Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Breast Cancer Res Treat ; 205(2): 267-279, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38453781

ABSTRACT

PURPOSE: Previous studies have reported the benefit of dual HER2-targeting combined to neoadjuvant chemotherapy in HER2-amplified breast cancer (HER2 + BC). Moreover, besides the cardiac toxicity following their association to Trastuzumab, anthracyclines chemotherapy may not profit all patients. The NeoTOP study was designed to evaluate the complementary action of Trastuzumab and Pertuzumab, and the relevance of an anthracycline-based regimen according to TOP2A amplification status. METHODS: Open-label, multicentre, phase II study. Eligible patients were aged ≥ 18 with untreated, operable, histologically confirmed HER2 + BC. After centralized review of TOP2A status, TOP2A-amplified (TOP2A+) patients received FEC100 for 3 cycles then 3 cycles of Trastuzumab (8 mg/kg then 6 mg/kg), Pertuzumab (840 mg/kg then 420 mg/kg), and Docetaxel (75mg/m2 then 100mg/m2). TOP2A-not amplified (TOP2A-) patients received 6 cycles of Docetaxel (75mg/m2) and Carboplatin (target AUC 6 mg/ml/min) plus Trastuzumab and Pertuzumab. Primary endpoint was pathological Complete Response (pCR) using Chevallier's classification. Secondary endpoints included pCR (Sataloff), Progression-Free Survival (PFS), Overall Survival (OS), and toxicity. RESULTS: Out of 74 patients, 41 and 33 were allocated to the TOP2A + and TOP2A- groups respectively. pCR rates (Chevallier) were 74.4% (95%CI: 58.9-85.4) vs. 71.9% (95%CI: 54.6-84.4) in the TOP2A + vs. TOP2A- groups. pCR rates (Sataloff), 5-year PFS and OS were 70.6% (95%CI: 53.8-83.2) vs. 61.5% (95%CI: 42.5-77.6), 82.4% (95%CI: 62.2-93.6) vs. 100% (95%CI: 74.1-100), and 90% (95%CI: 69.8-98.3) vs. 100% (95%CI: 74.1-100). Toxicity profile was consistent with previous reports. CONCLUSION: Our results showed high pCR rates with Trastuzumab and Pertuzumab associated to chemotherapy. They were similar in TOP2A + and TOP2A- groups and the current role of neoadjuvant anthracycline-based chemotherapy remains questioned. TRIAL REGISTRATION NUMBER: NCT02339532 (registered on 14/12/14).


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms , Carboplatin , DNA Topoisomerases, Type II , Docetaxel , Neoadjuvant Therapy , Receptor, ErbB-2 , Trastuzumab , Humans , Female , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Middle Aged , Trastuzumab/administration & dosage , Trastuzumab/adverse effects , Trastuzumab/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/mortality , Breast Neoplasms/genetics , Receptor, ErbB-2/metabolism , Adult , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Docetaxel/administration & dosage , Docetaxel/adverse effects , Carboplatin/administration & dosage , Carboplatin/adverse effects , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Cyclophosphamide/administration & dosage , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Poly-ADP-Ribose Binding Proteins/genetics , Anthracyclines/administration & dosage , Anthracyclines/therapeutic use , Epirubicin/administration & dosage
2.
BMC Cancer ; 21(1): 1287, 2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34856945

ABSTRACT

BACKGROUND: Breast cancer screening is currently predominantly based on mammography, tainted with the occurrence of both false positivity and false negativity, urging for innovative strategies, as effective detection of early-stage breast cancer bears the potential to reduce mortality. Here we report the results of a prospective pilot study on breast cancer detection using blood plasma analyzed by Fourier-transform infrared (FTIR) spectroscopy - a rapid, cost-effective technique with minimal sample volume requirements and potential to aid biomedical diagnostics. FTIR has the capacity to probe health phenotypes via the investigation of the full repertoire of molecular species within a sample at once, within a single measurement in a high-throughput manner. In this study, we take advantage of cross-molecular fingerprinting to probe for breast cancer detection. METHODS: We compare two groups: 26 patients diagnosed with breast cancer to a same-sized group of age-matched healthy, asymptomatic female participants. Training with support-vector machines (SVM), we derive classification models that we test in a repeated 10-fold cross-validation over 10 times. In addition, we investigate spectral information responsible for BC identification using statistical significance testing. RESULTS: Our models to detect breast cancer achieve an average overall performance of 0.79 in terms of area under the curve (AUC) of the receiver operating characteristic (ROC). In addition, we uncover a relationship between the effect size of the measured infrared fingerprints and the tumor progression. CONCLUSION: This pilot study provides the foundation for further extending and evaluating blood-based infrared probing approach as a possible cross-molecular fingerprinting modality to tackle breast cancer detection and thus possibly contribute to the future of cancer screening.


Subject(s)
Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Spectroscopy, Fourier Transform Infrared/methods , Adult , Area Under Curve , Breast Neoplasms/pathology , Case-Control Studies , DNA Fingerprinting , Disease Progression , Early Detection of Cancer/methods , Feasibility Studies , Female , Humans , Liquid Biopsy/methods , Machine Learning , Middle Aged , Pilot Projects , Prospective Studies , ROC Curve , Support Vector Machine
3.
NPJ Breast Cancer ; 7(1): 124, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34535679

ABSTRACT

To find metastatic recurrence biomarkers of triple-negative breast cancer (TNBC) treated by neoadjuvant chemotherapy and anti-EGFR antibodies (NAT), we evaluated tumor genomic, transcriptomic, and immune features, using MSK-IMPACT assay, gene arrays, Nanostring technology, and TIL assessment on H&E. Six patients experienced a rapid fatal recurrence (RR) and other 6 had later non-fatal recurrences (LR). Before NAT, RR had low expression of 6 MHC class I and 13 MHC class II genes but were enriched in upregulated genes involved in the cell cycle-related pathways. Their TIL number before NAT in RR was very low (<5%) and did not increase after treatment. In post-NAT residual tumors, RR cases showed high expression of SOX2 and CXCR4. Our results indicate that high expression of cell cycle genes, combined with cold immunological phenotype, may predict strong TNBC resistance to NAT and rapid progression after it. This biomarker combination is worth validation in larger studies.

4.
JCO Glob Oncol ; 7: 811-819, 2021 06.
Article in English | MEDLINE | ID: mdl-34086481

ABSTRACT

PURPOSE: Luminal, human epidermal growth factor receptor 2-negative breast cancer represents the most common subtype of breast malignancies. Neoadjuvant strategies of operable breast cancer are mostly based on chemotherapy, whereas it is not completely understood which patients might benefit from neoadjuvant hormone therapy (NAHT). MATERIALS AND METHODS: The SAFIA trial is a prospective multicenter, international, double-blind, neoadjuvant phase III trial, using upfront 21-gene Oncotype DX Breast Recurrence Score assay (recurrence score [RS] < 31) to select operable luminal human epidermal growth factor receptor 2-negative patients, for induction hormonal therapy HT (fulvestrant 500 mg with or without goserelin) before randomly assigning responding patients to fulvestrant 500 mg (with or without goserelin) plus either palbociclib (cyclin-dependent kinase 4/6 inhibitor) or placebo. The objectives of this interim analysis were to assess the feasibility of upfront RS determination on core biopsies in the Middle-East and North Africa region and evaluate the efficacy of induction NAHT in patients with an RS < 31. RESULTS: At the time of this interim analysis, 258 patients with relative risk were accrued, including 202 patients (RS < 31% to 78.3%) treated with induction NAHT and 182 patients evaluable so far for response. The feasibility of performing the Oncotype DX assays on core biopsy specimens was optimal in 96.4% of cases. Overall, 93.4% of patients showed hormone sensitivity and no difference in NAHT efficacy was noticed between RS 0-10, 11-25, and 26-30. Interestingly, patients with high RS (26-30) showed a trend toward a higher major response rate (P = .05). CONCLUSION: The upfront 21-gene assay performed on biopsies is feasible in our population and has allowed us to select patients with high hormone sensitivity (RS < 31). This approach could be an alternative to upfront surgery without significant risk of progression, particularly during pandemic times.


Subject(s)
Breast Neoplasms , Neoadjuvant Therapy , Africa, Northern , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Female , Humans , Middle East , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Prospective Studies , Receptor, ErbB-2 , Receptors, Estrogen
5.
Breast ; 41: 1-7, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29913374

ABSTRACT

BACKGROUND: Cardiac toxicity with a decrease of the left ventricular ejection fraction (LVEF) is the main side effect induced by trastuzumab. This study reports the fluctuation of LVEF over the 12 months of adjuvant trastuzumab in PHARE trial (NCT00381901). METHODS: LVEF assessment was performed every 3 months while patients received trastuzumab and after completion of treatment over the first 2 years and then every 6 months afterwards. The fluctuations of LVEF over time were described and a logistic regression model was performed investigating associated factors to LVEF perfect recovery at baseline value. RESULTS: A total of 1631 patients who received 12 months of trastuzumab from PHARE trial, were considered in the analysis. A total of 13 881 LVEF measurements were assessed. Baseline mean LVEF was 66.08% (standard error (SE): 0.15) and the mean relative LVEF decrease observed at 12-month was 3.61% (SE: 0.31). No clinical characteristic was significantly associated to LVEF fluctuation. After completion of trastuzumab, the relative difference progressively disappeared with beyond 30 months a relative difference value of 0.08% (SE: 0.42). Nevertheless, at 30 months, 48.53% of patients with available measures (379/781) did not fully recover their baseline LVEF value. CONCLUSION: The LVEF decreased during treatment with trastuzumab and rose up after the completion of treatment without coming back to the initial values for a substantial subset. These results would suggest investigating some strategies aimed to improve the ability to achieve a full recovery.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Breast Neoplasms/drug therapy , Trastuzumab/administration & dosage , Ventricular Function, Left/drug effects , Adult , Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/genetics , Cardiotoxicity/etiology , Chemotherapy, Adjuvant , Echocardiography , Female , Follow-Up Studies , Humans , Middle Aged , Receptor, ErbB-2/genetics , Trastuzumab/therapeutic use
6.
Oncotarget ; 9(28): 19463-19468, 2018 Apr 13.
Article in English | MEDLINE | ID: mdl-29731958

ABSTRACT

BACKGROUND: Since the approval of the first poly (adenosine diphosphate [ADP]) ribose polymerase inhibitor (PARPi; olaparib [Lynparza™]) for platinum-sensitive relapsed high grade ovarian cancer, with either germline or somatic BRCA1/2 deleterious variants, the strategies for BRCA1/2 are dynamically changing. Along with germline testing within the context of familial or sporadic ovarian cancer, patients are now being referred for BRCA1/2 genetic assay above all for treatment decisions: in this setting tumour BRCA assay can allow to identify an estimated 3-9% of patients with peculiar somatic BRCA1/2 mutations. These women could also benefit from PARPi therapy. This new type of approach is really challenging, in particular due to the technical and analytical difficulties regarding low quality DNA deriving from formalin-fixed, paraffin-embedded (FFPE) specimens. AIM: in this manuscript, we try to a) underline many issues related to BRCA1/2 analysis by next generation sequencing technologies (NGS), b) provide some responses to many questions regarding this new paradigm related to OvCa patients' management. Some considerations for incorporating genetic analysis of ovarian tumour samples into the patient pathway and ethical requirements are also provided. METHODS: we used our retrospective data based on thousands of ovarian cancer women sequenced for BRCA1/2 genes. DISCUSSION: tumor BRCA1/2 assay should be rapidly introduced in routine laboratory practice as first line testing by using harmonized pipelines based on consensus guidelines.

7.
Oncol Lett ; 14(1): 337-344, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28693173

ABSTRACT

The immunobiology of breast cancer (BC) subtypes, including luminal cancer, remains unclear. Cluster of differentiation (CD)8+ tumor-infiltrating lymphocytes (TIL) are essential components of tumor-specific cellular adaptive immunity. However, only few studies have addressed the significance of cluster of differentiation 8+(CD8+) TIL in patients with luminal BC. The present study aimed to evaluate the predictive and prognostic significance of CD8+ TIL in patients with luminal B/human epidermal growth factor receptor 2 (HER 2)-negative BC treated with anthracycline-based neoadjuvant chemotherapy (NC). A total of 31 patients who underwent breast-conserving surgery or mastectomy post-NC were enrolled. Immunostaining for CD8+ TIL was performed using rabbit monoclonal antibodies against human CD8+. Intra- and peritumoral CD8+ TIL expression levels were classified into high and low, based on the median value of each. CD8+ TIL expression data were demonstrated to be correlated with disease-free survival (DFS) and overall survival (OS), using Kaplan-Meier and Cox's proportional hazards regression tests. The results revealed that, among all clinicopathological characteristics, only pathological complete response (pCR) was significantly correlated with intratumoral CD8+ TIL expression (P=0.016). A total of 9/16 patients (56%) with high intratumoral CD8+ TIL expression achieved pCR, in contrast with 2 out of 15 patients (13.3%) with low expression (P=0.016). High expression of intratumoral CD8+ TIL was significantly associated with OS (log-rank test, P=0.023). Multivariate Cox regression analysis revealed that intratumoral expression of CD8+ TIL was an independent prognostic factor for OS [hazard ratio (HR)=2.82; 95% confidence interval (CI)=0.911-4.833, P=0.007], but not for DFS (HR=1.11; 95% CI=0.282-2.078; P=0.508). In conclusion, the results of the present study suggested that high intratumoral CD8+ TIL expression was significantly predictive of pCR post-NC, and represented an independent prognostic factor for improved OS. In contrast, low intratumoral CD8+ TIL expression was a strong predictor of lack of pCR to NC, as well as an independent prognostic factor for poor OS. Assessment of the immune response in conjunction with the usual parameters may aid in the further stratification of patients with luminal B/HER 2-negative BC regarding the prediction of pCR post-NC and overall prognosis.

8.
Anticancer Agents Med Chem ; 17(4): 491-499, 2017.
Article in English | MEDLINE | ID: mdl-27456662

ABSTRACT

Triple negative breast cancer (TNBC) is a heterogeneous disease entity constituting about 15% of breast cancer cases worldwide. TNBC is associated with poor prognosis and lack of sustained response to conventional chemotherapeutic agents. Tumoral heterogeneity and the presence of several subtypes of TNBC such as Basal like (BL)-1, BL-2, immune-modulatory, luminal androgen receptor, mesenchymal, and mesenchymal/stem like subtype and claudin low subtype, may explain some of the difficulties faced in managing this challenging disease subgroups. Although no approved targeted therapy is available for TNBCs, molecular-profiling efforts have revealed promising molecular targets such as the vascular endothelial growth factor (VEGF), epidermal growth factor receptor (EGFR), polyadenosine ribose polymerase inhibitors (PARPi) and DNA repair pathway, androgen pathway, and NOTCH pathway. TNBC is subject to intense research activities aiming at dissecting potential pathways, identifying potential molecular signatures and biomarkers in order to properly develop new targeted biologic modifiers. Despite this, there is a lack of approved predictive and prognostic biomarkers, and keeping in view the complexity of TNBC biology, research should be targeted towards identifying multi-factorial signatures rather than single markers. This review aims to summarize the current evidence, ongoing research and discuss future strategies for the treatment of patients with TNBC. In addition we have reviewed the recent advances in detecting predictive and prognostic biomarkers and identifying surrogate markers for early identification of potential responders to the new therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Targeted Therapy , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Antineoplastic Agents/chemistry , DNA Repair/drug effects , ErbB Receptors/metabolism , Hedgehog Proteins/metabolism , Humans , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Receptors, Androgen/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/pathology
9.
Lancet Oncol ; 16(16): 1700-10, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26596672

ABSTRACT

BACKGROUND: Patients with advanced HER2-positive breast cancer frequently develop CNS metastases. The metastases that progress after brain radiotherapy and HER2-targeted systemic therapy are a difficult therapeutic challenge. We aimed to assess the efficacy and safety of afatinib, an irreversible blocker of the ErbB protein family, alone or combined with vinorelbine, compared with treatment of the investigator's choice in women with HER2-positive breast cancer with progressive brain metastases during or after treatment with trastuzumab, lapatinib, or both. METHODS: We did this randomised, open-label, multicentre, phase 2 trial in 40 hospitals in Canada, Finland, France, Germany, Italy, Spain, South Korea, and the USA. Women older than 18 years with histologically confirmed HER2-overexpressing breast cancer and CNS recurrence or progression as determined by Response Evaluation Criteria in Solid Tumors (RECIST version 1.1) during or after treatment with trastuzumab, lapatinib, or both, were eligible. We randomly assigned patients (1:1:1) centrally to afatinib 40 mg orally once per day, afatinib 40 mg per day plus intravenous vinorelbine 25 mg/m(2) once per week, or investigator's choice of treatment in cycles of 3 weeks until disease progression, patient withdrawal, or unacceptable toxicity. Treatment assignment was not masked for clinicians or patients, but the trial team was masked until database lock to reduce bias. The primary endpoint, assessed in the intention-to-treat population, was patient benefit at 12 weeks, defined by an absence of CNS or extra-CNS disease progression, no tumour-related worsening of neurological signs or symptoms, and no increase in corticosteroid dose. Safety was assessed in all patients who received at least one dose of a study drug. This completed trial is registered with ClinicalTrials.gov, number NCT01441596. FINDINGS: Between Dec 22, 2011, and Feb 12, 2013, we screened 132 patients, of whom 121 were eligible and randomly assigned to treatment: 40 to afatinib alone, 38 to afatinib plus vinorelbine, and 43 to investigator's choice. All patients discontinued study treatment before the data collection cutoff on Oct 16, 2014. Patient benefit was achieved in 12 (30·0%; 95% CI 16·6-46·5) patients given afatinib alone (difference vs investigator's choice: -11·9% [95% CI -32·9 to 9·7], p=0·37), 13 (34·2%; 19·6-51·4) given afatinib plus vinorelbine (difference vs investigator's choice: -7·6% [-28·9 to 14·2], p=0·63), and 18 (41·9%; 27·0-57·9) given investigator's choice. The most common treatment-related grade 3 or 4 adverse events were diarrhoea (seven [18%] of 40 patients in the afatinib only group vs nine [24%] of 37 patients in the afatinib plus vinorelbine group vs two [5%] of 42 patients in the investigator's choice group) and neutropenia (none vs 14 [38%] vs four [10%]). INTERPRETATION: Patient benefit with afatinib-containing treatments was not different from that in patients given investigator's choice of treatments; however, adverse events were frequent and afatinib-containing treatments seemed to be less well tolerated. No further development of afatinib for HER2-positive breast cancer is currently planned. FUNDING: Boehringer Ingelheim.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Vinblastine/analogs & derivatives , Adult , Afatinib , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/analysis , Brain Neoplasms/chemistry , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Canada , Disease Progression , Disease-Free Survival , Europe , Female , Humans , Kaplan-Meier Estimate , Lapatinib , Middle Aged , Proportional Hazards Models , Protein Kinase Inhibitors/adverse effects , Quinazolines/adverse effects , Receptor, ErbB-2/analysis , Republic of Korea , Risk Factors , Time Factors , Trastuzumab/therapeutic use , Treatment Outcome , United States , Vinblastine/adverse effects , Vinblastine/therapeutic use , Vinorelbine
10.
Eur J Cancer ; 51(13): 1660-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26163096

ABSTRACT

BACKGROUND: This article reports, the cardiac toxicity according to 6- versus 12-month durations of adjuvant trastuzumab in PHARE randomised trial (NCT00381901). PATIENTS AND METHODS: Cardiac follow-up and Left Ventricular Ejection Fraction (LVEF) assessment by echocardiography or multigated acquisition scan were performed every 3 months while patients received trastuzumab and after completion of treatment over the first 2 years and every 6 months afterwards. The primary cardiac end-point was Cardiac Heart Failure (CHF) defined as New York Heart Association (NYHA) class III or IV. The secondary cardiac end-points were: cardiac events, cardiac dysfunctions defined by NYHA class I and II; LVEF decreases, cardiac recoveries. The cardiac subcommittee reviewed cardiac events and assessed if patients had favourable outcomes or not on the basis of trends from LVEF measurements. RESULTS: Among 3380 patients the cardiac dysfunction assessment included 14,055 and 13,218 LVEF measurements in the 12- and 6-month arms. The overall incidences of CHF were 0.65% (11/1690) and 0.53% (9/1690) in the 12 and 6 month arms, respectively (p>0.05). Cardiac dysfunction occurred in 5.9% (100/1690) and 3.4% (58/1690) of patients in the 12 and 6 month arms, respectively (p=0.001). Recoveries were observed for the majority patients and 0.79% (27/3380) of patients experienced an unfavourable cardiac outcome. CONCLUSION: PHARE confirm that the incidence of cardiac end-points remains low and mostly reversible after trastuzumab. Identification at baseline of cardiac risk categories of patients should be of interest to provide an optimal adaptation of adjuvant modalities and a shorter duration might be an option.


Subject(s)
Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Heart Failure/chemically induced , Protein Kinase Inhibitors/adverse effects , Trastuzumab/adverse effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Female , France/epidemiology , Heart Failure/diagnosis , Heart Failure/epidemiology , Heart Failure/physiopathology , Humans , Incidence , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Risk Assessment , Risk Factors , Stroke Volume/drug effects , Time Factors , Treatment Outcome , Ventricular Function, Left/drug effects
11.
Crit Rev Oncol Hematol ; 95(1): 88-104, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25900915

ABSTRACT

Breast cancer is heterogeneous in clinical, morphological, immunohistochemical and biological features, as reflected by several different prognostic subgroups. Neoadjuvant approaches are currently used for the "in vivo" efficacy assessment of treatments. Pathological complete response (pCR) has been reported as a reliable predictive factor of survival in that setting. However, pCR remains a subject of controversy in terms of definition and its evaluation methods. In addition, its predictive value for patient outcome in various breast cancer biological subtypes has been under debate. In this review, we will present the existing definitions of pCR, the impact of its evaluation methods on its rate and the assessment of its predictive value for patient outcome in the molecular subtypes of breast cancer (luminal A and B, Triple Negative and HER2-positive).


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Breast/pathology , Neoadjuvant Therapy , Biomarkers, Tumor/analysis , Breast Neoplasms/diagnosis , Female , Humans , Neoadjuvant Therapy/methods , Prognosis , Receptor, ErbB-2/analysis , Treatment Outcome
12.
Bull Cancer ; 98(7): 807-25, 2011 Jul.
Article in French | MEDLINE | ID: mdl-21727059

ABSTRACT

Apart from therapeutic advances related to new treatments, our practices in the management of early breast cancer have been modified by to key organizational settings (1) mass screening, substantially altering the presentation and epidemiology of breast cancer and (2) the development of guidelines to ensure that any patient management is in agreement with the demonstrated impact in the adjuvant treatment. In daily practice, the impact of screening and guidelines recommendations has put us now in a paradoxical situation: while the majority of non-metastatic breast cancers treated in the hexagon are node negative, most of the results of clinical studies on chemotherapy and targeted therapies today arise from populations predominantly node positive. Therefore, it seemed legitimate to convene a working group around a reflection on the directions of adjuvant chemotherapy in a growing node negative population in order to better respond to the questions of the field oncologists, trying to address the discrepancies between different existing guidelines.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Age Factors , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chemotherapy, Adjuvant/standards , Female , France , Genes, erbB-2/genetics , Humans , Lymph Nodes/pathology , Neoplasm Staging , Prognosis , Receptors, Estrogen/metabolism , Trastuzumab , Tumor Burden
13.
Oncologist ; 15(5): 476-83, 2010.
Article in English | MEDLINE | ID: mdl-20421265

ABSTRACT

BACKGROUND: Differences in the efficacy of various chemotherapies in patients with estrogen receptor (ER)(+) metastatic breast cancer are not well understood. In the present study, we assessed the efficacy of docetaxel in patients with metastatic breast cancer according to ER expression. METHODS: The efficacy of docetaxel in terms of the response rate and progression-free survival (PFS) time was analyzed according to ER expression in four randomized trials comparing a docetaxel-based regimen with a nontaxane regimen that included a total of 1,631 patients. The odds ratio for tumor response was estimated with logistic regression and a hazard ratio (HR) for PFS was estimated with Cox proportional hazards models. FINDINGS: ER expression was assessable in 1,037 patients included in these trials (64%). ER was expressed in 601 tumors (58%). Docetaxel was associated with a similarly higher response rate in both patients with ER(+) (odds ratio, 2.90; 95% confidence interval [CI], 1.72-4.87) and patients with ER(-) (odds ratio, 2.55; 95% CI, 1.44-4.51) disease. The lower hazard for disease progression with docetaxel was also similar in ER(+) (HR, 0.82; 95% CI, 0.67-1.00) and ER(-) (HR, 0.86; 95% CI, 0.70-1.07) cancers. The effect of docetaxel was not different in ER(+) and ER(-) disease, in terms of both the response rate and PFS time (interaction test, p = .77 and p = .93). INTERPRETATION: Docetaxel produces a higher response rate and lower risk for disease progression to a statistically similar extent in both patients with ER(+) and patients with ER(-) metastatic breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estrogen Receptor alpha/biosynthesis , Taxoids/therapeutic use , Adult , Aged , Breast Neoplasms/pathology , Disease-Free Survival , Docetaxel , Female , Humans , Middle Aged , Neoplasm Metastasis , Randomized Controlled Trials as Topic
14.
Cancer Invest ; 28(2): 195-200, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19968496

ABSTRACT

This study compares the outcome of 76 patients with N0 breast carcinoma, node-negative at axillary lymph node dissection (pN0) after neoadjuvant chemotherapy (NeoCT), treated with (RLNI+, 39 patients) or without (RLNI-, 37 patients) elective regional lymph node areas irradiation. For RLNI- and RLNI+ groups respectively at 10 years, survival without local-regional recurrence was 95% and 91% (p = .59), survival without distant metastasis was 97% and 78% (p = .018) and overall survival was 96% and 75% (p = .013). Clinical size < 4 cm was a strong pronostic factor.


Subject(s)
Breast Neoplasms/radiotherapy , Lymphatic Metastasis/radiotherapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Combined Modality Therapy , Female , Humans , Lymph Nodes/pathology , Middle Aged , Neoadjuvant Therapy , Radiotherapy, Adjuvant , Survival Analysis
15.
Indian J Surg Oncol ; 1(1): 19-26, 2010 Jan.
Article in English | MEDLINE | ID: mdl-22930614

ABSTRACT

Tamoxifen has been considered for several decades as the standard upfront hormonal therapy for patients with endocrine-sensitive early breast cancer. The efficacy and favorable toxicity profiles of third-generation aromatase inhibitors (AIs), anastrozole, letrozole and exemestane, in advanced disease led to their development in early breast cancer. Recent trial results consistently showed the superiority of AIs over tamoxifen in using the two following therapeutic approaches: either the upfront strategy (randomization of newly diagnosed patients: tamoxifen for 5 years vu AI for 5 years) or the sequencial strategy (randomization of newly diagnosed patients: tamoxifen (2-3 years) followed by AI or the inverse for a total of 5 years vs upfront AI for 5 years).Despite some common characteristics, a body of evidence on AIs suggests some specific differences between the three agents in terms of efficacy as well as toxicity profiles. Thus, these hormonal agents may not be considered interchangeable in clinical practice. This review will explore available results from AIs trials and will try to define their present role in the upfront adjuvant management of postmenopausal patients with breast cancer.

16.
Expert Opin Pharmacother ; 10(9): 1435-47, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19445563

ABSTRACT

The hormonal therapy of patients with endocrine-sensitive early breast cancer has mainly consisted, for several decades, of the gold standard tamoxifen. The efficacy and favorable toxicity profiles of third-generation aromatase inhibitors (AIs), anastrozole, letrozole and exemestane, in advanced disease led to their development in early breast cancer. Recent results consistently show the superiority of these agents over tamoxifen. Adjuvant trials evaluated AIs using four different therapeutic approaches: (1) Upfront strategy: randomization of newly diagnosed patients: tamoxifen for 5 years versus AI for 5 years. (2) Sequencial strategy: randomization of newly diagnosed patients: tamoxifen (2 - 3 years) followed by AI or the inverse for a total of 5 years versus upfront AI for 5 years. (3) Switch strategy: delayed randomization (or analysis) after 2 - 3 years of tamoxifen (patients free of disease): 2 - 3 years of tamoxifen versus 2 - 3 years of AI (total treatment 5 years). (4) Extended strategy: delayed randomization after 5 years of tamoxifen (patients free of disease): 2 - 5 years of AI versus placebo. Overall, AIs show evidence of superiority over tamoxifen in the adjuvant setting with proven improved efficacy and better toxicity profile. Despite some common characteristics, a body of evidence on AIs indicates some specific differences between the three agents in mechanism of action, pharmacokinetics, efficacy as well as toxicity profiles. Consequently, these hormonal agents may not be considered interchangeable in clinical practice. This review explores available results from AI trials and tries to define their present role in the adjuvant management of postmenopausal patients with breast cancer.


Subject(s)
Androstadienes/therapeutic use , Breast Neoplasms/drug therapy , Nitriles/therapeutic use , Triazoles/therapeutic use , Anastrozole , Animals , Breast Neoplasms/diagnosis , Breast Neoplasms/epidemiology , Disease Management , Early Detection of Cancer , Female , Humans , Letrozole , Randomized Controlled Trials as Topic/methods , Randomized Controlled Trials as Topic/trends
17.
Ther Clin Risk Manag ; 4(1): 189-204, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18728707

ABSTRACT

Following promising data for metastatic breast cancer in terms of efficacy and safety profile, third-generation aromatase inhibitors (AI), anastrozole, letrozole, and exemestane, underwent a full development in early setting. If recent results consistently show the superiority of these agents over tamoxifen, the therapeutic strategies of AIs in adjuvant setting are still debated. Beyond the choice of clinical strategy, the long duration of exposure to AI in adjuvant setting required a full determination of the long-term toxicity profile of these agents. While all three AIs have either favorable (decreased incidence of hot flashes, gynecologic and thromboembolic side-effects) or unfavorable (skeletal complications, arthralgia, musculoskeletal pain, sexual dysfunction) class adverse events, some variability between AIs has been reported in side-effects as well as gastrointestinal, urogenital, neurologic, and visual disturbances, confirming the lack of interchangeability between the three AIs. The overall therapeutic index of AIs appears today superior to that of tamoxifen with proven improved efficacy and better toxicity profile. This review will explore the results from the available adjuvant AIs trials with a particular emphasis on safety profiles, quality of life, and therapeutic index, helping to define the present role of AIs in the adjuvant management of postmenopausal patients with breast cancer.

18.
J Clin Oncol ; 26(12): 1980-6, 2008 Apr 20.
Article in English | MEDLINE | ID: mdl-18421049

ABSTRACT

PURPOSE: Taxanes (paclitaxel or docetaxel) have been sequenced or combined with anthracyclines (doxorubicin or epirubicin) for the first-line treatment of advanced breast cancer. This meta-analysis uses data from all relevant trials to detect any advantages of taxanes in terms of tumor response, progression-free survival (PFS), and survival. PATIENTS AND METHODS: Individual patient data were collected on eight randomized combination trials comparing anthracyclines + taxanes (+ cyclophosphamide in one trial) with anthracyclines + cyclophosphamide (+ fluorouracil in four trials), and on three single-agent trials comparing taxanes with anthracyclines. Combination trials included 3,034 patients; single-agent trials included 919 patients. RESULTS: Median follow-up of living patients was 43 months, median survival was 19.3 months, and median PFS was 7.1 months. In single-agent trials, response rates were similar in the taxanes (38%) and in the anthracyclines (33%) arms (P = .08). The hazard ratios for taxanes compared with anthracyclines were 1.19 (95% CI, 1.04 to 1.36; P = .011) for PFS and 1.01 (95% CI, 0.88 to 1.16; P = .90) for survival. In combination trials, response rates were 57% (10% complete) in taxane-based combinations and 46% (6% complete) in control arms (P < .001). The hazard ratios for taxane-based combinations compared with control arms were 0.92 (95% CI, 0.85 to 0.99; P = .031) for PFS and 0.95 (95% CI, 0.88 to 1.03; P = .24) for survival. CONCLUSION: Taxanes were significantly worse than single-agent anthracyclines in terms of PFS, but not in terms of response rates or survival. Taxane-based combinations were significantly better than anthracycline-based combinations in terms of response rates and PFS, but not in terms of survival.


Subject(s)
Anthracyclines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Taxoids/therapeutic use , Anthracyclines/administration & dosage , Breast Neoplasms/pathology , Disease-Free Survival , Female , Humans , Neoplasm Metastasis , Proportional Hazards Models , Randomized Controlled Trials as Topic , Survival Rate , Taxoids/administration & dosage
19.
J Clin Oncol ; 26(12): 1987-92, 2008 Apr 20.
Article in English | MEDLINE | ID: mdl-18421050

ABSTRACT

PURPOSE: Overall survival (OS) can be observed only after prolonged follow-up, and any potential effect of first-line therapies on OS may be confounded by the effects of subsequent therapy. We investigated whether tumor response, disease control, progression-free survival (PFS), or time to progression (TTP) could be considered a valid surrogate for OS to assess the benefits of first-line therapies for patients with metastatic breast cancer. PATIENTS AND METHODS: Individual patient data were collected on 3,953 patients in 11 randomized trials that compared an anthracycline (alone or in combination) with a taxane (alone or in combination with an anthracycline). Surrogacy was assessed through the correlation between the end points as well as through the correlation between the treatment effects on the end points. RESULTS: Tumor response (survival odds ratio [OR], 6.2; 95% CI, 5.3 to 7.0) and disease control (survival OR, 5.5; 95% CI, 4.8 to 6.3) were strongly associated with OS. PFS (rank correlation coefficient, 0.688; 95% CI, 0.686 to 0.690) and TTP (rank correlation coefficient, 0.682; 95% CI, 0.680 to 0.684) were moderately associated with OS. Response log ORs were strongly correlated with PFS log hazard ratios (linear coefficient [rho], 0.96; 95% CI, 0.73 to 1.19). Response and disease control log ORs and PFS and TTP log hazard ratios were poorly correlated with log hazard ratios for OS, but the confidence limits of rho were too wide to be informative. CONCLUSION: No end point could be demonstrated as a good surrogate for OS in these trials. Tumor response may be an acceptable surrogate for PFS.


Subject(s)
Anthracyclines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Taxoids/therapeutic use , Anthracyclines/administration & dosage , Biomarkers , Breast Neoplasms/pathology , Disease-Free Survival , Female , Humans , Neoplasm Metastasis , Proportional Hazards Models , Randomized Controlled Trials as Topic/methods , Reproducibility of Results , Survival Rate , Taxoids/administration & dosage , Treatment Outcome
20.
Drug Saf ; 29(9): 785-801, 2006.
Article in English | MEDLINE | ID: mdl-16944964

ABSTRACT

Third-generation aromatase inhibitors (AIs) are now being used for the adjuvant treatment of postmenopausal women with breast cancer, and are challenging tamoxifen, the previous 'gold standard' of care, in this setting. This review evaluates the potential clinical impact of anastrozole, letrozole and exemestane on the cardiovascular (CV) system of postmenopausal women with breast cancer. Some data for CV safety are available for AIs from the advanced disease setting; however, most derive from patients being treated for early disease. CV data on anastrozole for the treatment of early breast cancer were taken from the ATAC trial, in which anastrozole was compared with tamoxifen in the primary adjuvant setting, and the ABCSG trial 8/ARNO 95 combined analysis, in which switching to 3 years of anastrozole after 2 years of tamoxifen was compared with the standard 5 years of tamoxifen adjuvant therapy. Letrozole has been studied in the primary adjuvant setting and the adjuvant sequencing setting in the BIG 1-98 study, as well as in extended adjuvant endocrine therapy after 5 years of tamoxifen in the MA-17 trial. For exemestane, results were reviewed from the IES trial, in which switching to exemestane following 2-3 years of adjuvant tamoxifen was compared with continued tamoxifen treatment. All these trials clearly confirmed that all three AIs significantly reduce the risk of thromboembolic events compared with tamoxifen. Data on anastrozole versus tamoxifen from the ATAC trial (68 months' follow-up) showed a similar incidence of myocardial infarctions (MIs), CV deaths and overall deaths for both therapies; however, anastrozole appeared to be associated with a lower incidence of cerebrovascular events compared with tamoxifen. In addition, the ABCSG trial 8/ARNO 95 study reported no difference in terms of MIs for patients switching to anastrozole compared with patients continuing tamoxifen treatment. Data from BIG 1-98 (26 months' follow-up) suggested that primary adjuvant treatment with letrozole may be associated with a significantly greater incidence of CV events and a numerical increase of cerebrovascular and cardiac deaths compared with tamoxifen. However, no increase in CV events with letrozole was reported from the MA-17 trial. In the IES, updated data at 55 months' median follow up showed no significant difference in the incidence of MIs and cardiac deaths between patients who switched to exemestane compared with those who continued tamoxifen. In conclusion, a significantly reduced risk of thromboembolic disease was observed for all three AIs compared with tamoxifen. Anastrozole is, at this point, the only AI with a detailed benefit-risk profile from over 5 years' follow-up in the adjuvant setting. Thus far, no apparent CV-safety concerns have emerged. Preliminary data on letrozole and exemestane suggest that longer follow-up is needed for these two AIs before being able to fully assess their respective long-term CV toxicity profile. The present differences in CV-safety profiles suggest that third-generation AIs should not be considered as equivalents in clinical practice.


Subject(s)
Aromatase Inhibitors/adverse effects , Breast Neoplasms/drug therapy , Cardiovascular Diseases/chemically induced , Cardiovascular System/drug effects , Aromatase Inhibitors/pharmacology , Cytochrome P-450 Enzyme System , Drug Monitoring , Female , Humans , Polymorphism, Genetic , Postmenopause/drug effects , Risk Assessment , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...