Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
2.
Nat Med ; 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824244

ABSTRACT

Inhibition of histone lysine acetyltransferases (KATs) KAT6A and KAT6B has shown antitumor activity in estrogen receptor-positive (ER+) breast cancer preclinical models. PF-07248144 is a selective catalytic inhibitor of KAT6A and KAT6B. In the present study, we report the safety, pharmacokinetics (PK), pharmacodynamics, efficacy and biomarker results from the first-in-human, phase 1 dose escalation and dose expansion study (n = 107) of PF-07248144 monotherapy and fulvestrant combination in heavily pretreated ER+ human epidermal growth factor receptor-negative (HER2-) metastatic breast cancer (mBC). The primary objectives of assessing the safety and tolerability and determining the recommended dose for expansion of PF-07248144, as monotherapy and in combination with fulvestrant, were met. Secondary endpoints included characterization of PK and evaluation of antitumor activity, including objective response rate (ORR) and progression-free survival (PFS). Common treatment-related adverse events (any grade; grades 3-4) included dysgeusia (83.2%, 0%), neutropenia (59.8%, 35.5%) and anemia (48.6%, 13.1%). Exposure was approximately dose proportional. Antitumor activity was observed as monotherapy. For the PF-07248144-fulvestrant combination (n = 43), the ORR (95% confidence interval (CI)) was 30.2% (95% CI = 17.2-46.1%) and the median PFS was 10.7 (5.3-not evaluable) months. PF-07248144 demonstrated a tolerable safety profile and durable antitumor activity in heavily pretreated ER+HER2- mBC. These findings establish KAT6A and KAT6B as druggable cancer targets, provide clinical proof of concept and reveal a potential avenue to treat mBC. clinicaltrial.gov registration: NCT04606446 .

3.
Future Oncol ; 18(15): 1805-1816, 2022 May.
Article in English | MEDLINE | ID: mdl-35187951

ABSTRACT

The addition of a CDK4/6 inhibitor to endocrine therapy improves progression-free and overall survival in women with metastatic estrogen receptor-positive breast cancer. In that setting, CDK4/6 inhibitors induce a potent cell-cycle arrest (which may be accompanied by tumor senescence) but fail to induce apoptotic cell death. Venetoclax is a potent inhibitor of BCL2, a pro-survival protein overexpressed in the majority of estrogen receptor-positive cancers. Pre-clinical findings indicate that venetoclax augments tumor response to the CDK4/6 inhibitor palbociclib by triggering apoptosis, including in senescent cells. The PALVEN phase Ib trial will further examine this finding. The primary objective is to identify the maximum tolerated dose and determine the recommended phase II dose for palbociclib, letrozole and venetoclax combination therapy. Clinical Trial Registration: NCT03900884 (ClinicalTrials.gov).


The current 'gold standard' treatment for estrogen receptor-positive, HER2-negative metastatic breast cancer is endocrine therapy with a CDK4/6 inhibitor. This combination improves tumor response and patient outcomes, primarily by reducing tumor cell growth. Paradoxically, less killing of tumor cells is observed in the presence of a CDK4/6 inhibitor. The authors hypothesize that co-treatment with venetoclax, an inhibitor of the BCL2 survival protein, will help trigger tumor death, thereby further improving tumor responses and patient outcomes. As a first step, combination therapy comprising letrozole, palbociclib and venetoclax will be tested in a phase I trial to identify the recommended doses for subsequent studies.


Subject(s)
Breast Neoplasms , Receptors, Estrogen , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic , Clinical Trials, Phase I as Topic , Female , Humans , Letrozole/therapeutic use , Piperazines , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-bcl-2/genetics , Pyridines , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Sulfonamides
4.
Asia Pac J Clin Oncol ; 18(6): 560-569, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34908235

ABSTRACT

AIM: Palbociclib was approved in the United States in 2015 to treat estrogen receptor-positive/human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer (ABC). This study evaluated outcomes and safety in patients treated with palbociclib in Australia and India with hormone receptor-positive (HR+)/HER2- ABC before palbociclib became commercially available. METHODS: Postmenopausal women (≥18 years) with HR+/HER2- ABC who were appropriate candidates for letrozole therapy received palbociclib 125 mg once daily for 21 days followed by 7 days off, and letrozole 2.5 mg once daily (continuous). Safety, tumor response, and patient-reported outcomes (Australian cohort) were evaluated. RESULTS: In total, 252 patients received palbociclib plus letrozole (Australia, n = 152; India, n = 100). More patients in the Australian versus Indian cohort had received prior chemotherapy (advanced/metastatic setting: 45.9% vs. 32.0%), endocrine therapy (advanced/metastatic setting: 63.2% vs. 54.3%), and advanced/metastatic therapies (61.8% vs. 31.0%). The most frequently reported all-grade palbociclib-related treatment-emergent adverse events were neutropenia (66.7%), fatigue (35.3%), and stomatitis (26.6%); grade 3/4 neutropenia was reported as palbociclib-related in 62.7% of patients. Febrile neutropenia was reported in six patients (2.4%). Eight patients (3.2%) discontinued because of an adverse event. The objective response rate was 19.4% (95% CI, 14.7%-24.9%) overall and 2.3% in Australian patients with ≥2 lines of prior therapy for metastatic disease. Patient-reported quality of life scores were maintained throughout the study. CONCLUSIONS: In an expanded access setting in Australia and India, palbociclib plus letrozole was well tolerated in patients with HR+/HER2- ABC, with a safety profile consistent with previous reports.


Subject(s)
Breast Neoplasms , Neutropenia , Humans , Female , Letrozole/therapeutic use , Breast Neoplasms/pathology , Receptors, Estrogen/metabolism , Postmenopause , Quality of Life , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Australia , Receptor, ErbB-2/metabolism , Neutropenia/etiology
5.
Article in English | MEDLINE | ID: mdl-28603643

ABSTRACT

BACKGROUND: Young women diagnosed with breast cancer may be confronted by many difficult decisions, especially around fertility preservation prior to commencing cancer treatment. The information to be conveyed is complex, and it may be difficult to weigh up the risks and benefits of the different fertility preservation options available. This complexity is compounded by the widespread low levels of literacy and health literacy in Australia, which may result in greater difficulties in understanding available health information and in decision-making. METHODS/DESIGN: A working group of experts have developed a fertility-related online decision aid for a low health literacy population, guided by health literacy principles. The decision aid will be pilot tested with 30 women diagnosed with early breast cancer between 5 years and 6 months previously. To be eligible, at the time of diagnosis, women must be between 18 and 40 years (inclusive), pre-menopausal, have no history of metastatic disease, have not completed their families, be able to give informed consent and have low health literacy. Participants will be asked to reflect back to the time in which they were diagnosed. Participants will complete a questionnaire before and after reviewing the decision aid to determine the feasibility, use and acceptability of the decision aid. The decision aid will be modified accordingly. Participants may also choose to review a previously developed (high literacy) decision aid and provide feedback in comparison to the low health literacy decision aid. DISCUSSION: This project represents the first study to develop an online fertility decision aid developed from low health literacy models in the context of breast cancer. It is anticipated that the low health literacy decision aid will be useful and acceptable to young women with low health literacy who have been diagnosed with breast cancer and that it will be preferred over the high literacy decision aid. TRIAL REGISTRATION: ACTRN12615001364561p.

6.
Mol Oncol ; 9(1): 128-39, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25151299

ABSTRACT

PURPOSE: Metabolomics is a global study of metabolites in biological samples. In this study we explored whether serum metabolomic spectra could distinguish between early and metastatic breast cancer patients and predict disease relapse. METHODS: Serum samples were analysed from women with metastatic (n = 95) and predominantly oestrogen receptor (ER) negative early stage (n = 80) breast cancer using high resolution nuclear magnetic resonance spectroscopy. Multivariate statistics and a Random Forest classifier were used to create a prognostic model for disease relapse in early patients. RESULTS: In the early breast cancer training set (n = 40), metabolomics correctly distinguished between early and metastatic disease in 83.7% of cases. A prognostic risk model predicted relapse with 90% sensitivity (95% CI 74.9-94.8%), 67% specificity (95% CI 63.0-73.4%) and 73% predictive accuracy (95% CI 70.6-74.8%). These results were reproduced in an independent early breast cancer set (n = 40), with 82% sensitivity, 72% specificity and 75% predictive accuracy. Disease relapse was associated with significantly lower levels of histidine (p = 0.0003) and higher levels of glucose (p = 0.01), and lipids (p = 0.0003), compared with patients with no relapse. CONCLUSIONS: The performance of a serum metabolomic prognostic model for disease relapse in individuals with ER-negative early stage breast cancer is promising. A confirmation study is ongoing to better define the potential of metabolomics as a host and tumour-derived prognostic tool.


Subject(s)
Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Breast Neoplasms/surgery , Metabolome , Metabolomics , Models, Biological , Receptors, Estrogen , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Neoplasm Recurrence, Local , Predictive Value of Tests , Prognosis , Retrospective Studies
7.
Future Oncol ; 9(10): 1477-87, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24106899

ABSTRACT

AIM: Anthracyclines are commonly used in breast cancer, although they lack validated predictive biomarkers. We explored the interaction between TOP2A protein by quantitative immunofluorescence (QIF) and anthracycline sensitivity. PATIENTS & METHODS: Patients with estrogen receptor-negative breast cancer received neoadjuvant epirubicin. Pretreatment biopsies were analyzed using AQUA(®). Total, cytoplasmic (C) and nuclear (N) TOP2A protein concentrations were expressed as QIF scores and compared with pathologic complete response (pCR), TOP2A by immunohistochemistry, TOP2A mRNA, TOP2A and HER2 gene status, and Ki-67 level. RESULTS: In total, 76 cases were assessable. C, N, and total scores did not correlate with pCR, or other markers. The N:C ratio differed significantly by HER2 status. No pCRs occurred in patients in the lowest N:C quartile. CONCLUSION: Although no relevant correlation between TOP2A QIF scores and pCR was found, N:C ratio may have a negative predictive role, and may merit further exploration in a multifactorial predictive model that includes tumor and host factors.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Epirubicin/therapeutic use , Topoisomerase II Inhibitors/therapeutic use , Antigens, Neoplasm/genetics , Breast Neoplasms/mortality , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Female , Humans , Immunohistochemistry , Neoadjuvant Therapy , Poly-ADP-Ribose Binding Proteins , Prognosis , Protein Transport
8.
Mol Oncol ; 6(4): 437-44, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22687601

ABSTRACT

BACKGROUND: Metabolomics, a global study of metabolites and small molecules, is a novel expanding field. In this pilot study, metabolomics has been applied to serum samples from women with metastatic breast cancer to explore outcomes and response to treatment. PATIENTS AND METHODS: Pre-treatment and serial on-treatment serum samples were available from an international clinical trial in which 579 women with metastatic breast cancer were randomized to paclitaxel plus either a targeted anti-HER2 treatment (lapatinib) or placebo. Serum metabolomic profiles were obtained using 600 MHz nuclear magnetic resonance spectroscopy. Profiles were compared with time to progression, overall survival and treatment toxicity. RESULTS: Pre- and on-treatment serum samples were assessed for over 500 patients. Unbiased metabolomic profiles in the biologically unselected overall trial population did not correlate with outcome or toxicity. In a subgroup of patients with HER2-positive disease treated with paclitaxel plus lapatinib, metabolomic profiles from patients in the upper and lower thirds of the dataset showed significant differences for time to progression (N = 22, predictive accuracy = 89.6%) and overall survival (N = 16, predictive accuracy = 78.0%). CONCLUSIONS: In metastatic breast cancer, metabolomics may play a role in sub selecting patients with HER2 positive disease with greater sensitivity to paclitaxel plus lapatinib.


Subject(s)
Breast Neoplasms/blood , Breast Neoplasms/metabolism , Metabolome , Metabolomics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Clinical Trials, Phase III as Topic , Disease Progression , Female , Follow-Up Studies , Humans , Lapatinib , Neoplasm Metastasis , Paclitaxel/therapeutic use , Pilot Projects , Quinazolines/therapeutic use , Randomized Controlled Trials as Topic , Receptor, ErbB-2/metabolism , Survival Analysis , Time Factors , Treatment Outcome
9.
Breast Cancer Res ; 14(3): R70, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22551440

ABSTRACT

INTRODUCTION: Pre-clinical data suggest p53-dependent anthracycline-induced apoptosis and p53-independent taxane activity. However, dedicated clinical research has not defined a predictive role for TP53 gene mutations. The aim of the current study was to retrospectively explore the prognosis and predictive values of TP53 somatic mutations in the BIG 02-98 randomized phase III trial in which women with node-positive breast cancer were treated with adjuvant doxorubicin-based chemotherapy with or without docetaxel. METHODS: The prognostic and predictive values of TP53 were analyzed in tumor samples by gene sequencing within exons 5 to 8. Patients were classified according to p53 protein status predicted from TP53 gene sequence, as wild-type (no TP53 variation or TP53 variations which are predicted not to modify p53 protein sequence) or mutant (p53 nonsynonymous mutations). Mutations were subcategorized according to missense or truncating mutations. Survival analyses were performed using the Kaplan-Meier method and log-rank test. Cox-regression analysis was used to identify independent predictors of outcome. RESULTS: TP53 gene status was determined for 18% (520 of 2887) of the women enrolled in BIG 02-98. TP53 gene variations were found in 17% (90 of 520). Nonsynonymous p53 mutations, found in 16.3% (85 of 520), were associated with older age, ductal morphology, higher grade and hormone-receptor negativity. Of the nonsynonymous mutations, 12.3% (64 of 520) were missense and 3.6% were truncating (19 of 520). Only truncating mutations showed significant independent prognostic value, with an increased recurrence risk compared to patients with non-modified p53 protein (hazard ratio = 3.21, 95% confidence interval = 1.740 to 5.935, P = 0.0002). p53 status had no significant predictive value for response to docetaxel. CONCLUSIONS: p53 truncating mutations were uncommon but associated with poor prognosis. No significant predictive role for p53 status was detected. TRIAL REGISTRATION: ClinicalTrials.gov NCT00174655.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Doxorubicin/therapeutic use , Genes, p53 , Taxoids/therapeutic use , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adolescent , Adult , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Base Sequence , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Disease-Free Survival , Docetaxel , Female , Humans , Ki-67 Antigen/metabolism , Lymphatic Metastasis/genetics , Middle Aged , Mutation, Missense , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , Sequence Analysis, DNA , Sequence Deletion , Survival Analysis , Young Adult
10.
Lancet Oncol ; 13(4): e148-60, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22469125

ABSTRACT

As the mean age of the global population increases, breast cancer in older individuals will be increasingly encountered in clinical practice. Management decisions should not be based on age alone. Establishing recommendations for management of older individuals with breast cancer is challenging because of very limited level 1 evidence in this heterogeneous population. In 2007, the International Society of Geriatric Oncology (SIOG) created a task force to provide evidence-based recommendations for the management of breast cancer in elderly individuals. In 2010, a multidisciplinary SIOG and European Society of Breast Cancer Specialists (EUSOMA) task force gathered to expand and update the 2007 recommendations. The recommendations were expanded to include geriatric assessment, competing causes of mortality, ductal carcinoma in situ, drug safety and compliance, patient preferences, barriers to treatment, and male breast cancer. Recommendations were updated for screening, primary endocrine therapy, surgery, radiotherapy, neoadjuvant and adjuvant systemic therapy, and metastatic breast cancer.


Subject(s)
Aged , Breast Neoplasms/epidemiology , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/epidemiology , Carcinoma, Intraductal, Noninfiltrating/therapy , Breast Neoplasms/pathology , Breast Neoplasms, Male/epidemiology , Breast Neoplasms, Male/pathology , Breast Neoplasms, Male/therapy , Carcinoma, Intraductal, Noninfiltrating/pathology , Combined Modality Therapy , Decision Making , Europe/epidemiology , Female , Geriatric Assessment , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Practice Guidelines as Topic , Societies, Medical
11.
Breast ; 21(3): 336-42, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22406214

ABSTRACT

There are no clinical tools to functionally assess degree of DNA damage in breast cancer. The comet assay is an accepted research tool for assessing DNA damage, however, most cancer studies have assessed lymphocytes as surrogate cells. The aim of this pilot study was to use the comet assay in early breast cancer directly in tumor tissue to compare DNA damage between and within traditionally defined subgroups, and to explore intra-tumoral heterogeneity. Scrapings of tumor and healthy breast tissue were obtained at primary surgery from 104 women. Comet assay was applied to quantitatively assess DNA damage, revealing substantial inter- and intra-subgroup variation. Marked intra-tumoral heterogeneity was evident across all subgroups. The degree of DNA damage for an individual could not be predicted by breast cancer subgroup. Comet assay warrants further study as a potential clinical tool for identification of tumoral DNA damage and ultimately, individualised use of DNA damaging therapy.


Subject(s)
Breast Neoplasms/genetics , Comet Assay/methods , Cytogenetic Analysis/methods , DNA Damage , DNA, Neoplasm/genetics , Adult , Aged , Breast Neoplasms/pathology , Female , Humans , Italy , Micronucleus Tests , Middle Aged , Neoplasm Staging , Pilot Projects
12.
Breast ; 20 Suppl 3: S135-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22015281

ABSTRACT

Chemotherapy is the only evidence based adjuvant systemic treatment option in triple negative breast cancer (TNBC). Despite emerging results for targeted biological therapies for this subpopulation, lack of robust results does not currently support their use beyond the confines of a clinical trial. Conventional systemic chemotherapy remains the standard of care and is curative in a minority of patients. There is no defined standard chemotherapy and there is currently no robust, prospective, randomized data to advise different use of specific chemotherapy agents in TNBC as compared to non-TNBC. Data suggest high sensitivity to chemotherapy, however it is yet to be determined whether this increased sensitivity is agent/regimen specific or whether it reflects general chemosensitivity. This review will focus on systemic chemotherapy in early TNBC, particularly anthracyclines and platinums, and potential predictive tools to guide chemotherapy use.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Neoadjuvant Therapy/methods , Adult , Aged , Biopsy, Needle , Breast Neoplasms/mortality , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Follow-Up Studies , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Randomized Controlled Trials as Topic , Retrospective Studies , Risk Assessment , Survival Analysis , Treatment Outcome
14.
Future Oncol ; 7(2): 173-86, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21345137

ABSTRACT

Fulvestrant is a pure estrogen antagonist that binds, blocks and downgrades the estrogen receptor (ER). Its unique mechanism of action is its antitumor activity after progression on prior endocrine therapy. Fulvestrant has shown activity in ER-dependent cells that are ligand independent. Fulvestrant has been approved at 250 mg/month for postmenopausal women with hormone-sensitive advanced breast cancer after progression or recurrence on antiestrogen therapy. The fulvestrant 500 mg regimen has just received approval by the EMA and the US FDA, supported by dose-dependent ER downregulation and the recent results of the clinical trial CONFIRM. Fulvestrant in combination with systemic lowering of estrogen has shown no improvement over fulvestrant alone. Combination therapy with inhibitors of growth factor signaling may have greater efficacy and is under exploration. To enhance the benefit of fulvestrant and improve outcomes for individuals with ER-positive breast cancer, greater understanding of resistance mechanisms is required. A key issue is identification of patients with ER-positive disease who retain sensitivity to antiestrogen therapy after progression on tamoxifen and/or aromatase inhibitors.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Estradiol/analogs & derivatives , Estrogen Antagonists/therapeutic use , Postmenopause , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/chemistry , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms/pathology , Clinical Trials as Topic , Estradiol/adverse effects , Estradiol/chemistry , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogen Antagonists/adverse effects , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Humans , Neoplasm Staging , Premenopause , Receptors, Estrogen/antagonists & inhibitors , Treatment Outcome
15.
Int J Biochem Cell Biol ; 43(7): 1010-20, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20460168

ABSTRACT

Metabolomics, the study of metabolites and small intermediate molecules, may play a key role in further elucidation of breast cancer. This dynamic, simultaneous assessment of thousands of metabolites allows identification of the presence, concentration and fluxes of specific metabolites, and recognition of the critical metabolic pathways recruited in carcinogenesis. Studies of tumour cell and tissue allow focused analysis on the tumour, whilst studies of biofluids have the appeal of concurrent assessment of tumour and host. Elucidation of these metabolites and pathways may provide essential insights into both the intercellular environment and host/tumour interaction, allowing recognition of new biomarkers for diagnosis and prediction of outcome, new therapy targets and novel approaches for monitoring response and toxicity. Certainly, the field of metabolomics may evolve as a valuable, complementary clinical tool. In this review, current metabolomic data in breast cancer will be presented. The dominant metabolic pathways and metabolite disturbances associated with malignant transformation of breast cells will be outlined, leading to an overview of potential clinical implications for individuals with breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms , Glycolysis/physiology , Metabolic Networks and Pathways/physiology , Metabolomics/methods , Biomarkers, Tumor/analysis , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Choline/analysis , Choline/metabolism , Female , Humans , Prognosis
16.
Cancer Manag Res ; 2: 13-25, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-21188093

ABSTRACT

Lapatinib is a dual tyrosine kinase inhibitor of epidermal growth factor receptor (EGFR/ErbB1) and human epidermal growth factor receptor 2 (HER2/ErbB2). EGFR and HER2 overexpression is associated with aggressive breast cancer with a high risk of disease relapse and death. Although lapatinib targets both EGFR and HER2, its effects on HER2 appear to be more critical. The role of lapatinib in the first-line setting remains unclear. A phase II first-line monotherapy lapatinib trial in HER2-therapy-naïve metastatic breast cancer (MBC) patients confirms efficacy in HER2-positive tumors. Retrospective analysis of a phase III, first-line MBC study confirmed incremental benefit from lapatinib and paclitaxel over paclitaxel alone in HER2-positive disease. A prospective phase III study confirms superiority of letrozole and lapatinib over letrozole alone in HER2-positive MBC. Further investigation is required to define the potential first-line role for lapatinib. Particular strengths appear to be its manageable toxicity profile, lack of cross resistance with trastuzumab, activity in central nervous system disease, and synergy in combination with other anticancer therapy. Current limitations are lack of dosing recommendations from early trials, lack of predictive biomarkers beyond HER2 status, and lack of large prospective phase III trials for HER2-positive disease in the first-line setting. The role of lapatinib in HER2-negative disease is unclear.

17.
Nat Rev Clin Oncol ; 7(12): 725-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20975745

ABSTRACT

Recommendation of systemic adjuvant therapy and choice of optimal agents for early-stage breast cancer remains a challenge. Adjuvant therapy is indicated on the assumption of residual micrometastatic disease. Adjuvant assessment tools for prognosis and prediction of treatment benefit, including Adjuvant! Online, the St Gallen Consensus, Oncotype DX(®) and MammaPrint(®), aid clinical decision making. However, all of these tools have limitations that must be considered in their judicious application. Clinicopathological based tools are critically dependent on accurate, standardized measurement of parameters. Multigene tools are appealing for their objectivity and reproducibility, particularly regarding analysis of proliferation, but these approaches still overlook the biological heterogeneity within tumors evidenced by distinct cell subpopulations with different genomic patterns and function. The greatest treatment challenge remains for patients assessed as intermediate risk of relapse, a problem not overcome by multigene tools. Remarkable diversity in breast cancer dictates that adjuvant management must be biologically driven. Future identification of predictive biomarkers for specific chemotherapy sensitivity may allow targeted use of available agents, including anthracyclines, taxanes and DNA damaging agents. The presence of drug targets and targetable signaling pathways, rather than molecularly defined subgroups, may ultimately drive treatment decisions.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Chemotherapy, Adjuvant , Drug Therapy, Computer-Assisted , Gene Expression Profiling , Algorithms , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Cell Division , Combined Modality Therapy , Female , Humans , Ki-67 Antigen/analysis , Neoplasm Proteins/analysis , Neoplasm Staging , Neoplasm, Residual , Practice Guidelines as Topic , Predictive Value of Tests , Prognosis , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Reproducibility of Results , Risk Assessment
19.
20.
Breast ; 19(5): 312-21, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20382530

ABSTRACT

Triple negative breast cancer (TNBC) accounts for approximately 15% of breast cancer cases. TNBC is an immunohistochemically defined subtype, with significant diversity within the subtype. Generally TNBC occurs in younger women and is marked by high rates of relapse, visceral and CNS metastases, and early death. Current therapy fails to curtail the innate aggressive behaviour of TNBC in the majority of patients. The poor prognosis coupled with a lack of targeted use of therapies is reflected in the high mortality. In a minority of patients with highly chemosensitive disease, no robust clinical evidence exists to guide use of current cytotoxics. Critical to optimal future management are accurate identification of truly triple negative disease and adequately powered prospective TNBC trials to establish treatment efficacy and define predictive biomarkers.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Angiogenesis Inhibitors/therapeutic use , Anthracyclines/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epothilones/therapeutic use , ErbB Receptors/antagonists & inhibitors , Female , Gene Expression Profiling , Genes, BRCA1 , Humans , Immunohistochemistry , Platinum/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Taxoids/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...