on breast cancer mortality. The authors hypothesized that patient, physician, and system barriers are all responsible for treatment delays. Therefore, a study was conducted to define prevalent barriers to treatment from the patient's perspective. A modified 43-item Likert-scale questionnaire was administered to patients with clinical stage III locally advanced breast cancer (LABC) who had experienced a delay in treatment of 3 months or more. Between October 2008 and January 2010, 153 patients presented with LABC; 43 patients (28.1%) met eligibility, and 40 completed the questionnaire. Among the patient barriers reported, 38% of patients delayed care for fear of losing their breast and 47% awaited previously scheduled routine appointments instead of seeking care. Among the physician barriers reported, 20% of physicians of initial contact did not believe the breast lump/symptom was related to cancer and 15% did not believe it needed a biopsy. Among the system barriers reported, the most prevalent were delays in performing diagnostic tests and obtaining insurance authorization for tests, treatment, or physician visits. Substantial delays were seen in 28.1% of patients from presentation to when they sought therapy at City of Hope Comprehensive Cancer Center. The high prevalence of patient barriers versus physician/system barriers suggests that increased educational efforts for patients and health care professionals are needed.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Adult , Aged , Female , Humans , Middle Aged , Neoplasm Staging , Pilot Projects , Risk Factors
5.
Ann Surg Oncol ; 20(8): 2576-81, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23468047

ABSTRACT

BACKGROUND: There are few data on the long-term outcome of patients with microinvasive (T1mi) breast cancer. Moreover, predictors of lymph node involvement and the impact of multifocal microinvasion are not well understood. METHODS: Patients with T1mi cancer, defined as tumors ≤1 mm, surgically managed at our institute and who underwent axillary lymph node evaluation were identified. Specimen slides were independently reviewed. Multivariate analysis was used to identify factors predictive of lymph node involvement. RESULTS: Forty-five patients with T1mi cancer were identified. Median patient age was 52 years, and median size of in situ disease was 4 cm. Nine tumors (20.0 %) had more than one focus of microinvasion. Lymph nodes metastasis were identified in 9 patients: 1 macrometastasis (2.2 %), 4 micrometastases (8.9 %), and 4 isolated tumor cells (8.9 %). Seven of 9 patients with lymph node involvement underwent adjuvant chemotherapy. Estrogen receptor-negative invasive disease was a significant predictor of lymph node metastasis by multivariable analysis (p < 0.02). There was also a trend toward lymph node involvement in patients with multifocal microinvasion compared to unifocal disease (33.3 vs. 16.7 %, respectively). At a median follow-up of 83 months, 3 patients (6.3 %) had disease recurrence (1 local, 1 distant, 1 local and distant). All patients with recurrence initially had tumor-free lymph nodes and only one focus of microinvasion. CONCLUSIONS: Microinvasive breast cancer clearly has the ability to metastasize and recur, but in this series, only 2 % of patients with nodal macrometastasis. Only two patients experienced local recurrence, neither of whom had lymph node metastasis. The importance of identifying nodal micrometastasis in T1mi disease needs to be further explored.


Subject(s)
Breast Neoplasms/pathology , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/secondary , Lymph Node Excision , Neoplasm Recurrence, Local/pathology , Neoplasms, Multiple Primary/pathology , Adult , Aged , Aged, 80 and over , Axilla , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Carcinoma in Situ/metabolism , Carcinoma in Situ/therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/therapy , Chemotherapy, Adjuvant , Female , Humans , Lymphatic Metastasis , Mastectomy, Segmental , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Micrometastasis , Neoplasms, Multiple Primary/metabolism , Neoplasms, Multiple Primary/therapy , Prognosis , Receptors, Estrogen/metabolism , Recurrence , Sentinel Lymph Node Biopsy
7.
Oncologist ; 16(6): 760-71, 2011.
Article in English | MEDLINE | ID: mdl-21543509

ABSTRACT

Newer treatments have improved survival for patients with metastatic breast cancer over the last two decades, and a battery of new cytotoxic and targeted therapies is continuing to enhance this trend. This review outlines recent data and ongoing research in this area, by highlighting new developments (regarding approved but relatively new classes of cytotoxic and targeted agents) and also new classes of targeted therapy that are undergoing clinical evaluation. Mechanisms for synergy between agents are discussed where data are available, as is information on the rationale behind the development of agents that inhibit angiogenesis, DNA repair, histone deacetylases, heat shock proteins, or various signaling pathways in tumor proliferation. The abundance of clinical research surrounding anticancer agents, together with ongoing cancer biology research, is expected to further increase the available pool of therapeutic options for metastatic breast cancer. Concomitantly, in the absence of an effective targeted monotherapy, a better understanding of the interplay between biologic and cytotoxic anticancer agents will improve our ability to rationally design combination regimens with better efficacy and tolerability.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Clinical Trials as Topic , Drug Synergism , Female , Humans , Molecular Targeted Therapy , Neoplasm Metastasis , Signal Transduction
8.
J Clin Oncol ; 28(25): 3917-21, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20679610

ABSTRACT

PURPOSE: To explore the antitumor activity of the aromatase inhibitor, anastrozole, in the treatment of premenopausal women with hormone receptor-positive, metastatic breast cancer who have been rendered functionally postmenopausal with the use of the luteinizing hormone-releasing hormone agonist, goserelin. PATIENTS AND METHODS: Premenopausal women with estrogen and/or progesterone receptor-positive, metastatic or recurrent breast cancer were enrolled in this prospective, single-arm, multicenter phase II trial. Patients were treated with goserelin 3.6 mg subcutaneous monthly and began anastrozole 1-mg daily 21 days after the first injection of goserelin. Patients continued on treatment until disease progression or unacceptable toxicity. RESULTS: Thirty-five patients were enrolled of which 32 were evaluable for response and toxicity. Estradiol suppression was assessed, with mean estradiol levels of 18.7 pg/mL at 3 months and 14.8 pg/mL at 6 months. One participant (3.1%) experienced a complete response, 11 (34.4%) experienced partial response, and 11 (34.4%) experienced stable disease for 6 months or longer for a clinical benefit rate of 71.9%. Median time to progression was 8.3 months (range, 2.1 to 63+) and median survival was not been reached (range, 11.1 to 63+). The most common adverse events were fatigue (50%), arthralgias (53%), and hot flashes (59%). There were no grade 4 to 5 toxicities. CONCLUSION: The combination of goserelin plus anastrozole has substantial antitumor activity in the treatment of premenopausal women with hormone receptor-positive metastatic breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Goserelin/administration & dosage , Neoplasms, Hormone-Dependent/drug therapy , Nitriles/administration & dosage , Triazoles/administration & dosage , Adult , Anastrozole , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estradiol/metabolism , Female , Humans , Middle Aged , Neoplasm Metastasis , Premenopause
9.
Cancer ; 116(13): 3195-204, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20564623

ABSTRACT

BACKGROUND: Several publications reporting on health disparities document that ethnic minorities disproportionately experience delays in healthcare access, delivery, and treatment. However, few studies examine factors underlying access and receipt of healthcare among cancer survivors from the patient perspective. This study explores diagnostic and therapeutic care delays among a multiethnic sample of breast and cervical cancer survivors and examines contextual factors influencing diagnostic and therapeutic care delays. METHODS: Population-based sampling and a cross-sectional design were used to recruit 1377 survivors (breast cancer, n = 698; cervical cancer, n = 679). This multiethnic sample included 449 European American, 185 African American, 468 Latina American, and 275 Asian American survivors. RESULTS: Latina Americans were more likely to report diagnostic delays (P = .003), whereas African Americans were more likely to report therapeutic delays (P = .007). In terms of cancer type, cervical cancer survivors were more likely to report diagnostic (P = .004) and therapeutic delays (P = .000) compared with breast cancer survivors. "Fear of finding cancer" was the most frequently cited reason for diagnostic delays, and "medical reasons" were most frequently cited for therapeutic delays. CONCLUSIONS: Due in part to a higher proportion of diagnostic and therapeutic delays, ethnic minorities endure greater cancer burden, including poorer survival and survivorship outcomes. The medical community must recognize the impact of existing psychological and cultural dimensions on diagnostic care, as well as the personal and healthcare system level barriers that contribute to therapeutic delays.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Delayed Diagnosis/psychology , Ethnicity , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/therapy , Black or African American/psychology , Aged , Asian/psychology , Attitude to Health , Breast Neoplasms/ethnology , Breast Neoplasms/mortality , Female , Healthcare Disparities , Hispanic or Latino/psychology , Humans , Middle Aged , Patient Acceptance of Health Care , Socioeconomic Factors , Survivors , Uterine Cervical Neoplasms/ethnology , Uterine Cervical Neoplasms/mortality , White People/psychology
10.
Oncologist ; 14(9): 883-90, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19726455

ABSTRACT

BACKGROUND: Substantial evidence supports the use of adjuvant trastuzumab with chemotherapy for patients with human epidermal growth factor receptor (HER)-2(+) breast cancer; however, a lesser amount of data is available to guide use of this therapy in older patients and in those with significant medical comorbidities. The goal of the current study was to understand how patient age and health status impact oncologists' decisions to recommend adjuvant therapy in older women with HER-2(+) breast cancer. METHODS: Medical oncologists (n = 151) participated in an online survey regarding treatment recommendations for a hypothetical patient of varying age and health status with tumor stage 2, nodal stage 2, estrogen receptor-negative, HER-2(+) breast cancer. Survey respondents recommended either chemotherapy plus trastuzumab, chemotherapy alone, trastuzumab alone, or no therapy. The effect of age and health status on therapeutic recommendations was assessed. FINDINGS: As the hypothetical patient's age increased or health status deteriorated, oncologists were less likely to recommend a combination of chemotherapy plus trastuzumab. In contrast, oncologists were more likely to recommend either trastuzumab alone or no therapy for patients with advanced age and deteriorating health status. Chemotherapy alone was recommended by only 7.5% of respondents, on average. INTERPRETATION: With limited evidence-based data for the treatment of older women with early-stage HER-2(+) breast cancer, medical oncologists recommend a diverse array of therapeutic approaches. With increasing age and declining health status they were less likely to recommend chemotherapy plus trastuzumab and more likely to recommend single-agent trastuzumab or no therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Health Status , Medical Oncology/statistics & numerical data , Practice Patterns, Physicians'/statistics & numerical data , Adult , Age Factors , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Chemotherapy, Adjuvant , Decision Making , Female , Health Care Surveys , Humans , Male , Middle Aged , Neoplasm Staging , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Risk Assessment , Trastuzumab , Treatment Outcome
11.
J Clin Oncol ; 26(33): 5386-92, 2008 Nov 20.
Article in English | MEDLINE | ID: mdl-18955446

ABSTRACT

PURPOSE: To determine the impact of age and health status on adjuvant treatment recommendations for older patients with breast cancer from the perspective of medical oncologists and primary care physicians with geriatric expertise. PATIENTS AND METHODS: One hundred fifty-one oncologists and 158 primary care physicians with geriatric expertise participated in an online survey. The survey described hypothetical patients of varying ages (70, 75, 80, and 85 years) and health status (good, average, and poor) who had node-positive, hormone receptor-positive, human epidermal growth factor receptor 2 (HER-2)/neu-negative; and hormone receptor-negative, HER-2/neu-positive breast cancers. The effects of patient age and health status on the survey participants' adjuvant treatment recommendations were examined using generalized estimation equation methods. RESULTS: The majority of both oncologists and primary care physicians recommended some form of adjuvant therapy for patients of all ages (70, 75, 80, and 85 years) and health status. Both oncologists and primary care providers were less likely to recommend adjuvant treatment as a patient's age increased or health status declined (P < .0001). There were no significant differences in treatment recommendations among primary care physicians and oncologists for patients with hormone receptor-negative, HER-2/neu-positive tumors (P = .54). However, primary care providers were more likely than oncologists to recommend no adjuvant treatment for patients age 75 years or older with hormone receptor-positive, HER-2/neu-negative tumors (P < .01). CONCLUSION: Age and health status influence oncologists' and primary care providers' adjuvant treatment recommendations. Evidence-based guidelines for breast cancer treatment in older adults taking into account age and health status are needed.


Subject(s)
Breast Neoplasms/therapy , Age Factors , Aged , Aged, 80 and over , Breast Neoplasms/chemistry , Chemotherapy, Adjuvant , Data Collection , Female , Geriatrics , Health Status , Humans , Medical Oncology , Physicians, Family , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis
12.
Am J Surg ; 192(4): 416-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16978940

ABSTRACT

BACKGROUND: Previous studies on the efficacy of primary treatments for ductal carcinoma-in-situ (DCIS) have focused on local recurrence rates. Our objective was to detail the outcomes of local invasive recurrence, distant recurrence, and breast cancer mortality in patients previously treated for DCIS. METHODS: Clinical, pathologic, and outcome data were collected prospectively for 1236 patients with pure DCIS accrued from 1972 through 2005. RESULTS: There were 150 recurrences (87 DCIS and 63 invasive). Invasive local recurrence after mastectomy was rare (0.5% of patients) and after breast preservation was more frequent (12.0% of patients). The 12-year probabilities of breast cancer-specific mortality after mastectomy and after breast preservation were 0.8% and 1.0%, respectively. The 12-year breast cancer-specific mortality and distant disease probability for the 63 patients with invasive recurrences were 12% and 15%, respectively. CONCLUSIONS: Regardless of initial treatment, most patients with invasive local recurrence after treatment for DCIS can be treated and cured.


Subject(s)
Breast Neoplasms/mortality , Carcinoma, Intraductal, Noninfiltrating/mortality , Neoplasm Recurrence, Local/mortality , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Carcinoma, Intraductal, Noninfiltrating/secondary , Carcinoma, Intraductal, Noninfiltrating/surgery , Disease-Free Survival , Female , Follow-Up Studies , Humans , Mastectomy , Neoplasm Invasiveness , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Prospective Studies , Survival Rate , Treatment Outcome
13.
Clin Breast Cancer ; 5 Suppl 1: S18-23, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15347435

ABSTRACT

Adjuvant hormonal therapy in the treatment of women with early-stage, hormone receptor (HR)-positive breast cancer is now considered the standard of care. Adjuvant tamoxifen decreases the risk of breast cancer recurrence and death in women with early-stage breast cancer when taken for 5 years. The benefits of tamoxifen are counterbalanced by toxicities including an increased risk of endometrial cancer and thromboembolic events. The selective aromatase inhibitors (AIs)--including anastrozole, letrozole, and exemestane--are challenging the role of tamoxifen as the adjuvant hormonal therapy of choice in postmenopausal women. Results of the Arimidex and Tamoxifen Alone or in Combination trial favor the use of anastrozole over tamoxifen as initial adjuvant hormonal therapy, with improvement in disease-free survival (DFS) and a favorable toxicity profile. The results of 2 large adjuvant trials using AIs sequentially with tamoxifen in postmenopausal women with early-stage, HR-positive breast cancer have been reported. The MA-17 study randomized women to placebo or letrozole for 5 years after completion of 4.5-6 years of initial tamoxifen. The Intergroup Exemestane Study (IES) randomized women following 2-3 years of adjuvant tamoxifen to continue to receive tamoxifen or switch to exemestane for a total of 5 years of adjuvant hormonal therapy. The MA-17 and IES trials demonstrated superior DFS with the AI and corroborated the smaller GROCTA-4B and Italian Tamoxifen Arimidex trials, which studied sequential therapy with aminoglutethamide or anastrozole. There is now substantial medical evidence supporting the use of AIs in postmenopausal women with early-stage, HR-positive breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Aromatase Inhibitors/administration & dosage , Breast Neoplasms/drug therapy , Tamoxifen/administration & dosage , Aged , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Female , Humans , Middle Aged , Neoplasm Staging , Postmenopause , Randomized Controlled Trials as Topic
14.
Oncologist ; 8(6): 514-20, 2003.
Article in English | MEDLINE | ID: mdl-14657529

ABSTRACT

Patients with metastatic breast cancer consist of a heterogeneous group of patients whose prognoses and clinical courses can vary depending on host factors, such as comorbidity and age, and on tumor factors, such as hormone-receptor status, grade, and anatomical site of disease. Although the median survival time for patients with metastatic breast cancer is 2-4 years, subsets of patients with either indolent or limited metastatic disease may have prolonged survival times. Further, expectations of treatment, both in terms of efficacy and of toxicity, vary greatly based upon the specific treatment, patient characteristics, and tumor characteristics. Thus, the goals of treatment for patients with metastatic breast cancer are influenced by estimates of prognoses as well as a balance between physician and patient preferences regarding efficacy and toxicity considerations. Traditionally, objective measures of response and survival have been the targeted end points in clinical trial design and in physician selection of therapy for metastatic breast cancer. More recently, issues of quality of life have surfaced as important end points, especially from the perspective of the patient. The decision-making process in selecting the optimal treatment for patients with metastatic breast cancer is, therefore, a multidimensional process involving subjective as well as objective goals of treatment. Ultimately, the benefits of treatment must justify the risks and toxicities of the treatment, and the impact of treatment should be measured in relation to specified goals. Both physician and patient perspectives are important in establishing the objectives of treatment, and this process is optimally an interactive and ongoing process throughout the course of disease.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Metastasis , Patient Care Planning , Breast Neoplasms/therapy , Clinical Trials as Topic , Decision Making , Female , Humans , Physician-Patient Relations , Prognosis
15.
Mol Cancer Ther ; 2(9): 855-62, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14555704

ABSTRACT

Treatment of solid tumors with combinations of chemotherapeutic agents has not led to significant increases in long-term survival. Recent studies support a role for inhibitors of checkpoint arrest as a means to enhance the cytotoxicity of chemotherapy. We have shown previously that triptolide (PG490), an oxygenated diterpene derived from a Chinese medicinal plant, induces apoptosis in cultured tumor cells and sensitizes tumor cells to topoisomerase inhibitors by blocking p53-mediated induction of p21. Here we extend our studies to a tumor xenograft model and evaluate the efficacy and safety of PG490-88 (14-succinyl triptolide sodium salt), a water-soluble prodrug of PG490. We also look at the combination of PG490 or PG490-88 with CPT-11, a topoisomerase I inhibitor, in cultured cells and in the tumor xenograft model. We show that PG490-88 is a safe and potent antitumor agent when used alone, causing tumor regression of lung and colon tumor xenografts. We also show that PG490-88 acts in synergy with CPT-11 to cause tumor regression. A phase I trial of PG490-88 for solid tumors began recently and safety and optimal dosing data should accrue within the next 12 months. Our findings that PG490-88 causes tumor regression and that it acts in synergy with DNA-damaging chemotherapeutic agents suggest a role as an antineoplastic agent and chemosensitizer for the treatment of patients with solid tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Colonic Neoplasms/drug therapy , Diterpenes/therapeutic use , Lung Neoplasms/drug therapy , Animals , Camptothecin/therapeutic use , Cell Cycle/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Inhibitors/pharmacology , Epoxy Compounds , Humans , Irinotecan , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Phenanthrenes/therapeutic use , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
16.
Curr Treat Options Oncol ; 4(2): 133-40, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12594939

ABSTRACT

The role of hormonal therapy for the treatment of patients with early stage breast cancer has been evaluated in many studies. The results of these studies establish tamoxifen as the gold standard of hormonal therapy for the adjuvant treatment of hormone receptor-positive invasive breast cancer in pre- and postmenopausal women. Studies show tamoxifen reduces the risk of invasive breast cancer in women at increased risk for the disease, including women with ductal carcinoma in situ. Tamoxifen has adverse effects such as hot flashes, increased risk of uterine cancer in postmenopausal women, and rare occurrence of thromboembolic disease. Despite the multiple therapeutic roles of tamoxifen, alternatives are needed. Aromatase inhibitors (AI) are drugs with antiestrogenic activity. AIs function by inhibiting the peripheral conversion of adrenally synthesized androstenedione to estradiol through inhibition of the aromatase enzyme. AIs do not suppress estradiol synthesis by the ovary adequately. Therefore, AIs are effective in reducing circulating estradiol levels in postmenopausal women, but not premenopausal women. Selective nonsteroidal AIs, including anastrozole (Arimidex; AstraZeneca, Wilmington, DE) and letrozole (Femara; Novartis, East Hanover, NJ), and the steroidal AI exemestane (Aromasin; Pharmacia, Peapack, NJ) have been associated with increased specificity and improved therapeutic index compared to nonselective AIs such as aminoglutethamide. Nonsteroidal and steroidal AIs have demonstrated to be superior to megestrol acetate in second-line therapy of postmenopausal women with metastatic breast cancer, and selective nonsteroidal AIs have shown to be superior to tamoxifen in first-line therapy of postmenopausal women with metastatic breast cancer. The ATAC (Arimidex, tamoxifen, alone, or in combination) trial is the only published randomized trial comparing the efficacy of an AI to tamoxifen for the adjuvant treatment of women with early breast cancer. This large study showed that at a median follow-up time of 33 months, anastrozole alone results in significant improvement in disease-free survival rates, reduction in contralateral breast cancers, and increased tolerability, compared to tamoxifen in postmenopausal women. Although the long-term effects of AIs are not known, the early positive results of the ATAC trial led to the approval of anastrozole by the US Food and Drug Administration for use as adjuvant hormonal therapy for postmenopausal women with hormone receptor-positive invasive breast cancer. Thus, there is an alternative to tamoxifen for postmenopausal women with relative/absolute contraindications to tamoxifen use or patients who choose not to take tamoxifen because of its side-effect profile. New AIs may challenge the position of tamoxifen as the gold standard for the treatment of early stage breast cancer in postmenopausal women.


Subject(s)
Aromatase Inhibitors , Breast Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Adult , Aged , Breast Neoplasms/pathology , Female , Humans , Middle Aged , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Tamoxifen/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...