144 results on '"An JN"'
Search Results
2. Abstract P1-17-03: Statin use, site of recurrence, and survival among post-menopausal women taking bisphosphonates as adjuvant therapy for breast cancer (SWOG S0307)
- Author
-
Alison Stopeck, JN Ingle, Elizabeth Claire Dees, William E. Barlow, Carla I. Falkson, Gabriel N. Hortobagyi, Jieling Miao, Mark Clemons, Julie Gralow, Ahg Paterson, D Kizub, and Patricia A. Thompson
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Statin ,business.industry ,medicine.drug_class ,medicine.medical_treatment ,Hazard ratio ,Cancer ,Bisphosphonate ,medicine.disease ,Zoledronic acid ,Breast cancer ,Internal medicine ,medicine ,Adjuvant therapy ,business ,medicine.drug - Abstract
Purpose: Statins may mediate suppression of molecular pathways conferring benefit in cancer. Statins have shown anti-tumor effects in preclinical studies and have been associated with decreased recurrence and improved disease-specific survival. While designed to target cholesterol biosynthesis, statins can also have liver, bone and brain effects. We collected data on statin use in the S0307 adjuvant bisphosphonate trial to test the hypothesis that statin use may decrease risk of recurrence to liver, bone and brain as well as second primary (contralateral) breast cancers, and may act synergistically with bisphosphonates to decrease the risk of recurrence to bone. Patients and Methods: In S0307, 6097 patients diagnosed with Stage I-III breast cancer who had undergone surgery and were receiving adjuvant systemic therapy were randomized to receive zoledronic acid, clodronate, or ibandronate for 3 years. No significant difference was found in disease-free survival (DFS) among the 3 groups, including a sub-analysis of patients > age 55. Statin use was infrequent in younger women in S0307, consequently we analyzed statin use in those > age 55. Cox proportional hazard models were used to determine which variables were independently associated with DFS and to estimate hazard ratios (HR) and 95% confidence intervals (CI). Results: Among women aged ≥ 55 years, 684 (27%) reported taking a statin at baseline and 1,848 did not. Both groups were similar in terms of hormone receptor and HER2 status (p = 0.82). Median age in the statin group was 64.3 versus 61.0 years in the no statin group, mean BMI 31.2 v. 29.5, mean tumor size 2.1cm v. 2.3cm, negative lymph nodes 60% v. 54%, Stage I disease 47% v. 36%, and receipt of chemotherapy 62% v. 71% (all p < 0.01). In the statin group, 122 (17.8%) experienced a DFS event compared to 313 (16.9%) in the no statin group (HR 1.18, CI 0.95-1.46). No difference was observed by statin use in overall recurrence (p=0.28), distant recurrence (p=0.64), or recurrences to the bone (p=0.64), liver (p=0.38) or brain (p=0.65) at initial recurrence. There was no synergy between statin use and specific bisphosphonates. Recurrence and statin useOutcomeGroup 1: On stan at baseline n=684Group 2: No statin at baseline n=1848DFS events122 (17.8%)313 (16.9%)Died without recurrence51 7.5%)97 (5.2%)Recurrence71 (10.4%)216 (11.7%)Contralateral breast cancer9 (1.3%)17 (0.9%)Distant recurrence48 (7%)157 (8.5%)Bone as 1st site of distant recurrence (% distant recurrence)31 (65%)76 (48%)Liver as 1st site of distant recurrence (% distant recurrence)6 (13%)24 (16%)Brain/CNS as 1st site of distant recurrence (% distant recurrence)5 (10%)17 (11%) Conclusions: We found no evidence that statins reduce risk of second primary breast cancers or distant metastases among post-menopausal women with early-stage breast cancer. Despite promising preclinical data, they did not appear to act in synergy with a specific bisphosphonate. Though women in the statin group had less advanced disease at study entry, statin use was not associated with improved DFS. Results are limited by lack of information about type of statin used, adherence, or initiation of statin in control group. Citation Format: Kizub D, Miao J, Stopeck A, Thompson P, Paterson AH, Clemons M, Dees EC, Ingle JN, Falkson CI, Barlow W, Hortobagyi GN, Gralow JR. Statin use, site of recurrence, and survival among post-menopausal women taking bisphosphonates as adjuvant therapy for breast cancer (SWOG S0307) [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P1-17-03.
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P3-08-01: Characteristics, outcomes and prognostic factors of luminal androgen receptor (LAR) triple-negative breast cancer (TNBC)
- Author
-
Kevin J. Thompson, Matthew P. Goetz, Victoria Cafourek, M-Y Polley, Roberto A. Leon-Ferre, Hongfang Liu, Fergus J. Couch, Krishna R. Kalari, Xiaojia Tang, JN Ingle, MC Liu, Poulami Barman, Erin E. Carlson, JC Carter, Judy C. Boughey, DW Visscher, and Vivian Negron
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Apocrine ,Cancer ,Histology ,medicine.disease ,Lower risk ,Breast cancer ,Internal medicine ,Cohort ,medicine ,Luminal androgen receptor ,business ,Triple-negative breast cancer - Abstract
Background: The LAR subtype is a genomically distinct subset of TNBC. Using a large cohort of non-metastatic TNBC patients (pts) with long term follow-up, we sought to further characterize the clinicopathologic features and outcomes of LAR vs non-LAR TNBC. Methods: From a cohort of 9982 women with surgically-treated non-metastatic breast cancer, 605 met criteria for TNBC (ER/PR Results: 58 (20%) tumors were classified as LAR and 225 (80%) as non-LAR. Compared to non-LAR, LAR pts were older (mean age 65 vs 54) and more often postmenopausal (79%vs53%), both p=0.01. Apocrine histology was more common among LAR tumors (21%vs0%), which were also lower grade (grade3: 69%vs95%) and had lower Ki-67 (Ki-67>15%: 64%vs82%), all p Conclusions: LAR TNBCs occurred in older women, were lower grade, and had lower TIL density than nonLAR tumors. While significant differences in IDFS or OS were not demonstrated, LAR pts exhibited a numerically lower risk of a disease event at 3yrs, but higher risk by 10yrs compared to nonLAR pts. In the entire cohort, higher N stage, absence of AdjCT and lower TILs were independently associated with poorer outcomes. Citation Format: Leon-Ferre RA, Polley M-Y, Liu H, Kalari KR, Boughey JC, Liu MC, Cafourek V, Negron V, Ingle JN, Thompson KJ, Tang X, Barman P, Carlson E, Visscher DW, Carter JC, Couch FJ, Goetz MP. Characteristics, outcomes and prognostic factors of luminal androgen receptor (LAR) triple-negative breast cancer (TNBC) [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P3-08-01.
- Published
- 2019
- Full Text
- View/download PDF
4. Abstract P1-06-07: Mayo clinic TNBC outcome calculator: A clinical calculator to predict disease relapse and survival in women with triple-negative breast cancer
- Author
-
Vivian Negron, M-Yc Polley, MC Liu, David W. Hillman, Victoria Cafourek, JN Ingle, Roberto A. Leon-Ferre, Matthew P. Goetz, Hongfang Liu, Judith A. Gilbert, Fergus J. Couch, Judy C. Boughey, Krishna R. Kalari, and DW Visscher
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Proportional hazards model ,medicine.medical_treatment ,Breast surgery ,Lumpectomy ,medicine.disease ,Breast cancer ,Internal medicine ,Cohort ,Medicine ,Stromal tumor ,business ,Mastectomy ,Triple-negative breast cancer - Abstract
Purpose: Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype with substantial risks of disease recurrence. While cytotoxic chemotherapy is commonly administered and reduces recurrence, disease outcomes vary considerably and few prognostic tools are available for risk stratification for TNBC patients. We constructed and validated clinical calculators for invasive-disease free survival (IDFS) and overall survival (OS) for TNBC and compared their performance against AJCC-based models which include only tumor size and nodal status. Methods: From a surgical cohort of 9,982 patients who underwent breast cancer surgery at Mayo Clinic between January 1985 and December 2012, 605 centrally reviewed TNBC patients were identified and used to construct Cox models for IDFS and OS. Patients treated with neoadjuvant chemotherapy were excluded. Variables considered included age, menopausal status, tumor size, nodal status, Nottingham grade, type of breast surgery (mastectomy vs. lumpectomy), adjuvant radiation therapy, adjuvant chemotherapy, Ki67, stromal tumor infiltrating lymphocytes (sTILs), and neutrophil-to-lymphocyte ratio (NLR). Missing values were imputed using single imputation with all variables (including outcomes) included in the imputation model. Backward step-down procedure was used for model selections. The final models were internally validated for calibration and discrimination using bootstrapping methods and compared with AJCC-based models. Results: For both IDFS and OS, higher sTIL's, less extensive nodal involvement, use of adjuvant chemotherapy, and lower NLR were significant predictors of improved clinical outcomes. Premenopausal status and younger age were additionally predictive of improved IDFS and OS, respectively. Models for IDFS and OS have good calibration and are associated with bias-corrected C-indices of 0.68 and 0.71, respectively, as compared with C-indices of 0.59 and 0.62 for AJCC-based models. Conclusions: Our data indicate that a clinical calculator that includes sTIL's, NLR, menopausal status, age, nodal involvement as well as chemotherapy use can provide significantly greater prediction of clinical risk than tumor size and nodal status alone. These tools may be used to identify TNBC patients at elevated risk of disease relapse and to aid physician's communication with patients regarding their long-term disease outlook and planning treatment strategies. External validation is required to further evaluate broader applicability of this tool, which was developed utilizing a single-institutional experience. Citation Format: Polley M-YC, Leon-Ferre RA, Liu H, Gilbert J, Cafourek V, Hillman DW, Negron V, Boughey JC, Liu MC, Ingle JN, Kalari K, Couch F, Visscher DW, Goetz MP. Mayo clinic TNBC outcome calculator: A clinical calculator to predict disease relapse and survival in women with triple-negative breast cancer [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P1-06-07.
- Published
- 2018
- Full Text
- View/download PDF
5. Abstract P1-03-04: Molecular subtyping of androgen receptor-positive patients using gene expression profiles
- Author
-
T Alaparthi, Judy C. Boughey, Xiaojia Tang, JN Ingle, L Wang, Matthew A. Bockol, Richard M. Weinshilboum, Jason P. Sinnwell, Peter T. Vedell, Krishna R. Kalari, VJ Suman, Kevin J. Thompson, Erin E. Carlson, and Matthew P. Goetz
- Subjects
Cancer Research ,Fuzzy clustering ,Rand index ,Computational biology ,Biology ,medicine.disease ,Subtyping ,Non-negative matrix factorization ,Hierarchical clustering ,Breast cancer ,Oncology ,medicine ,Cluster analysis ,Triple-negative breast cancer - Abstract
Breast cancer is a heterogeneous disease, and unsupervised clustering approaches using gene expression data have identified 3-6 distinct subtypes of triple negative breast cancer (TNBC). A genomically and clinically distinct subtype of TNBC is referred to as LAR (Luminal Androgen Receptor). Tumors with this subtype typically express high levels of the AR and exhibit alterations within genes involved in the PI3K pathway (e.g. PIK3CA mutations). Prospective studies are underway using drugs that target the AR alone or in combination with PI3K and CDK 4/6 inhibitors. Given the importance of accurately identifying this subtype, we sought to develop an online tool that uses submitted gene expression data to confidently characterize LAR samples by corroborating the classification with previously published clustering approaches. Methods: We have investigated TNBC RNA-Seq data from The Cancer Genome Atlas (TCGA) breast cancer study (N=123 samples) by cluster analysis. Analysis of the average silhouette width in both biased and unbiased K-means clustering approaches demonstrated LAR and basal as two distinct and significant clusters. A shrunken centroid model of 426 differentially expressed genes, named as CABAL (Clustering Among BAsal and Luminal androgen receptor), was constructed by comparing LAR and basal subtypes. Results: We applied the CABAL model to classify the four TNBC microarray datasets that were previously used in clustering experiments as well as an independent RNA-Seq data cohort. Non-negative matrix factorization (NMF) and fuzzy clustering were applied to the samples (N=1046). Clustering similarity among the methods was assessed with the adjusted rand index, and CABAL demonstrated significant similarity with both fuzzy and NMF clustering methods. Similarly, hierarchical clustering analysis performed on the pooled cohort of 1046 samples recapitulated the CABAL classification with an area under the receiver operating curve of 0.91. Conclusions: Confident and robust identification of samples with the LAR phenotype is paramount in the assessment of clinical associations and therapeutic efficacy. To facilitate LAR identification, we have provided a web-based prediction tool of the CABAL classification, integrated with the NMF and fuzzy clustering results to identify candidate LAR samples. The end user is provided with the pair-wise adjusted rand indexes, thus reinforcing in the clustering characterizations. Further, our online LAR depiction tool provides a set of graphical and tabular summaries, which will be illustrated, while providing additional molecular characterizations of the PAM50 and Metabric classifications. The availability of this tool could advance the genomic research and treatment of TNBC patients. Citation Format: Thompson KJ, Alaparthi T, Sinnwell JP, Carlson EE, Tang X, Bockol M, Vedell PT, Ingle JN, Suman V, Weinshilboum RM, Wang L, Boughey JC, Kalari KR, Goetz MP. Molecular subtyping of androgen receptor-positive patients using gene expression profiles [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P1-03-04.
- Published
- 2019
- Full Text
- View/download PDF
6. Abstract 5774: Indo-Canadian parternship for oral cancer screening in rural India
- Author
-
Datta, Madhurima, primary, Chaitanya, Nallan CSK, additional, Ndvn, Shyam, additional, Palat, Gayatri, additional, Jacob, Jean, additional, Chandran, Priya, additional, Rapelli, Vineela, additional, Jn, Jagannath, additional, Broughton, Sandra, additional, Sutcliffe, Simon, additional, and Laronde, Denise Marie, additional
- Published
- 2020
- Full Text
- View/download PDF
7. Abstract PD1-04: CSMD1 SNPs selectively affect anastrozole response in postmenopausal breast cancer patients
- Author
-
MP Goetz, Aman U. Buzdar, T Dudenkov, Krishna R. Kalari, J Cairns, Richard M. Weinshilboum, Paul E. Goss, MJ Ellis, L Wang, Mark E. Robson, JN Ingle, Michiaki Kubo, and Lois E. Shepherd
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Anastrozole ,Single-nucleotide polymorphism ,Affect (psychology) ,medicine.disease ,030226 pharmacology & pharmacy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Medicine ,business ,medicine.drug - Abstract
BACKGROUND: Based on prospective clinical trials, there is no evidence for differences in efficacy between the 3 aromatase inhibitors (AIs) anastrozole, exemestane, and letrozole. The purpose of this study was to identify germline genetic variants associated with response to AIs and to help identify novel mechanisms associated with drug disease efficacy. METHODS: A genome-wide association study (GWAS) was performed for 624 patients (Steroids 2015;99:32-38) to identify SNPs associated with estrogen level change in women with estrogen receptor (ER) positive breast cancer treated with anastrozole. Replication of associated SNPs was performed in a GWAS from the MA.27 trial that compared adjuvant anastrozole and exemestane treatment of post-menopausal women with ER+ breast cancer. Functional studies were subsequently performed to determine SNP effects and underlying mechanisms. RESULTS: Our initial GWAS identified SNPs within CSMD1 that were associated with changes in estrogen levels during anastrozole therapy. An additional SNP in CSMD1 was also associated with breast cancer events in CCTG MA.27. Functionally, we showed that CSMD1 regulates CYP19 expression in a SNP-, and in an anastrozole- dependent fashion. These phenomena were not observed for either letrozole or exemestane. In MA.27, an anastrozole- specific effect was also seen with the minor allele having a protective effect on time to distant metastasis (HR=0.49, p=0.00259), but this was not the case for exemestane (HR=0.71, p=0.111). Our in vitro functional studies indicated that overexpression of CSMD1 sensitized anastrozole or letrozole resistant cells to anastrozole but not to the other two AIs. The SNP in CSMD1 that was associated with increased CSMD1 and CYP19 expression levels increased anastrozole sensitivity, but not letrozole or exemestane in lymphoblastoid cell lines (LCLs) homozygous for either WT or variant CSMD1 SNP genotypes. Based on these observations, we explored whether anastrozole has additional mechanisms beyond its function as a CYP19 inhibitor. Utilizing an estrogen response element (ERE) luciferase reporter assay in a CYP19 CRISPR knockout breast cancer T47D cell line and a surface plasmon resonance (SPR) assay, we found that anastrozole can also function as an ERα agonist, and can bind to, and result in, proteasome dependent ERα degradation, especially in the presence of E2. Treatment of these CYP19 CRISPR knockout cells with anastrozole in the presence of increasing concentrations of E2 results in greater sensitivity compared with anastrozole alone, while the addition of E2, as expected, does not improve letrozole or exemestane sensitivity. These same observations were also seen in letrozole and anastrazole resistant cells. CONCLUSIONS: Our findings suggest that anastrozole might be more effective than letrozole or exemestane in patients with the CSMD1 SNP. Furthermore, anastrozole can function as an ERα agonist, binding to ERα and resulting in its degradation, especially in the presence of E2. These findings should help to make it possible to develop precision endocrine therapies for women who are candidates for AIs. Citation Format: Cairns J, Ingle J, Dudenkov T, Kalari K, Buzdar A, Kubo M, Robson M, Ellis M, Goss P, Shepherd L, Goetz M, Weinshilboum R, Wang L. CSMD1 SNPs selectively affect anastrozole response in postmenopausal breast cancer patients [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr PD1-04.
- Published
- 2017
- Full Text
- View/download PDF
8. Abstract P6-11-08: Safety and efficacy results from phase I study of BYL 719 plus nab-paclitaxel in HER 2 negative metastatic breast cancer
- Author
-
P Sharma, J Ward, AR Brown, JN Scott, C Lehn, Anne O'Dea, Andrew K. Godwin, Raymond P. Perez, Stephen K. Williamson, Greg Reed, Qamar J. Khan, S Lewis, Vandana G. Abramson, Takefumi Komiya, and Ja De Jong
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Taxane ,business.industry ,medicine.medical_treatment ,Cancer ,Neutropenia ,medicine.disease ,Metastatic breast cancer ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Breast cancer ,Paclitaxel ,chemistry ,Concomitant ,Internal medicine ,medicine ,business - Abstract
Introduction Mutations/deregulations in the phosphatidylinositol-3-kinase (PI3K) pathway are common in breast cancer, Inhibition of the PI3K pathway is recognized as a promising target for the treatment of breast cancer. Although taxanes are effective early on in advanced stage breast cancer, resistance often develops. It has been demonstrated that activation of the PI3K/AKT pathway confers resistance to paclitaxel, and in preclinical models, concomitant inhibition of the PI3K pathway enhances the efficacy of taxanes. BYL719 is a potent oral, class I PI3K inhibitor which strongly inhibits the PI3K alpha isoforms and is significantly less active against the other class I isoforms. Targeting the alpha isoform of PI3K is expected to improve the therapeutic window over inhibitors with less isoform specificity. Nab-Paclitaxel is a solvent-free, nanoparticle, albumin-based paclitaxel which takes advantage of the antitumor activity of paclitaxel while decreasing the toxicities typically associated with the solvent (Cremophor) used to administer the most common formulation of paclitaxel. Methods A 3+3 dose-escalation design evaluated three dose levels of BYL719 (250mg, 300mg, and 350mg) administered PO once daily (D1-28) with nab-Paclitaxel (100 mg/m2 intravenously D 1, 8, 15) every 28 days in patients with metastatic HER 2 negative breast cancer. The aims of the study were to 1) determine the recommended phase II dose (RPTD) of BYL719 + nab-Paclitaxel, 2) assess pharmacokinetics of BYL and nab-paclitaxel, and 3) assess preliminary efficacy. Results 10 patients were enrolled at 3 dose levels of BYL719 and 3 patients were enrolled in expansion cohort at the RPTD of BYL719 of 350 mg PO daily plus nab-paclitaxel 100mg/m2 (D 1, 8, 15). Median age was 61years; 54% (7/13) of patients were hormone receptor positive and 46% (6/13) triple negative. 85% (11/13) had visceral disease, 69% (9/13) had received prior chemotherapy for metastatic disease and 85% (11/13) had received prior taxane in adjuvant/metastatic setting. There were no DLTs in the three cohorts and the MTD of BYL was not reached. Hyperglycemia (G3:31%, G4:0%) and neutropenia (G3:15%, G4:8%), were the most common grade 3/4 adverse events. There were no Grade 3/4 diarrhea or rash. Best overall response for 12 patients was 58% (7/12) (complete response=1, partial response=6), and an additional 33% (4/12) demonstrated stable disease. Objective responses were noted in both hormone positive and triple negative disease. Median duration of response is 6.5 months (range 2-14 months). No pharmacokinetic interactions were detected when BYL and nab-paclitaxel were co-administered. Discussion: This phase I study demonstrates that combination of BYL719 and nab-paclitaxel was well tolerated and shows encouraging efficacy in metastatic HER2 negative breast cancer. Enrollment in the phase II portion of the trial at the RPTD (BYL719 350mg PO daily plus nab-paclitaxel 100mg/m2 D1,8,15 every 28 days) continues. Ongoing analysis of PI3K pathway alterations in tumor and cfDNA will be correlated with clinical response. Citation Format: Sharma P, Abramson VG, O'Dea A, Lewis S, Scott JN, Ward J, De Jong JA, Lehn C, Brown AR, Williamson SK, Perez RP, Komiya T, Godwin AK, Reed GA, Khan QJ. Safety and efficacy results from phase I study of BYL 719 plus nab-paclitaxel in HER 2 negative metastatic breast cancer [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr P6-11-08.
- Published
- 2017
- Full Text
- View/download PDF
9. Abstract P5-07-01: LncRNA MIR2052HG regulates ERα level and endocrine resistance through LMTK3 by recruiting early growth response protein 1
- Author
-
Krishna R. Kalari, Richard M. Weinshilboum, Michiaki Kubo, J Cairns, L Wang, Lois E. Shepherd, MP Goetz, and JN Ingle
- Subjects
MAPK/ERK pathway ,Cancer Research ,Oncology ,Transcription (biology) ,EGR1 ,Biology ,Estrogen receptor alpha ,Molecular biology ,Protein kinase B ,Chromatin immunoprecipitation ,Transcription factor ,Protein kinase C - Abstract
BACKGROUND: A GWAS for the MA.27 aromatase inhibitors (AIs) adjuvant trial (4,406 controls and 252 cases) identified variant (V) SNPs in a long noncoding (lnc) RNA, MIR2052HG, that were associated with longer breast cancer free interval (HR= 0.37, P= 2.15E-07). V SNPs (MAF= 0.32 to 0.42) were associated with lower MIR2052HG and ERα expression in the presence of AIs. MIR2052HG maintained ERα both by promoting AKT/FOXO3-mediated ESR1 transcription and by limiting ubiquitin-mediated ERα degradation. (Cancer Res 76:7012-23, 2016). Our goal was to further elucidate MIR2052HG's mechanism of action. METHODS: RNA-Binding Protein Immunoprecipitation (RBPI) assays were performed to demonstrate that the transcription factor, early growth response protein 1 (EGR1), worked together with MIR2052HG to regulate lemur tyrosine kinase-3 (LMTK3) transcription in MCF7/AC1 and CAMA-1 cells. The location of EGR1 on the LMTK3 gene locus was mapped using chromatin immunoprecipitation (ChIP) assays. The co-localization of MIR2052HG RNA and the LMTK3 gene locus was determined using RNA-DNA dual fluorescent in situ hybridization (FISH). SNP effects were evaluated using a panel of human lymphoblastoid cell lines. RESULTS: TCGA analysis revealed LMTK3 and MIR2052HG expression were highly correlated in ERα-positive breast cancer patients. We found that the MIR2052HG transcript was located in the LMTK3 gene locus by RNA-DNA FISH. Among all of the 12 potential LMTK3 transcription factors identified in the Encode database that were examined by RBPI, only EGR1 showed an interaction with MIR2052HG. CHIP assays confirmed EGR1 binding to the two putative EGR1 binding sites in LMTK3 gene.Depletion of MIR2052HG reduced the binding of EGR1 to the LMTK3 promoter and decreased LMTK3 expression, suggesting that it might function as a scaffold. Mechanistically, decreased LMTK3 levels further increased protein kinase C (PKC) activity and downstream AKT activity, leading to reduced ESR1 mRNA levels via increased pFOXO3. At the protein level, in MIR2052HG depleted cells, increased PKC activity increased the phosphorylation of MEK, ERK, and RSK1, leading to increased ERα phosphorylation at Ser167 and increased ERα degradation. Conversely, overexpression of LMTK3 in MIR2052HG depleted cells reversed these phenotypes. MIR2052HG regulated LMTK3 and ERα expression in a SNP- dependent fashion: the MIR2052HG V SNP, relative to wild-type (WT) genotype, increased LMTK3/ERα expression in response to androstenedione due to increased binding between EGR1 and the LMTK3 promoter in LCLs. However, AI treatment reduced this binding in MIR2052HG variant cells but increased binding in WT cells, resulting in decreased LMTK3/ERα in V cells and increased expression in WT cells. CONCLUSIONS: Our findings support a model in which the protective MIR2052HG variant genotype regulates LMTK3 via MIR2052HG/EGR1, and LMTK3 regulates ERα stability via the PKC/MEK/ERK/RSK1 axis. This regulation may explain the effect of the MIR2052HG variant genotype on cell proliferation and response to AIs in MA.27. These findings provide new insight into the mechanism of action of MIR2052HG and suggest that LMTK3 may be a new therapeutic target in ERα-positive breast cancer patients treated with AIs. Citation Format: Cairns J, Ingle JN, Shepherd LE, Kubo M, Goetz MP, Weinshilboum RM, Kalari KR, Wang L. LncRNA MIR2052HG regulates ERα level and endocrine resistance through LMTK3 by recruiting early growth response protein 1 [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P5-07-01.
- Published
- 2018
- Full Text
- View/download PDF
10. Abstract P3-05-06: Prognostic value of the neutrophil-to-lymphocyte ratio (NLR) and its relation to stromal tumor infiltrating lymphocytes (sTILs) in triple negative breast cancer (TNBC)
- Author
-
Roberto A. Leon-Ferre, Vivian Negron, M-Y Polley, DW Visscher, MC Liu, Matthew P. Goetz, Hongfang Liu, Judy C. Boughey, JN Ingle, David W. Hillman, Victoria Cafourek, Krishna R. Kalari, Judith A. Gilbert, and Fergus J. Couch
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Univariate analysis ,Proportional hazards model ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,Breast cancer ,Internal medicine ,medicine ,Stromal tumor ,Neutrophil to lymphocyte ratio ,business ,Neoadjuvant therapy ,Triple-negative breast cancer - Abstract
Background: While TNBC remains the most aggressive type of breast cancer (BC), substantial heterogeneity in biology and outcomes exists among TNBC subtypes. Historically, risk stratification of TNBC has been based on anatomic factors such as tumor size, nodal involvement and presence of distant metastases. However, these features alone fail to accurately predict outcomes. Tumor immune infiltration (sTILs) and distribution of immune cell subsets in the perip heral blood (NLR) have emerged as variables reported to be associated with outcomes in TNBC. We sought to evaluate whether NLR and sTILs provided independent prognostic information in TNBC. Methods: From a cohort of 9,982 women who underwent BC surgery at Mayo Clinic, Rochester, MN between Jan 1985 and Dec 2012, we identified 605 centrally-confirmed TNBC tumors. Patients (pts) with prior BC, bilateral BC, non-invasive disease, stage IV, neoadjuvant therapy, endocrine therapy, or adenoid cystic histology were excluded. For eligible tumors, clinical and pathologic variables were evaluated, including peripheral blood NLR and central assessment of sTILs per the 2014 International TILs Working Group recommendations. We calculated the Pearson correlation coefficient (PCC) between NLR and sTILs and constructed Cox Proportional Hazards Models to evaluate their association with invasive-disease free (IDFS) and overall survival (OS). NLR and sTILs were both analyzed as continuous variables. Results: Most pts had T1-2 (95%) and N0-1 disease (86%). Median OS follow-up was 10.6yrs. Median IDFS was 12yrs (95%CI 10.2-16.7) and median OS was 18.8yrs (95%CI 15.6-20.8). NLR and sTILs were available in 408 and 599 pts, respectively. The median NLR and sTIL content were 2.29 (0.14-10.50) and 20% (0-90%), respectively. NLR and sTILs were poorly correlated (PCC 0.0237). On univariate analysis (UVA), a higher NLR was associated with worse IDFS (HR 1.13; 95%CI 1.02-1.26, p=0.02) and OS (HR 1.17; 95%CI 1.04-1.31, p=0.01). Each 1% increment in sTILs was associated with improved IDFS (HR 0.99; 95%CI 0.98-0.99, p Conclusions: A lower NLR and a higher sTIL content were each associated with improved IDFS and OS among pts with nonmetastatic TNBC on UVA. However, when evaluated on a MVA, only sTILs remained independently associated with IDFS and OS. Our data suggest that the effect of sTILs on outcomes may not be modified by the NLR. Citation Format: Leon-Ferre RA, Polley M-Y, Liu H, Gilbert J, Cafourek V, Hillman D, Negron V, Boughey JC, Liu MC, Ingle JN, Kalari K, Couch FJ, Visscher DW, Goetz MP. Prognostic value of the neutrophil-to-lymphocyte ratio (NLR) and its relation to stromal tumor infiltrating lymphocytes (sTILs) in triple negative breast cancer (TNBC) [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P3-05-06.
- Published
- 2018
- Full Text
- View/download PDF
11. Abstract P3-07-29: Role of germline BRCA status and tumor homologous recombination (HR) deficiency in response to neoadjuvant weekly paclitaxel followed by anthracycline-based chemotherapy
- Author
-
Matthew P. Goetz, RW Weinshilboum, Judy C. Boughey, Kirsten Timms, Donald W. Northfelt, Amy Lynn Conners, Eric D. Wieben, Ann M. Moyer, DW Visscher, J Jones, Richard Gray, Travis J. Dockter, Sarah A. McLaughlin, Steven N. Hart, L Wang, EP Elkin, JN Ingle, A-R Hartman, Xiaojia Tang, Sara J. Felten, Peter T. Vedell, A Moreno Aspitia, Krishna R. Kalari, VJ Suman, and Katie N. Jones
- Subjects
Gynecology ,Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Anthracycline ,business.industry ,medicine.medical_treatment ,BRCA mutation ,Cancer ,medicine.disease ,Carboplatin ,chemistry.chemical_compound ,Germline mutation ,Breast cancer ,chemistry ,Trastuzumab ,Internal medicine ,medicine ,skin and connective tissue diseases ,business ,medicine.drug - Abstract
Background: Both HR deficiency and BRCA mutation status predict response to platinum-based therapy and BRCA mutation status predicts docetaxel resistance. However, the association of either biomarker with response to the individual elements of either AC or taxanes (T) is unknown since T is commonly given concomitantly with or after anthracyclines (A). We evaluated the association of HRD and BRCA mutation status with response to neoadjuvant weekly T followed by AC or (F)EC in high-risk breast cancer. Methods: We studied 140 high risk Stage I-III breast cancer patients (pts), enrolled in the breast cancer genome guided therapy study (BEAUTY), obtaining biopsies for DNA/RNA sequencing and MRI imaging to assess response to neoadjuvant weekly T (+trastuzumab+/-pertuzumab for HER2+ disease) followed by AC or (F)EC. Germline BRCA status and HR status of tumor samples (Myriad laboratories) were obtained. HR deficient tumor was defined as HRD score ≥42 or BRCA mutation. MRI response by changes in tumor size after 12 weeks of T was classified by WHO criteria. pCR was defined as ypT0/Tis ypN0. Both MRI response after T and pCR (after T and AC) were examined in terms of germline BRCA mutation (gBRCAmut vs. gBRCAwt) and tumor HR deficiency. Results: Of 140 pts enrolled, 8 withdrew consent and 2 carboplatin treated pts were excluded. Germline data were available for 124/130 pts. 12 patients had BRCA deleterious germline mutations (4 BRCA1, 8 BRCA2). MRI partial (PR)/complete response (CR) rate to T was 47.3% (95% CI: 37.8-57.0%) in the BRCAwt group and 66.7% (95% CI: 34.9-90.1%) in the BRCAmut group. No MRI CR's were observed in BRCA1 mut pts. In contrast, pCR rate was 50% in the 12 gBRCAmut pts (95% CI: 21.1-78.9%) and 31.3% in the 112 gBRCAwt pts (95% CI: 22.8-40.7%). HR deficiency status has thus far been determined for 74 pts: 26 pts have HD deficient tumors: 18 TNBC, 5 Luminal B, 2 ER-/HER2+; and 1 ER+/HER2+. Determination of HR deficiency is ongoing and will be reported for the full cohort in terms of 12 week MRI response to T and pCR to T+AC. HR deficientMolecular Subtypeyes (%)no (%)TBD (%)Luminal A0/112/11 (18.2)9/11 (81.8)Luminal B5/37 (13.5)13/37 (35.1)19/37 (51.3)Luminal NOS0/21/2 (50)1/2 (50)ER+/Her2+1/17 (5.8)14/17 (82.4)2/17 (11.8)ER-/Her2+2/20 (10)11/20 (55)7/20 (35)Triple Negative18/43 (41.9)6/43 (18.6)17/43 (39.5)germline BRCA statusMRI partial response after T (%)MRI complete response after T (%)pCR after T&AC (%)BRCA11/4 (25)0/42/4 (50)BRCA25/8 (62.5)2/8 (25)4/8 (50)BRCAwt35/112 (31.3)18/112 (16.1)35/112 (31.3) Conclusion: In the setting of neoadjuvant weekly T followed by AC, pCR rates were non-significantly higher in pts with BRCA1 mutations. While we observed no overall association between BRCA mutation status and response rates to taxanes; nearly all MRI responses to taxanes (partial and complete) were observed in the BRCA2 group. Prospective studies are needed to validate these findings and to determine whether BRCA status can be used to select therapy. HR deficiency is uncommon in luminal A and HER2+, frequent in TNBC, and the association of HRD with both MRI response to taxanes and pCR will be reported at the meeting. Citation Format: Boughey JC, Kalari KR, Suman VJ, McLaughlin SA, Moreno Aspitia A, Moyer AM, Northfelt DW, Gray RJ, Vedell PT, Tang X, Dockter TJ, Jones KN, Felten SJ, Conners AL, Hart SN, Visscher DW, Wieben ED, Ingle JN, Hartman A-R, Timms K, Elkin E, Jones J, Wang L, Weinshilboum RW, Goetz MP. Role of germline BRCA status and tumor homologous recombination (HR) deficiency in response to neoadjuvant weekly paclitaxel followed by anthracycline-based chemotherapy. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P3-07-29.
- Published
- 2016
- Full Text
- View/download PDF
12. Abstract P6-13-04: A phase I trial to evaluate the safety of the addition of alisertib to fulvestrant in hormone receptor positive (HR+), advanced breast cancer
- Author
-
VJ Suman, JN Ingle, Tufia C. Haddad, A B D'Assoro, MP Goetz, and Mateusz Opyrchal
- Subjects
Oncology ,Gynecology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,Everolimus ,Fulvestrant ,business.industry ,medicine.medical_treatment ,Cancer ,Neutropenia ,medicine.disease ,chemistry.chemical_compound ,Breast cancer ,chemistry ,Internal medicine ,Alisertib ,Mucositis ,Medicine ,business ,medicine.drug - Abstract
Background: During tumor progression, activation of Aurora A kinase (AURKA) is associated with epithelial to mesenchymal transition (EMT) reprogramming and expansion of a subpopulation of tumor initiating cells harboring a CD44+/CD24low/- phenotype [D'Assoro, Oncogene 2014]. These cells are characterized by their capacity to self-renew, resist drug therapies, and promote distant metastases. In ER+ breast cancer (BC) models, activation of AURKA is associated with down-regulation of ERα expression and resistance to endocrine therapy. Alisertib, a selective inhibitor of AURKA, can reverse EMT and restore tumor ERα expression and sensitivity to endocrine therapy [Opyrchal, PLoS One 2014]. As a single agent in HR+ advanced BC, alisertib was associated with a 6-month clinical benefit rate of 54% and median PFS of 7.9 months [Melichar, Lancet Oncol 2015]. The objectives of this phase I trial were to determine the maximum-tolerated dose (MTD) and evaluate the toxicities and clinical activity of alisertib with fulvestrant in patients (pts) with HR+ advanced BC. Methods: In this standard 3+3 dose-escalation phase I study, pts were assigned to two different oral doses of alisertib (40-50 mg BID on days 1-3, 8-10, 15-17 q 28-day cycle) in combination with standard dose fulvestrant (500 mg IM on day 1 and 15 of cycle 1 and then day 1 q 28-day cycle thereafter). Eligibility included HR+ advanced BC, postmenopausal status, measurable disease or nonmeasurable bone disease by RECIST v1.1, ECOG performance status ≤ 1, unlimited prior endocrine therapies, and ≤ 2 chemotherapy regimens in the metastatic setting. Results: Ten pts enrolled September 2014 - April 2015, and 9 were evaluable for the primary endpoint (one excluded due to ineligibility). The median pt age was 59 (range 48, 73). Prior endocrine therapies included AI (9, 100%), fulvestrant (6, 67%), and everolimus/exemestane (5, 56%). Eight pts (89%) had prior chemotherapy. A median of 4 cycles of therapy have been administered (range 1+, 9+). There were no severe (grade 3+) toxicities reported during cycle 1 at either dose level, thus the MTD was not reached. The cycle 1 grade 1/2 adverse events regardless of attribution were fatigue (6, 67%), neutropenia (5, 56%), anemia (5, 56%), leukopenia (4, 44%), diarrhea (3, 33%), nausea (3, 33%), and mucositis (1, 11%). As of June 3, 2015, 2 pts have discontinued treatment due to disease progression, and 7 remain on treatment with stable disease (Table). One pt with bone only disease had a near CR on PET scan. Dose LevelAlisertib Dose (BID)Treatment Cycles≥ Grade 3 Toxicity, All CyclesProgression-Free Survival (days)1 (n=3)40 mg4, 7+, 9+ 117, 170+, 223+2 (n=6)50 mg1+, 2, 3+, 3+, 4+, 5+grade 4 neutropenia (1 pt)28+, 56, 56+, 57+, 112+, 116++ indicates patients still receiving treatment Conclusion: Alisertib in combination with fulvestrant was well-tolerated. The recommended phase II dose is 50 mg twice daily on days 1-3, 8-10, and 15-17 q 28-day cycle with standard dose fulvestrant. Promising antitumor activity was observed. Correlative tissue evaluation of AURKA expression and other EMT biomarkers is underway. Funding: This work was funded by Takeda Oncology and supported by NIH Grant K12 CA90628 [TCH]. Citation Format: Haddad TC, D'Assoro AB, Suman VJ, Opyrchal M, Goetz MP, Ingle JN. A phase I trial to evaluate the safety of the addition of alisertib to fulvestrant in hormone receptor positive (HR+), advanced breast cancer. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P6-13-04.
- Published
- 2016
- Full Text
- View/download PDF
13. Abstract P3-07-51: Regulation of DNA methyltransferases via TRAF6 determines breast cancer response to decitabine
- Author
-
Judy C. Boughey, Katie N. Jones, Jason P. Sinnwell, JN Ingle, Richard Gray, Donald W. Northfelt, DW Visscher, Richard M. Weinshilboum, Iii Copland Ja, Sara J. Felten, Ping Yin, Ann M. Moyer, L Wang, Jia Yu, Zhenkun Lou, Matthew P. Goetz, TJ Docter, Kevin J. Thompson, Bo Qin, VJ Suman, A Moreno Aspitia, Sarah A. McLaughlin, Eric D. Wieben, Krishna R. Kalari, and Amy Lynn Conners
- Subjects
Cancer Research ,Methyltransferase ,Decitabine ,Methylation ,Biology ,medicine.disease_cause ,Molecular biology ,DNA methyltransferase ,Oncology ,DNA methylation ,medicine ,DNMT1 ,Epigenetics ,Carcinogenesis ,medicine.drug - Abstract
Background: Tumorigenesis involves both genetic and epigenetic changes. Epigenetic alterations are reversible and are promising cancer therapeutic targets. Decitabine (5-aza-2'-deoxycytidine), a DNA methyltransferase inhibitor, is FDA approved for hematological malignancies. However, the effect of decitabine in breast cancer is not completely understood. Previous reports indicated that one decitabine mechanism involves regulation of protein levels for DNMT1, the major DNA methyltransferase that methylates hemimethylated CpG di-nucleotides in DNA. However, the E3 ligase involved in this process has not been identified. Whether decitabine also regulates DNMT3A and 3B in a similar fashion remains unclear. Therefore, our goals were to 1) understand mechanisms underlying decitabine action, 2) test the antitumor activity of decitabine in breast cancer models and 3) identify biomarkers associated with response to decitabine. Methods and Results: Western blots of breast cancer cell lines showed that DNMT1, DNMT3A, and DNMT3B protein levels decreased following decitabine treatment without a reduction in mRNA levels. Bioinformatic analysis of DNA methyltransferase sequences revealed a potential TRAF6 binding motif, and the interaction with TRAF6 (TNF receptor-associated factor 6) was confirmed by IP. TRAF6 functions as an E3 ligase. To determine whether TRAF6 might be the E3 ligase responsible for the degradation of DNMTs after decitabine treatment, we knocked down TRAF6 by RNA interference or knocked out the TRAF6 gene by CRISPR/Cas9. Down regulation of TRAF6 attenuated DNMT ubiquitination and increased DNMT protein levels, suggesting that TRAF6 might mediate proteasome-dependent degradation of all three DNMTs. This was further confirmed by reconstituting the knockout cells with WT and a TRAF6-C70A mutant, followed by assessing DNMT protein levels. Global DNA methylation was also increased after TRAF6 depletion and was confirmed in TRAF6 knock out cells in which DNMT levels were unaffected by decitabine. Cell cytotoxicity and colony forming assays showed that TRAF6 knockout cells were resistant to decitabine, suggesting that a major decitabine mechanism of action is through the regulation of TRAF6 which, in turn, degrades DNMTs, leading to decreased global methylation. Finally, decitabine significantly induced TRAF6 at both mRNA and protein levels, a process that might create positive feedback leading to increased degradation of DNMT proteins upon decitabine treatment. Based on these results, we further hypothesized that levels of the three DNMTs might influence decitabine response. Using 18 breast cancer patient derived xenograft (PDX) models, we found a wide range of DNMT protein levels regardless of ER/HER2 status. DNMT levels in the PDX models were directly associated with sensitivity to decitabine treatment, confirming our hypothesis. Conclusion: Our data showed that decitabine might be an effective agent for treating breast cancer and revealed a novel mechanism underlying decitabine treatment. Baseline DNMT protein levels may serve as a biomarker for predicting decitabine drug response. Citation Format: Yu J, Qin B, Boughey JC, Moyer AM, Visscher DW, Sinnwell JP, Yin P, Thompson KJ, Docter TJ, Kalari KR, Suman VJ, Wieben ED, Felten SJ, Conners AL, Jones KN, McLaughlin SA, Copland JA III, Moreno Aspitia A, Northfelt DW, Gray RJ, Ingle JN, Lou Z, Weinshilboum R, Goetz MP, Wang L. Regulation of DNA methyltransferases via TRAF6 determines breast cancer response to decitabine. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P3-07-51.
- Published
- 2016
- Full Text
- View/download PDF
14. Abstract P2-08-01: Prospective trial of endocrine therapy alone in patients with estrogen receptor positive, HER2-negative, node-negative breast cancer: Results of the TAILORx low risk registry
- Author
-
Charles E. Geyer, J Zujweski, Timothy F. Goggins, Jeffrey L. Berenberg, Daniel F. Hayes, M. Keane, Kathleen I. Pritchard, Adam Brufsky, JN Atkins, IA Mayer, Deborah Toppmeyer, KS Albain, GW Sledge, Virginia G. Kaklamani, E. A. Perez, Elizabeth Claire Dees, H. L. Gomez Moreno, John A. Olson, Joseph A. Sparano, DF Makower, Richard Gray, and RP Reddi
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Population ,Estrogen receptor ,03 medical and health sciences ,Breast cancer ,Internal medicine ,medicine ,education ,Gynecology ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,030503 health policy & services ,Cancer ,medicine.disease ,Primary tumor ,0305 other medical science ,business ,Oncotype DX ,Tamoxifen ,Mastectomy ,medicine.drug - Abstract
Background: The Trial Assigning Individualized Options for Treatment (TAILORx) is a prospective trial evaluating the role of endocrine therapy or chemoendocrine therapy in patients 18-75 years of age with estrogen receptor (ER)-positive, HER2-negative breast cancer, a primary tumor between 0.6-5.0 cm, and negative axillary nodes, a population for whom chemotherapy is typically recommended or least considered based on National Comprehensive Cancer Center Network (NCCN) guidelines. Methods: The trial was designed to demonstrate non-inferiority of endocrine therapy compared with chemoendocrine therapy in the randomized group with an Oncotype DX Recurrence Score (RS) of 11-25. Patients with a low RS < 11 were assigned to endocrine therapy alone and with a high RS > 25 assigned to chemoendocrine therapy, and both groups were followed in a prospective registry. The definition of an intermediate RS differed in this trial (RS 11-25) from the original reports (RS 18-30) in order to reduce the risk of chemotherapy undertreatment in patients with a mid-range or low RS (Sparano & Paik. J Clin Oncol 2008; 26:721-728). Results: The trial enrolled 10,273 patients between April 2006 and October 2010, of whom 6907 patients (67.2%) had a mid-range RS of 11-25, 1737 (16.9%) had a high RS > 25, and 1639 (15.9%) had a low RS of < 11. At the fourth planned interim analysis, the ECOG-ACRIN data monitoring committee recommended that the study continue as planned for the randomized group with a RS 11-25, and that the results be released to the investigators for the low risk group with a RS 60 (39%); tumor size < 1 cm (8%), 1-2 cm (61%), > 2 cm (31%); histologic grade low (34%), intermediate (59%), high (7%); breast conservation (68%) or mastectomy (32%). Initial endocrine therapy included tamoxifen in 35%, aromatase inhibitors in 59%, ovarian function suppression in 3%, and unspecified therapy in 3%; 5 patients received adjuvant chemotherapy (1 of whom relapsed). Five-year rates (and 95% confidence intervals [CI]) for low RS group were 99.2% (98.5, 99.6%) for distant relapse free interval, 98.5% (97.7, 99.1%) for relapse-free interval, 93.7% (92.2, 94.9%) for invasive disease free survival, and 98.2% (97.3, 98.7%) for overall survival. Information regarding ER, PR, and HER2 RNA expression will be presented. Conclusions: Despite meeting guidelines for recommending or at least considering adjuvant chemotherapy based on classical clinicopathologic features, the risk of recurrence was very low at 5 years in patients with ER-positive, HER2-negative, axillary node-negative breast cancer and a low RS of < 11 treated with endocrine therapy alone without chemotherapy. Citation Format: Sparano JA, Gray RJ, Makower DF, Pritchard KI, Albain KS, Hayes DF, Geyer Jr CE, Dees EC, Perez EA, Olson Jr JA, Zujweski J, Keane MM, Gomez Moreno HL, Reddi RP, Goggins TF, Mayer IA, Brufsky AM, Toppmeyer DL, Kaklamani VG, Atkins JN, Berenberg JL, Sledge Jr GW. Prospective trial of endocrine therapy alone in patients with estrogen receptor positive, HER2-negative, node-negative breast cancer: Results of the TAILORx low risk registry. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P2-08-01.
- Published
- 2016
- Full Text
- View/download PDF
15. Abstract 5774: Indo-Canadian parternship for oral cancer screening in rural India
- Author
-
Simon Sutcliffe, Sandra Broughton, Jean Jacob, Shyam Ndvn, Madhurima Datta, Priya Chandran, Nallan Csk Chaitanya, Gayatri Palat, Denise M. Laronde, Jagannath Jn, and Vineela Rapelli
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Mortality rate ,Cancer ,medicine.disease ,Oncology ,Family medicine ,medicine ,Rural area ,Family history ,business ,Developed country ,Survival rate ,Socioeconomic status ,Gutka - Abstract
Aims: Oral cancer is ranked among the top three commonly occurring cancers in India with over 120,000 cases reported annually. Advanced stage diagnosis is common due to poor access to healthcare and low socioeconomic status, limiting the 5-year survival rate to a dismal 27%. In India, 60-80% of patients present with advanced disease as compared to 40% in developed countries. Oral cancer screening is a quick, painless, and non-invasive exam to detect suspicious lesions that can reduce mortality rates significantly. The aim of the project was to determine the feasibility of conducting oral cancer screening in rural areas around Hyderabad, India in collaboration with Two Worlds Cancer Collaboration, Canada; MNJ Cancer Hospital, India and two dental colleges in Hyderabad. Methods: Planning and communication between the Indian and Canadian partners were facilitated by Zoom videoconferencing. Ethics approval for the feasibility phase was obtained in both countries. A feasibility study was planned in a rural village. Prior to screening, the Urulla village community school required infrastructure changes such as furniture and electricity. Fluorescence Visualization (FV) units were shipped from Canada to India to assess if they improve the sensitivity and specificity of screening in this setting. The Canadian team trained the Indian clinicians on use of FV and study methodology. The feasibility study was conducted in March 2018 where participants received a detailed risk habit evaluation, intraoral and extraoral examination. Results: A total of 114 participants were screened, 62% males and 38% females with a median age of 55 years. Four percent had a family history of oral cancer, 33% had a smoking history of which the majority (65%) smoked beedis. Nineteen percent chewed pan, gutka or other chewing substances. Seventy-one percent drank alcohol, primarily beer/toddy (65%). Screening data was complete for 112 participants of which 16 (14.2%) had intraoral lesions. Lesion presence was not significantly associated with smoking, chewing habit or alcohol drinking (P=0.370, P=0.207, P=0.393). The majority of lesions were on the labial and buccal mucosa (79%). Forty-three percent of lesions showed a loss of fluorescence. Out of the 16 lesions, 7 lesions were clinically deemed as high-risk and recommended to undergo biopsy. Loss of fluorescence was noted in 6 out of 7 lesions, and was highly significant with high-risk lesion presence (p Conclusion: The high evidence of disease warrants the need for oral cancer screening in rural India. The feasibility study paved way for the pilot study which aims to screen over 1000 participantsin rural areas around Hyderabad using additional adjunct tools like FV and Quantitative Cytology to improve accuracy and semi-automate the screening process. Acknowledgements: This study was supported through fundraising efforts by Two Worlds Cancer Collaboration, an NPO based in Kelowna, Canada. Two Worlds aims to provide cancer palliative care in low resource countries. Citation Format: Madhurima Datta, Nallan CSK Chaitanya, Shyam Ndvn, Gayatri Palat, Jean Jacob, Priya Chandran, Vineela Rapelli, Jagannath Jn, Sandra Broughton, Simon Sutcliffe, Denise Marie Laronde. Indo-Canadian parternship for oral cancer screening in rural India [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5774.
- Published
- 2020
- Full Text
- View/download PDF
16. Abstract P2-03-09: Autocrine motility factor signaling pathway promotes aggressive behavior and migration in breast cancer
- Author
-
Pamela A. Althof, AZ VanDyke, SP Thayer, D Maroni, JN Sanmann, Prosenjit Mondal, Cassie J Liu, JD Price, and JM Stevens
- Subjects
Cancer Research ,Autocrine Motility Factor ,Breast cancer ,Oncology ,medicine ,Cancer research ,Signal transduction ,Biology ,medicine.disease - Abstract
Background: Autocrine motility factor (AMF) is secreted by cancer cells and acts in an autocrine or paracrine fashion to bind to its receptor, AMFR, at the cell surface of tumor cells. The AMF-AMFR pathway has been shown to promote proliferation, anti-apoptosis, motility and migration, invasion, and metastasis pathways in various cancers. However, our understanding of the AMF-AMFR pathway in breast cancer is limited. We propose that the AMF signaling pathway is an unexplored mechanism for breast cancer tumor aggression and its receptor, AMFR, may be a potential therapeutic target. Methods: Tumor tissue obtained from consented female (n = 31) and male (n = 1) patients were analyzed by the Affymetrix OncoscanTM genome-wide microarray platform and examined for somatic copy-number alterations (SCNAs) of AMFR. cBioPortal was used to investigate SCNA of AMFR and gene expression of AMF on primary breast cancer tumors from METABRIC (n = 1,784) and TCGA Pan-Cancer Atlas (n = 981) datasets. In vitro, AMF and AMFR gene expression in luminal A (MCF-7) and triple-negative breast cancer (MDA-MB-231) cell lines were assessed by qPCR. Cell migration assays were performed on MDA-MB-231 cells to investigate their migration towards AMF with AMFR present or knocked down by siRNA. Results: Microarray analysis of 32 tumors revealed that a single-copy loss of AMFR occurred 79% of the time in luminal A tumors (n = 19); 67% of the time in luminal B tumors (n = 6); 33% of the time in ER+, PR+, HER2+ tumors (n = 3); and 0% of the time in triple-negative breast cancer (TNBC) tumors (n = 4), suggesting that the loss of AMFR results in less aggressive tumors that have good overall prognosis. To extend our findings to a larger patient cohort, SCNA analysis of the METABRIC and TCGA Pan-Cancer Atlas datasets revealed that single-copy loss of AMFR occurred in 64.53% and 68.14% of luminal A tumors, 58.51% and 59.90% of luminal B tumors, 23.62% and 48.72% of HER2 overexpression tumors, and 29.80% and 39.77% of TNBC tumors, respectively. Therefore, AMFR appears most frequently deleted in the tumor genomes of good prognosis breast cancer molecular subtypes (luminal tumors). Gene expression analysis of AMF in the METABRIC (using z-scores) and TCGA Pan-Cancer Atlas (using batch-normalized numbers) datasets revealed median mRNA expressions of -0.3351 and 4862 in luminal A tumors, -0.05415 and 5841 in luminal B tumors, 0.5758 and 9390 in HER2 overexpression tumors, and 0.754 and 8798 in TNBC tumors, respectively, suggesting that the AMF-AMFR pathway is more active in aggressive breast cancers. Similarly, we observed that AMF and AMFR are transcriptionally overexpressed by 7-fold and 16-fold, respectively, in the TNBC cell line MDA-MB-231 compared to the luminal A breast cancer cell line MCF-7. When AMFR is knocked down in MDA-MB-231 cells, focused migration towards AMF is abolished. Conclusion: AMF-AMFR pathway activity correlates with aggressive cancer cell behavior and enhanced migration in breast cancer. Single-copy loss of AMFR in tumor genomes is associated with less aggressive tumors with better overall prognosis. AMFR may be an attractive therapeutic target. Citation Format: Liu C, Price JD, Maroni D, Stevens JM, VanDyke AZ, Althof PA, Mondal P, Sanmann JN, Thayer SP. Autocrine motility factor signaling pathway promotes aggressive behavior and migration in breast cancer [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P2-03-09.
- Published
- 2019
- Full Text
- View/download PDF
17. Abstract P3-08-01: Characteristics, outcomes and prognostic factors of luminal androgen receptor (LAR) triple-negative breast cancer (TNBC)
- Author
-
Leon-Ferre, RA, primary, Polley, M-Y, additional, Liu, H, additional, Kalari, KR, additional, Boughey, JC, additional, Liu, MC, additional, Cafourek, V, additional, Negron, V, additional, Ingle, JN, additional, Thompson, KJ, additional, Tang, X, additional, Barman, P, additional, Carlson, E, additional, Visscher, DW, additional, Carter, JC, additional, Couch, FJ, additional, and Goetz, MP, additional
- Published
- 2019
- Full Text
- View/download PDF
18. Abstract P5-04-05: Glucocorticoid receptor activation inhibits proliferation of endoxifen resistant breast cancer cells and resensitizes cells to hormonal therapy
- Author
-
Jones, CJ, primary, Goetz, MP, additional, Ingle, JN, additional, and Hawse, JR, additional
- Published
- 2019
- Full Text
- View/download PDF
19. Abstract P1-17-03: Statin use, site of recurrence, and survival among post-menopausal women taking bisphosphonates as adjuvant therapy for breast cancer (SWOG S0307)
- Author
-
Kizub, D, primary, Miao, J, additional, Stopeck, A, additional, Thompson, P, additional, Paterson, AH, additional, Clemons, M, additional, Dees, EC, additional, Ingle, JN, additional, Falkson, CI, additional, Barlow, W, additional, Hortobagyi, GN, additional, and Gralow, JR, additional
- Published
- 2019
- Full Text
- View/download PDF
20. Abstract GS2-01: Age-related breast cancer risk estimates for the general population based on sequencing of cancer predisposition genes in 19,228 breast cancer patients and 20,211 matched unaffected controls from US based cohorts in the CARRIERS study
- Author
-
Couch, FJ, primary, Hu, C, additional, Hart, SN, additional, Gnanaolivu, RD, additional, Lilyquist, J, additional, Lee, KY, additional, Gao, C, additional, Eckloff, B, additional, Samara, R, additional, Klebba, J, additional, Auer, P, additional, Bernstein, L, additional, Gaudet, M, additional, Haiman, C, additional, Palmer, JR, additional, Yao, S, additional, Domchek, SM, additional, Weitzel, JN, additional, Goldgar, DE, additional, Nathanson, KL, additional, Kraft, P, additional, and Polley, EC, additional
- Published
- 2019
- Full Text
- View/download PDF
21. Abstract P2-03-09: Autocrine motility factor signaling pathway promotes aggressive behavior and migration in breast cancer
- Author
-
Liu, C, primary, Price, JD, additional, Maroni, D, additional, Stevens, JM, additional, VanDyke, AZ, additional, Althof, PA, additional, Mondal, P, additional, Sanmann, JN, additional, and Thayer, SP, additional
- Published
- 2019
- Full Text
- View/download PDF
22. Abstract P1-03-04: Molecular subtyping of androgen receptor-positive patients using gene expression profiles
- Author
-
Thompson, KJ, primary, Alaparthi, T, additional, Sinnwell, JP, additional, Carlson, EE, additional, Tang, X, additional, Bockol, M, additional, Vedell, PT, additional, Ingle, JN, additional, Suman, V, additional, Weinshilboum, RM, additional, Wang, L, additional, Boughey, JC, additional, Kalari, KR, additional, and Goetz, MP, additional
- Published
- 2019
- Full Text
- View/download PDF
23. Abstract P5-11-10: Biomarkers associated with resistance or response to CDK4/6 treatment in patients with metastatic hormone-receptive positive breast cancer
- Author
-
Mullins, JN, primary, Chaudhry, A, additional, Ryder, J, additional, Valasareddy, P, additional, Jain, A, additional, Ranganath, H, additional, Hare, F, additional, and Vidal, GA, additional
- Published
- 2019
- Full Text
- View/download PDF
24. Abstract P3-01-21: Circulating tumor cells of breast cancer origin identified by fluorescence in situ hybridization and may be an early predictor of therapy failure in early breast cancer
- Author
-
Liu, C, primary, Althof, PA, additional, Maroni, D, additional, Stevens, JM, additional, Grabow, CE, additional, VanDyke, AZ, additional, Price, JD, additional, Sanmann, JN, additional, and Thayer, SP, additional
- Published
- 2019
- Full Text
- View/download PDF
25. Abstract P5-07-01: Successful whole transcriptome analysis of 25-year-old breast tumor samples from the phase III trial SWOG-8814 by next generation sequencing (NGS): Standardized analytical methods for exploratory and validation studies
- Author
-
Daniel F. Hayes, Julie Gralow, Lisa A. Carey, EM Beasley, S Shak, Kunbin Qu, JN Ingle, Kathleen I. Pritchard, Robert B. Livingston, Gabriel N. Hortobagyi, F Collin, Audrey Goddard, CK Osborne, Peter M. Ravdin, W. Barlow, KS Albain, Diana B. Cherbavaz, Crager, Amy P. Sing, MC Liu, FL Baehner, Nancy E. Davidson, James M. Rae, Debu Tripathy, and J-H Jeong
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Adjuvant chemotherapy ,medicine.disease ,Bioinformatics ,DNA sequencing ,Breast tumor ,Transcriptome ,Breast cancer ,Quartile ,Internal medicine ,Medicine ,Median absolute deviation ,business ,Science study - Abstract
BACKGROUND: We previously reported that low 21 gene Recurrence Scores (RS) identify patients with ER-positive, lymph node-positive breast cancer who may not benefit from anthracycline-based adjuvant chemotherapy added to tamoxifen (SWOG-8814A NCI correlative science study; Albain et al. Lancet Oncol 2010). New exploratory and comprehensive quantitative analyses now permit whole transcriptome NGS on residual RNA extracted from FFPE blocks 12-18 years post-fixation. Herein, we report methodology details and feasibility results (see companion abstract, Albain et al., for clinical outcomes correlations). METHODS and RESULTS: Sequencing was carried out in Illumina HiSeq 2000 instruments, yielding 4.2 trillion data points. Messenger RNA expression was quantified using 3rd quartile normalization. Both Library (RNA) and Sequencing Standards showed high quality coverage as measured by median uniquely mapped reads over a 13 month window (168M and 182M, respectively, including duplicate reads). The median absolute deviation (MAD) of the relative log expression (RLE) of mapped reads for the Library and Sequencing Standards was 0.22 and 0.05, respectively. The Library Standard variation was greater than the Sequencing Standard, as library preparation was manual. Of 360 patient samples with sufficient RNA (≥ 100 ng total RNA), 354 (98.3%) were successfully sequenced and included in the final analysis data set. Average library yield was 39 ng/μL. Only 5 libraries failed yield requirements and one library failed expression quality metrics. The median insert length was 120 bp with the first and third quartiles 93 and 152 bp, respectively. After removal of duplicate reads, 82% of reads were uniquely mapped, and the median library size was 8.95M (number of unique mapped reads). Sequences with counts CONCLUSIONS: High quality whole transcriptome NGS is feasible from decades-old clinical trial FFPE specimens that have not been stored in any special fashion. Controlled laboratory, bioinformatics and biostatistics methods, with inclusion of appropriate process controls, ensure robustness and reliability of the NGS process. This in turn results in the discovery and validation of biologically and clinically relevant variations from prior landmark clinical trials. SUPPORT: NCI CA 180888, CA180819, CA180821, CA180820, CA180863; in part, Genomic Health, Inc. Citation Format: Cherbavaz DB, Hayes DF, Qu K, Crager MR, Barlow WR, Goddard AD, Beasley EM, Jeong J, Collin F, Liu M-L, Rae JM, Ravdin PM, Tripathy D, Gralow JR, Livingston RB, Osborne CK, Ingle JN, Pritchard KI, Davidson NE, Carey LA, Sing AP, Baehner FL, Hortobagyi GN, Shak S, Albain KS. Successful whole transcriptome analysis of 25-year-old breast tumor samples from the phase III trial SWOG-8814 by next generation sequencing (NGS): Standardized analytical methods for exploratory and validation studies. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P5-07-01.
- Published
- 2016
- Full Text
- View/download PDF
26. Abstract GS1-02: NSABP B-47 (NRG oncology): Phase III randomized trial comparing adjuvant chemotherapy with adriamycin (A) and cyclophosphamide (C) → weekly paclitaxel (WP), or docetaxel (T) and C with or without a year of trastuzumab (H) in women with node-positive or high-risk node-negative invasive breast cancer (IBC) expressing HER2 staining intensity of IHC 1+ or 2+ with negative FISH (HER2-Low IBC)
- Author
-
Virginia F. Borges, Eleftherios P. Mamounas, Jonathan Polikoff, Norman Wolmark, KS Albain, P Rastogi, TD Moore, Reena S. Cecchini, Louise Provencher, JN Atkins, Soonmyung Paik, Joseph P. Costantino, Sandra M. Swain, C Stokoe, Louis Fehrenbacher, J-F Boileau, Charles E. Geyer, and André Robidoux
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Cyclophosphamide ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,03 medical and health sciences ,Regimen ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Docetaxel ,Trastuzumab ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,business ,Prospective cohort study ,medicine.drug - Abstract
Background: Adjuvant trastuzumab (H) reduces cancer recurrence and improves survival in patients (pts) with HER2-amplified or overexpressing (IHC 3+ staining intensity) IBC. Two of the landmark trials that demonstrated the efficacy of H-based eligibility on HER2 testing performed at local site laboratories were found to contain a cohort of pts without amplification or IHC overexpression on tissue submitted for central testing. These HER2-low cohorts appeared to benefit from the addition of H, and efforts at external HER2 testing validation and laboratory explorations did not negate these findings. NSABP B-47 was performed to determine if these findings would be confirmed in a large prospective randomized trial. The primary aim was to determine whether the addition of H to chemotherapy (CT) regimens of AC→WP or TC (choice per investigator discretion) would improve invasive disease-free survival (IDFS). Methods: From 2/8/2011 to 2/10/2015, 3270 women were enrolled with 1630 pts randomly assigned to Arm 1 [TC: docetaxel 75mg/m2, cyclophosphamide 600 mg/m2 every 3 weeks x 6 cycles; or AC→WP: doxorubicin 60 mg/m2 and cyclophosphamide 600 mg/m2 every 2 or 3 weeks x 4 cycles followed by paclitaxel 80 mg/m2 every week x 12], and 1640 pts to Arm 2 [same CT regimens + 12 months of H]. Pts were stratified by IHC score (1+ vs 2+), number of positive nodes (0-3, 4-9, ≥10), hormone receptor status (ER or PgR positive vs both negative), and CT (TC vs AC→WP). Overall 58.5% were ≥50 years, 57% had tumors with IHC 1+, 17.3% were ER- and PgR-, 19.9% were node negative, and 27.4% had ≥4 positive nodes. TC was the intended CT regimen for 44.2%. Results: As of 7/31/2017, the median follow-up time was 46.1 months. We observed 264 IDFS events, which triggered the definitive analysis for the primary endpoint. The addition of H to CT showed a 5-year IDFS of 89.6% compared to 89.2% for CT alone (HR 0.98; 95%CI 0.77-1.26; P=0.90). The findings did not differ by level of HER2 IHC expression, level of lymph node involvement, or hormone receptor status. 5-year point estimates for RFI were 92.0% for CT+H compared to 92.2% for CT alone (HR 0.995; 95%CI 0.75-1.32; P=0.97). 5-year estimates for DRFI were 92.7% for CT+H and 93.5% for CT alone (HR 1.10; 95%CI 0.81-1.49; P=0.55). The addition of H did not change OS significantly with 5-year point estimates of 94.8% in CT+H vs 96.2% in CT alone (HR 1.33; 95%CI 0.91-1.94; P=0.14). 4.3% of women in the CT arm experienced Grade 4 or 5 toxicities compared to 5.0% in CT+H. Conclusion: The addition of H to CT did not demonstrate a reduction in IDFS, RFI, or DRFI in women with non-overexpressing but IHC measurable HER2 IBC. This prospective study did not confirm the retrospective findings in NSABP B-31 or N9831. The threshold of HER2 expression or genetic amplification for H benefit remains unchanged. Support: NCI U10-180868, -180822, -44066, UG1-189867, and Genentech, Inc. Citation Format: Fehrenbacher L, Cecchini RS, Geyer CE, Rastogi P, Costantino JP, Atkins JN, Polikoff J, Boileau J-F, Provencher L, Stokoe C, Moore TD, Robidoux A, Borges V, Albain KS, Swain SM, Paik S, Mamounas EP, Wolmark N. NSABP B-47 (NRG oncology): Phase III randomized trial comparing adjuvant chemotherapy with adriamycin (A) and cyclophosphamide (C) → weekly paclitaxel (WP), or docetaxel (T) and C with or without a year of trastuzumab (H) in women with node-positive or high-risk node-negative invasive breast cancer (IBC) expressing HER2 staining intensity of IHC 1+ or 2+ with negative FISH (HER2-Low IBC) [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr GS1-02.
- Published
- 2018
- Full Text
- View/download PDF
27. Abstract P5-05-03: Estrogen receptor beta elicits anti-cancer effects in triple negative breast cancer through suppression of NFκB signaling
- Author
-
MP Goetz, Zhifu Sun, Jason S. Carroll, John R. Hawse, Zhenqing Ye, Igor Chernukhin, Adam W Nelson, Kirsten G M Aspros, and JN Ingle
- Subjects
Cancer Research ,EZH2 ,Cancer ,Biology ,medicine.disease ,Transcriptome ,Oncology ,Cistrome ,Histone methyltransferase ,Cancer research ,medicine ,Chromatin immunoprecipitation ,Estrogen receptor beta ,Triple-negative breast cancer - Abstract
Background: Triple Negative Breast Cancer (TNBC) affects approximately 15-20% of BC patients, yet accounts for a disproportionately higher rate of BC morbidity and mortality, in part due to lack of targeted therapies. Using well-validated antibodies, Estrogen Receptor Beta (ERβ) protein has been shown to be expressed in approximately 25% of TNBCs and is associated with improved patient outcomes. Using multiple ERβ +/- TNBC cell lines and PDX models, we have demonstrated that ligand-mediated activation of ERβ by estradiol (E2) decreases cell proliferation, invasion, and migration in vitro, as well as primary tumor growth and metastatic spread in vivo. Methods: To determine the mechanisms by which ERβ elicits these anti-cancer effects, we elucidated the ERβ transcriptome and cistrome via Microarray and ChIPseq, respectively, in TNBC cells stably expressing ERβ in a doxycycline-inducible manner. We also performed gene expression and luciferase assays to assess the impact of ERβ on NFκB signaling, followed by ChIP-PCR and ChIPseq to assess how ERβ modifies chromatin architecture near NFκB target genes. Results: Pathway analysis of ERβ-regulated genes identified NFκB signaling as one of the most suppressed pathways in response to E2 treatment. Indeed, numerous NFκB target genes were among the most down-regulated genes following E2 treatment but only in the presence of ERβ expression. Chromatin Immunoprecipitation followed by sequencing (ChIPseq) revealed that ERβ primarily associated with estrogen response elements (EREs), but was also enriched around NFκB binding sites following E2 treatment. In fact, 12% of all ERβ binding sites were enriched for NFκB response elements and ERβ was shown to physically associate with NFκB protein. Using an NFκB reporter construct and qPCR, ERβ was shown to block TNFα-mediated induction of NFκB signaling and NFκB target gene expression. Globally, RNAseq identified 200 genes to be significantly regulated by TNFα in TNBC cells, of which 81 were significantly altered in the presence of E2+TNFα. ChIPseq demonstrated that ligand-mediated activation of ERβ significantly diminished an activating histone mark (H3K27Ac) at many of these NFκB target genes while enhancing a repressive mark (H3K27Me3). These modifications are also associated with recruitment of the histone methyltransferase, EZH2, to enhancer elements of these NFκB target genes. Drug-mediated blockade of HDAC and EZH2 activity reversed suppression of NFκB target gene expression by ERβ. Conclusions: Our data suggest that ERβ may elicit its anti-cancer effects in part via formation of a novel co-repressor complex consisting of ERβ, NFκB, and EZH2. These data are in keeping with prior observations of the importance of NFκB signaling as it relates to TNBC cell proliferation and invasion, and that decreased expression of NFκB target genes is associated with improved outcomes in TNBC patients. Currently, a Mayo Breast SPORE prospective study is underway to investigate the role of estradiol in ERβ expressing TNBC and to further evaluate the cross-talk between ERβ and NFκB signaling in TNBC. Citation Format: Aspros K, Nelson A, Ye Z, Sun Z, Chernukhin I, Carroll J, Ingle J, Goetz M, Hawse J. Estrogen receptor beta elicits anti-cancer effects in triple negative breast cancer through suppression of NFκB signaling [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P5-05-03.
- Published
- 2019
- Full Text
- View/download PDF
28. Abstract P5-04-05: Glucocorticoid receptor activation inhibits proliferation of endoxifen resistant breast cancer cells and resensitizes cells to hormonal therapy
- Author
-
Calley J. Jones, John R. Hawse, MP Goetz, and JN Ingle
- Subjects
Endoxifen ,Cancer Research ,Glucocorticoid receptor ,Oncology ,business.industry ,Cancer research ,Hormonal therapy ,Medicine ,Breast cancer cells ,business - Abstract
Background: Despite the prevalent treatment options for ERα-positive breast cancer patients, and their initial efficacy for many women, ERα-positive disease still accounts for more breast cancer related deaths than any other subtype. Relapse in these patients is largely due to the development of resistance to anti-estrogen therapies such as tamoxifen. While tamoxifen and its resistance mechanisms have been extensively studied from both the bench and the bedside, relatively little is known about its active metabolite endoxifen. Our group has provided evidence that endoxifen is the most potent and clinically relevant metabolite of tamoxifen, suggesting that its characterization may be crucial to understanding tamoxifen resistance. Methods: We have developed novel endoxifen resistant MCF7 and T47D cell lines through chronic exposure to endoxifen during a period of 12-24 months. Using these models and their respective controls, we compared global gene expression profiles of endoxifen resistant cells to tamoxifen resistant cells and found marked differences between the two models. Additionally, we subjected treatment naïve cells to a genome-wide, CRISPR-mediated knockout screen to identify genes, and their associated pathways, that are likely involved in mediating endoxifen resistance. Results: Analysis of CRISPR guide RNAs enriched or depleted in response to chronic endoxifen treatment revealed that disruption of genes regulated by dexamethasone (Dex), a potent glucocorticoid receptor (GR) agonist, enhanced cells' ability to survive and proliferate in the presence of endoxifen. These data suggest that GR activation may inhibit endoxifen resistance, and that treatment of resistant cells with Dex may restore endoxifen efficacy. Indeed, Dex treatment significantly inhibited the proliferation rates of endoxifen resistant cells by 50-60% with little to no inhibitory effects in endoxifen sensitive models. Further, Dex was shown to synergize with endoxifen in resistant cells to further suppress cell proliferation, implying that Dex treatment could be utilized as an effective therapy for endocrine resistant disease. Conditioned media harvested from cells chronically exposed to Dex also resulted in substantial inhibition of endoxifen resistant cell proliferation rates. To explore potential mechanisms of these effects, we performed RNA-seq on both treatment-naïve and endoxifen resistant cells following Dex treatment. Out of 246 genes significantly regulated by Dex in endoxifen resistant cells, we identified 61 genes that were not differentially regulated in treatment naïve cells. These genes may provide insights into the mechanisms of GR activity specific to endoxifen resistant cells. Conclusions: To our knowledge, we have developed the first models of endoxifen resistance and have demonstrated that global transcriptomic changes that occur during this process are substantially different than those observed in tamoxifen resistant models. We have shown that activation of GR signaling elicits significant growth-inhibitory effects specifically in the setting of endoxifen resistance. These data identify the GR pathway as a potential novel therapeutic target for the treatment of endocrine resistant breast cancer. Citation Format: Jones CJ, Goetz MP, Ingle JN, Hawse JR. Glucocorticoid receptor activation inhibits proliferation of endoxifen resistant breast cancer cells and resensitizes cells to hormonal therapy [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P5-04-05.
- Published
- 2019
- Full Text
- View/download PDF
29. Abstract P3-01-21: Circulating tumor cells of breast cancer origin identified by fluorescence in situ hybridization and may be an early predictor of therapy failure in early breast cancer
- Author
-
JM Stevens, AZ VanDyke, CE Grabow, JN Sanmann, JD Price, Pamela A. Althof, D Maroni, SP Thayer, and Cassie J Liu
- Subjects
Cancer Research ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,Cancer ,SCNA ,medicine.disease ,Primary tumor ,Radiation therapy ,Circulating tumor cell ,Breast cancer ,Oncology ,Adjuvant therapy ,Cancer research ,medicine ,business ,Fluorescence in situ hybridization - Abstract
Background: Most modern methods of detecting circulating tumor cells (CTCs) involve identifying cells with epithelial markers. This approach presents challenges, as not all epithelial cells found in circulation originate from the tumor and not all CTCs express epithelial markers. We propose using a size-exclusion filtration system to enrich for CTCs in peripheral blood followed by fluorescence in situ hybridization (FISH) of the filtered cells to identify cells of tumor origin in the early-stage breast cancer patients. We further hypothesize that the presence of CTCs may be indicators of therapy failure in early-stage breast cancer patients. Methods: Patients diagnosed with breast cancer (n = 9) were consented for CTC evaluation. Primary tumor DNA was analyzed by the Affymetrix OncoscanTM genome-wide microarray platform and investigated for somatic copy-number alterations (SCNAs). For each patient, two FISH probes were then identified for two regions of gain or a region of gain and a region of loss from the microarray results. Blood samples from patients were obtained before surgery, radiation therapy, endocrine therapy, and at 6-month or 1-year follow-up visits. Blood samples were filtered using ScreenCell® Cyto V2 devices, and FISH was performed. Cells were categorized as normal (diploid for all FISH probes), suspicious (single SCNA detected by FISH), or CTC (two SCNAs detected by FISH). Patients were identified as having CTCs present in their circulation when ≥2 CTCs were observed or when one CTC and >15 suspicious cells were observed. Results: The microarray data revealed that luminal A tumors ranged from 2-43 SCNAs; luminal B tumors ranged from 15-20 SCNAs; and ER+, PR+, HER2+ tumors ranged from 46-98 SCNAs. Although a correlation appears to exist between tumor genetic complexity and molecular subtype, the degree of complexity was highly varied within each subtype. We found that neither complexity of tumor profile, molecular subtype, nor stage could predict the presence of CTCs in patients. Molecular SubtypeSCNAsCTCsSuspicious Cells OnlyLuminal A2-433/52/5Luminal B15-200/21/2ER+, PR+, HER2+46-982/20/2 In pre-surgical blood samples, we detected CTCs in 63% of patients with stage 1 disease and in 60% of patients with luminal A tumors, 0% of patients with luminal B tumors, and 100% of patients with triple-positive tumors. StageCTCsSuspicious Cells OnlyIA/B5/82/8IIA0/11/1 Although limited in number, ongoing investigation revealed that one of our patients in early follow-up with a luminal A, stage IB tumor was identified to have persistent CTCs at 1-year after starting hormonal adjuvant therapy, suggesting residual tumor burden not detected by standard clinical modalities; this finding also suggests that this patient may be at highest risk for relapse and should be considered for additional therapies. Conclusion: Size-exclusion filtration followed by FISH analysis can accurately identify CTCs in early-stage breast cancer patients. Tumor complexity, molecular subtype, and stage did not predict the presence of CTCs in circulation. Our method for CTC detection may be able to serve as a diagnostic tool for treatment failure. Citation Format: Liu C, Althof PA, Maroni D, Stevens JM, Grabow CE, VanDyke AZ, Price JD, Sanmann JN, Thayer SP. Circulating tumor cells of breast cancer origin identified by fluorescence in situ hybridization and may be an early predictor of therapy failure in early breast cancer [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P3-01-21.
- Published
- 2019
- Full Text
- View/download PDF
30. Abstract GS4-03: Randomized comparison of adjuvant tamoxifen (T) plus ovarian function suppression (OFS) versus tamoxifen in premenopausal women with hormone receptor-positive (HR+) early breast cancer (BC): Update of the SOFT trial
- Author
-
Fleming, G, primary, Francis, PA, additional, Láng, I, additional, Ciruelos, EM, additional, Bellet, M, additional, Bonnefoi, HR, additional, Climent, MA, additional, Pavesi, L, additional, Burstein, HJ, additional, Martino, S, additional, Davidson, NE, additional, Geyer, CE, additional, Walley, BA, additional, Coleman, RE, additional, Kerbrat, P, additional, Buchholz, S, additional, Ingle, JN, additional, Rabaglio-Poretti, M, additional, Colleoni, M, additional, and Regan, MM, additional
- Published
- 2018
- Full Text
- View/download PDF
31. Abstract P1-06-07: Mayo clinic TNBC outcome calculator: A clinical calculator to predict disease relapse and survival in women with triple-negative breast cancer
- Author
-
Polley, M-YC, primary, Leon-Ferre, RA, additional, Liu, H, additional, Gilbert, J, additional, Cafourek, V, additional, Hillman, DW, additional, Negron, V, additional, Boughey, JC, additional, Liu, MC, additional, Ingle, JN, additional, Kalari, K, additional, Couch, F, additional, Visscher, DW, additional, and Goetz, MP, additional
- Published
- 2018
- Full Text
- View/download PDF
32. Abstract P5-07-01: LncRNA MIR2052HG regulates ERα level and endocrine resistance through LMTK3 by recruiting early growth response protein 1
- Author
-
Cairns, J, primary, Ingle, JN, additional, Shepherd, LE, additional, Kubo, M, additional, Goetz, MP, additional, Weinshilboum, RM, additional, Kalari, KR, additional, and Wang, L, additional
- Published
- 2018
- Full Text
- View/download PDF
33. Abstract GS1-02: NSABP B-47 (NRG oncology): Phase III randomized trial comparing adjuvant chemotherapy with adriamycin (A) and cyclophosphamide (C) → weekly paclitaxel (WP), or docetaxel (T) and C with or without a year of trastuzumab (H) in women with node-positive or high-risk node-negative invasive breast cancer (IBC) expressing HER2 staining intensity of IHC 1+ or 2+ with negative FISH (HER2-Low IBC)
- Author
-
Fehrenbacher, L, primary, Cecchini, RS, additional, Geyer, CE, additional, Rastogi, P, additional, Costantino, JP, additional, Atkins, JN, additional, Polikoff, J, additional, Boileau, J-F, additional, Provencher, L, additional, Stokoe, C, additional, Moore, TD, additional, Robidoux, A, additional, Borges, V, additional, Albain, KS, additional, Swain, SM, additional, Paik, S, additional, Mamounas, EP, additional, and Wolmark, N, additional
- Published
- 2018
- Full Text
- View/download PDF
34. Abstract P3-03-06: Telomerase polymorphism and breast cancer
- Author
-
Alvares, MM, primary, Rodrigues, KS, additional, Matos, JN, additional, and Oliveira, DM, additional
- Published
- 2018
- Full Text
- View/download PDF
35. Abstract P2-09-07: Inhibition of aurora-A by MLN8237 decreases SMAD5 expression and increases effectiveness of chemotherapeutic agents in breast cancer cells
- Author
-
Eva Galanis, JN Ingle, Mateusz Opyrchal, Ianko D. Iankov, and A B D'Assoro
- Subjects
Cancer Research ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,chemistry.chemical_compound ,Breast cancer ,Oncology ,chemistry ,Paclitaxel ,embryonic structures ,Alisertib ,Cancer cell ,Immunology ,Cancer research ,Medicine ,Epithelial–mesenchymal transition ,business ,Adjuvant - Abstract
BACKGROUND: Breast cancer affects thousands of women each year. Epithelial to Mesenchymal transition (EMT) has been associated with increased metastatic potential of cancer cells as well as resistance to chemotherapy. Predicatively, presence of EMT leads to worse prognosis in patients with breast cancer. We have recently showed that Aurora-A plays a key role in development of EMT and increased ability of breast cancer cells for self renewal. Therefore we hypothesized that inhibition of Aurora-A in breast cancer cells will lead to increased sensitivity to chemotherapy. RESULTS: Increased Aurora-A expression, as assessed by IHC staining, in patients with locally advanced disease was associated with worse prognosis. We show that in ER+ cell line, MCF7, activating MAPK pathway through overexpression of Raf1 leads to increase in Aurora-A activity. These cells have decreased sensitivity to treatment with paclitaxel when compared to untransfected cells in MTT assays. This effect is even more exacerbated with over-expression of Aurora-A. The sensitivity to paclitaxel was restored with inhibition of Aurora-A by specific inhibitor, Alisertib. This was further explored in triple negative (MDA-MB-231) or Her-2/neu expressing (SUM149) cell lines. These cells express higher levels of Aurora-A. Inhibition of Aurora-A activity in combination with paclitaxel was superior when compared to either therapy alone. Similar effect was seen with use of anthracyclines. Inhibition of Aurora-A resulted in decreased SMAD5 expression as well as decreased Akt phosphorylation. Current studies are looking at a role of Aurora-A in activating SMAD5 and its role in resistance to chemotherapy. In vivo experiments evaluating combination therapies in breast cancer animal model are ongoing. CONCLUSIONS: Inhibition of Aurora-A by a specific inhibitor, MLN8237, resulted in decreased SMAD5 expression and increased effectiveness of chemotherapeutic agents in breast cancer cells. These results contribute to better understanding of signaling pathways involved in resistance of breast cancer cells to chemotherapy. This knowledge could be very useful in developing more effective treatments for breast cancer patients in neo-adjuvant, adjuvant and metastatic settings. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P2-09-07.
- Published
- 2013
- Full Text
- View/download PDF
36. Abstract P2-06-01: cMethDNA is a quantitative circulating methylated DNA assay for detection of metastatic breast cancer and for monitoring response to therapy
- Author
-
Wei Wen Teo, JN Ingle, CB Umbricht, Mary Jo Fackler, Zhen Zhang, Judy C. Boughey, Stacie Jeter, Zoila Areli Lopez Bujanda, S Sukumar, Kandace P. McGuire, LA Cope, Pedram Argani, Antonio C. Wolff, K Visvanathan, Lisa A. Carey, Ta King, and Clarence Wang
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Cancer ,Methylation ,Biology ,medicine.disease ,Metastatic breast cancer ,Primary tumor ,Breast cancer ,CpG site ,Genetic marker ,Internal medicine ,DNA methylation ,medicine - Abstract
Background- The ability to consistently detect cell-free tumor-specific DNA in peripheral blood of patients with metastatic breast cancer provides the opportunity to detect changes in tumor burden and to monitor response to treatment. Studies of cell-free DNA in the peripheral blood of breast cancer patients suggest that methylated DNA markers in serum or plasma could be used for detection of advanced disease, monitoring of therapeutic response, and for early detection of disease recurrence. Methods- A genome-wide serum DNA methylome array (Illumina HumanMethylation27 BeadChip) analysis was conducted on cell-free circulating DNA in serum from women with stage IV recurrent breast cancer, and 232 key CpG loci were identified. Methylation for this panel of 10 gene loci was evaluated using our newly developed cMethDNA assay to detect miniscule amounts of methylated DNA in Training and Test sets of sera from a total of 112 women (n = 55 normal, n = 57 metastatic breast cancer). The clinical sensitivity and specificity of the assay, along with technical reproducibility, was determined. To evaluate the concordance of DNA methylation patterns, the 10 gene panel was tested on 22 DNA sets of primary tumor, metastases and serum from the same patient. Finally, the ability of cMethDNA to monitor response to therapy was evaluated in 28 patients with metastatic disease. Results- A normal laboratory threshold of 7 cumulative methylation units was set and assay parameters were locked, based on Receiver Operating Characteristic (ROC) analyses of DNA from 300 ul of patient sera in the Training set (normal, n = 28; cancer, n = 24; 92% sensitivity, 96% specificity, and AUC = 0.950). Evaluation of the Test set of patient sera (normal, n = 27; cancer n = 33) resulted in detection of metastatic breast cancer with 91% sensitivity, 100% specificity, and AUC = 0.994 (0.984-1.005, p Conclusion- Together, our data suggest that the cMethDNA test 1) can detect tumor DNA shed into blood, 2) reflect the methylation alterations typical of the primary tumor and its metastatic lesions, and 3) reflect response to treatment after chemotherapy. Next, we will test the clinical utility of cMethDNA in independent clinical trial sample sets where it's complementary and independent roles will be examined against CA15.3 and CTC assays. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P2-06-01.
- Published
- 2013
- Full Text
- View/download PDF
37. Abstract P2-11-01: Effects of chemotherapy on the ovary: What you didn't know
- Author
-
Marlene H. Frost, Daniel Satele, Debra L. Barton, SL Thompson, and JN Senn-Reeves
- Subjects
Gynecology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,medicine.drug_class ,medicine.medical_treatment ,Oophorectomy ,Androgen ,medicine.disease ,Menopause ,Follicle-stimulating hormone ,chemistry.chemical_compound ,Dehydroepiandrosterone sulfate ,Sex hormone-binding globulin ,Oncology ,chemistry ,Estrogen ,Follicular phase ,medicine ,biology.protein ,business - Abstract
Background: It has long been known that chemotherapy can result in premature menopause, causing follicular senescence and estrogen depletion with associated hot flashes and mood alterations. What is less well appreciated is whether the stroma of the ovary is equally impacted after chemotherapy, resulting in androgen deprivation. The aim of this pilot study was to evaluate whether androgen levels are adversely affected after chemotherapy and whether this is associated with unwanted symptoms. Methods: Women who were premenopausal, newly diagnosed with breast cancer, and about to undergo adjuvant chemotherapy were followed longitudinally. Women with adrenal insufficiencies, taking steroids, oral contraceptives, or had had previous chemotherapy were excluded. Self report questionnaires regarding sexual function, fatigue, mood, menstrual symptoms; menstrual diaries; and blood were collected at 4 points: before treatment, mid chemotherapy, post chemotherapy and 6 months later. Serum concentrations of dehydroepiandrosterone sulfate (DHEA-S) (adrenal hormone), bioavailable testosterone (bioT), androstenedione (Adione) (stromal hormone), estrone (E1), estradiol (E2) (follicular hormone), sex hormone binding globulin (SHBG), and follicle stimulating hormone (FSH) were evaluated. Descriptive statistics, comparisons of means by two sided t-tests and Pearson correlation coefficients were computed. Six month post treatment data are reported. Results: 24 women were accrued and 21 provided serum and questionnaires through 6 months. All sex steroid hormones decreased during chemotherapy and did not return to baseline by 6 months for the group as a whole. At 6 months, 14 women were postmenopausal per FSH, E2 and menstrual diaries and 7 had resumed menses. There were no significant differences in hormone concentrations at baseline between women who ended up menopausal from those who resumed menses. However, at 6 months, postmenopausal women had significantly lower concentrations than premenopausal women of E2 (289 pg/ml pre- 9 pg/ml post), E1 (132 pg/ml pre- 22 pg/ml post), and Adione (102 ng/dL pre- 56 ng/dL post), but not DHEA-S or bioT (all p < .01). E2 was significantly correlated with Adione (R = .47 p = .03), and E1 (R = .57, p = .007). Low to moderate correlations were found between hormone concentrations and symptoms. The use of tamoxifen was significantly, and negatively correlated with the total score on the Female Sexual Function Index (r = −.572, p = .005), indicating worse sexual function for women on tamoxifen. Conclusion: These data support the hypothesis that the post chemotherapy ovary suffers both follicular and stromal dysfunction, as noted by lower Adione, which is specific to the ovarian stroma. Adione concentrations in the postmenopausal group women are similar to published reports of women post oophorectomy. This is the first longitudinal study we are aware of to evaluate ovarian stromal function in women undergoing chemotherapy. This total hormone depletion may be why women experiencing chemotherapy induced menopause report severe and distressing menopausal symptoms such as hot flashes. Estrogen is often implicated, but androgen deprivation in this population should be taken into consideration when planning interventions to improve health related quality of life. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr P2-11-01.
- Published
- 2012
- Full Text
- View/download PDF
38. Abstract P2-13-02: Effect of aspirin (ASP) or celecoxib (CC) use on outcomes in postmenopausal breast cancer patients randomized to adjuvant exemestane or anastrozole: NCIC CTG MA.27
- Author
-
C. Elliott, L. Han, M. J. Ellis, Lois E. Shepherd, Mark Clemons, Crnjevic T Badovinac, Karen A. Gelmon, Manuela Rabaglio, GW Sledge, JN Ingle, Kathleen I. Pritchard, George Thomas Budd, Paul E. Goss, J-Aw Chapman, and M. Higgins
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Randomization ,business.industry ,Anastrozole ,Cancer ,medicine.disease ,Placebo ,chemistry.chemical_compound ,Breast cancer ,Exemestane ,chemistry ,Internal medicine ,Concomitant ,medicine ,Clinical endpoint ,business ,medicine.drug - Abstract
Background: ASP is hypothesized to decrease the risk of breast cancer (BRCA) and BRCA recurrences. Thus we performed an exploratory analysis of ASP and CC use within the NCIC CTG MA.27 adjuvant trial comparing exemestane (E;N=3789) to anastrozole (A; N=3787) with a factorial second randomization to CC or placebo (P). Neither A or E was superior in breast cancer outcomes. Baseline low-dose ASP use is an accepted surrogate of cardiovascular risk factors and was used as a stratification factor across all 4 arms. Randomization to CC-P was discontinued after 18 months (n = 1622) due to concerns of cardiac toxicity. Methods: Patients taking >81mg of ASP daily at baseline were ineligible for randomization and use of >81mg of ASP daily was not allowed. Women enrolled during CC randomization were included in the comparison of E and A, stratified by whether they had been randomized to CC [yes, no;N=1622] and concomitant low-dose ASP [≤81 mg/day (yes, no); N=2209]. Other stratification factors included: lymph-nodes (negative, positive, or unknown); prior adjuvant chemotherapy (yes, no). The primary endpoint, event-free-survival (EFS), was defined as time from randomization to time of locoregional or distant disease recurrence, new primary breast cancer, or death from any cause. Secondary endpoints included overall survival (OS) defined as time from randomization to time of death from any cause and distant disease-free-survival (DDFS), defined as time from randomization to time of distant disease recurrence. Univariate (uni) assessment of CC and ASP use was assessed with stratified log-rank test, adjusting for lymph-node status and adjuvant chemotherapy and applied by intention-to-treat. Exploratory multivariate (multi) analyses (N = 1622) had forced inclusion of treatment and used step-wise forward stratified Cox modeling to examine the effects of CC, ASP use and baseline patient characteristics on outcomes; a factor was added with Wald test statistic p ≤ 0.05. Results: At median follow-up of 4.1 years, 186/1622 (11%) patients had an EFS event; 125 (8%) had died from any cause, and 80 (5%) had distant BRCA relapse. CC did not have significant uni association with outcomes: EFS p-value=0.92; OS p-value p = 0.56; DDFS p-value=0.55. ASP use was associated with worse EFS [p = 0.006, HR 1.48 (95% CI 1.12–1.96)], worse OS [p = 0.0002, HR 1.87 (95% CI (1.35–2.61)], and non-significant difference in DDFS (p = 0.72). CC had no multi association with EFS, OS or DDFS. ASP use had no multi association with EFS and DDFS (p > 0.05). However, ASP use had multi prognostic association with worse OS [p = 0.01; HR 1.67 (95% CI 1.13–2.49)]. Conclusions: Users of either CC or low dose ASP had similar DDFS to non-users in MA.27. As expected, low-dose ASP users (with presumptive cardiovascular risk factors) had worse OS than non-users. Inadequate numbers of patients randomized to CC and lack of randomization to aspirin leaves inadequate evidence from MA.27 to determine whether anti-inflammatories influence breast cancer outcomes. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr P2-13-02.
- Published
- 2012
- Full Text
- View/download PDF
39. P2-14-02: NCIC CTG MA.27: Clinical Tolerability and Overall Survival of Racial and Ethnic Minority Women on Aromatase Inhibitor Therapy
- Author
-
Beverly Moy, Paul E. Goss, C. Elliott, Maitre A Le, JN Ingle, Lois E. Shepherd, Karen A. Gelmon, and J-Aw Chapman
- Subjects
Gynecology ,Cancer Research ,medicine.medical_specialty ,Aromatase inhibitor ,business.industry ,medicine.drug_class ,Anastrozole ,medicine.disease ,chemistry.chemical_compound ,Exact test ,Breast cancer ,Oncology ,Exemestane ,chemistry ,Tolerability ,Internal medicine ,medicine ,Adjuvant therapy ,Pacific islanders ,business ,medicine.drug - Abstract
Background Aromatase inhibitors (AIs) are standard adjuvant therapy for postmenopausal women with hormone receptor-positive breast cancer. We previously reported that racial/ethnic minority women receiving an AI in NCIC CTG MA.17 experienced fewer hot flashes, fatigue, and arthralgias than Caucasians (Moy B et al. Ann Oncol 2006;17:1637). Here we examined whether race affected clinical outcomes in the MA.27 AI trial comparing exemestane (E) with anastrozole (A). Methods: Fisher's exact test was used to compare observed side effects (0 vs grades 1–5) between minority and Caucasian women. Adverse events (AE), including menopausal symptoms, were assessed according to the Common Terminology Criteria of the National Cancer Institute (version 3.0). ITT univariate test of race effects on overall survival (OS) was determined with a stratified log-rank test, while multivariate testing was with stratified Cox regression. Results: Among 7312 for whom race was known, distribution was: Caucasian (n=6939; 95%); black (n=235; 3%); Asian/Native Hawaiian/Pacific Islander (n=93); 1%; American Indian (n=39; 1%); and mixed race (n=6). Among women treated with E, minorities reported a significantly lower incidence of hot flashes (45% vs. 56%; p=0.003) and fatigue (34% vs. 46%; p=0.001) compared to Caucasians. Similarly with A, minority women reported significantly fewer hot flashes (47% vs. 58%; p=0.002) and lower cholesterol (12% vs. 18%; p=0.01); however, they reported more headaches than Caucasians (16% vs. 10%; p=0.01). Caucasian women were more likely to discontinue therapy due to AE or side effects (32% vs. 24%). There was a significant OS interaction (p=0.02) between race and treatment with minority women on E having fewer deaths than those on A. Caucasian women had HR of E to A of 0.98 (95% CI 0.81−1.20), p=0.85, while minorities had HR of E to A of 0.72 (95% CI 0.33−1.58), p=0.41. Conclusions: Minority women tolerated AIs better and were also more compliant than Caucasians, supporting our previous findings. A significant interaction between race and type of AI therapy for OS was seen, favoring exemestane in minority women. Replication of these findings in larger cohorts is necessary. Since race/ethnicity may serve as a surrogate for genetic differences, our findings suggest pharmacogenomic differences which require further research. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P2-14-02.
- Published
- 2011
- Full Text
- View/download PDF
40. PD01-06: Endoxifen Exhibits Potent Anti-Tumor Activity and Regulates Different Genes Than Tamoxifen in an Aromatase Expressing MCF7 Model Resistant to Letrozole
- Author
-
Kathryn E. Reinicke, JN Ingle, Angela Brodie, Ann L. Oberg, Matthew Bidwell Goetz, Paul Haluska, VJ Suman, Joel M. Reid, Matthew M. Ames, Mary J. Kuffel, D Grill, and Xiaonan Hou
- Subjects
Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,medicine.drug_class ,Letrozole ,Estrogen receptor ,Endocrinology ,Oncology ,Amphiregulin ,Estrogen ,In vivo ,Internal medicine ,Gene expression ,biology.protein ,Medicine ,Aromatase ,business ,Tamoxifen ,medicine.drug - Abstract
Background: First in human studies of Z-endoxifen hydrochloride (E), the active metabolite of tamoxifen (T), are underway in metastatic breast cancer (BC). Previous data have demonstrated the superiority of aromatase inhibitors (AI's) over T in estrogen receptor (ER) + BC. Using an in vivo aromatase expressing model (MCF7/AC1), we compared the antitumor activity of E with T and Letrozole (L), as well as the antitumor activity and global gene expression changes of E with T in an L-resistant model. Methods: MCF7/AC1 tumors were stimulated with androstenedione. Once tumor size reached 300 mm3, mice (30/group) were randomly assigned to one of five treatment groups: control (daily, po), T (500 μg/day, sc), endoxifen 25 mg/kg/day p.o.(LDE) endoxifen 75 mg/kg/day p.o. (HDE) or letrozole, 10 μg/day s.c for 4 weeks. Tumors were harvested from control, T, and E groups while the L group continued treatment until the development of resistance defined as an increase in tumor volume of at least 300% from day 1. Mice with L-resistant tumors were randomly assigned to T (n=4) or E (n=5) for 4 weeks and then sacrificed. Gene expression in L-resistant tumors was quantified using Affymetrix U133+2 and changes in gene expression profiles [comparing T and E with L-resistant (n=3)] were analyzed. Genes identified as significantly different were confirmed by real-time RT-PCR assays. Results: At the 4 week time point, both doses of E and L resulted in greater anti-tumor activity than control (Wilcoxon rank sum test: all p < 0.0001); however, tumor burden did not differ between T and control (p=0.095). HDE resulted in significantly less tumor burden than T (p=0.002) but was similar to L. In mice that continued on L, resistance developed at 24 weeks in 9/25 mice. These mice were randomly assigned to either T (n=4) or E (n=5) for 4 weeks. Tumor volume (expressed as a% of its size prior to randomization) was significantly different comparing E (73.3%; range: 69.3 to 80.75%) versus T (148.39%; range: 114.07 to 165.99%) (Wilcoxon rank sum test p=0.016). Compared to control, microarray studies identified 1518 unique probe sets regulated by E (p Conclusions: Using the MCF7/AC1 model previously used to show the superiority of AI's over T, HDE demonstrated similar antitumor activity to L and was superior to T. In cells resistant to L, E was superior to T and gene expression changes demonstrate that E down-regulates while T activates estrogen regulated genes. These findings support the ongoing development of E for the treatment of ER+ BC. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr PD01-06.
- Published
- 2011
- Full Text
- View/download PDF
41. P3-16-09: Endoxifen, a Newly Developed Breast Cancer Drug, Has Anabolic Actions on the Mouse Skeleton
- Author
-
JN Ingle, Muzaffer Cicek, K.D. Peters, Matthew P. Goetz, Sarah B. Grygo, Kevin S. Pitel, Malayannan Subramaniam, Thomas C. Spelsberg, John R. Hawse, Xianglin Wu, Urszula T. Iwaniec, RT Turner, and GL Evans
- Subjects
Bone mineral ,Cancer Research ,medicine.medical_specialty ,Bone density ,medicine.diagnostic_test ,biology ,business.industry ,Osteoblast ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Osteoclast ,Internal medicine ,Ovariectomized rat ,Osteocalcin ,biology.protein ,Medicine ,Quantitative computed tomography ,business ,Tamoxifen ,medicine.drug - Abstract
Background Commonly used endocrine therapies for breast cancer, such as aromatase inhibitors in postmenopausal women and tamoxifen in premenopausal women, have deleterious effects on bone mineral density. Therefore, the identification of novel cancer therapies which either maintain or improve bone mass are of clinical need. We have recently demonstrated that endoxifen is the most active tamoxifen metabolite with regard to inhibiting the growth of ERα+ breast cancer cells and these studies have led to the development of endoxifen as a novel anti-breast cancer drug for which first-in-human studies are now underway. At present, there are no data regarding endoxifen's effects on bone. Methods: The effects of endoxifen on osteoblast (OB) and osteoclast (OC) maturation and gene expression were monitored by cell differentiation assays and real-time PCR. Dual-energy X-ray absorptiometry (DXA), peripheral Quantitative Computed Tomography (pQCT) and micro-Computed Tomography (μCT) were used to determine changes in bone density, mass and architecture following 45 days of oral endoxifen administration (50mg/kg/day) to 3-month-old ovariectomized (OVX) C57BL/6 mice relative to vehicle control treated animals. Alterations in the numbers and activity of OBs and OCs were determined by histomorphometry and serum levels of P1NP and CTX-1 respectively. Results: Endoxifen treatment of mouse derived bone marrow stromal cells and human OBs led to significant increases in the expression of critical bone marker genes such as Runx2, osterix, osteocalcin, osteoprotegerin and alkaline phosphatase in a dose dependent manner. Daily administration of endoxifen to OVX mice led to significant increases in total body bone mineral density (BMD) (6%) and content (BMC) (9%), which was accompanied by a 50% decrease in fat tissue mass as determined by DXA. pQCT analysis of the tibial metaphysis revealed dramatic increases in BMD (35%) and BMC (20%), as well as trabecular density (52%), cortical content (62%), cortical area (60%) and cortical thickness (78%). μCT analysis of the femoral metaphysis revealed increases in bone volume/total volume (200%), trabecular number (38%) and trabecular thickness (18%), as well as decreased trabecular spacing (29%). Interestingly, there was nearly a 50% increase in the numbers of OCs derived from endoxifen treated mice which was associated with elevated expression of OC marker genes such as NFATcl, RANK, c-fms and cathepsin-K compared to control treated animals. Approximately 4 times as many OBs and OCs were observed on the bone surfaces of endoxifen treated mice which correlated with nearly 2-fold increases in serum levels of the bone formation (P1NP) and resorption (CTX-1) markers. Conclusions: These data are the first to demonstrate that endoxifen has anabolic effects on the mouse skeleton which are similar to that of estrogen. Additionally, these data reveal that endoxifen's mechanism of action in bone is different than that reported for tamoxifen and other selective estrogen receptor modulators in mice as it increases, rather than decreases, bone formation and remodeling. Therefore, the use of endoxifen for the treatment of endocrine responsive breast cancer may avoid the detrimental skeletal effects of many conventional endocrine therapies. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P3-16-09.
- Published
- 2011
- Full Text
- View/download PDF
42. P4-11-13: Influence of Two Years of Exemestane on Bone Mineral Density in Postmenopausal Women at Increased Risk of Developing Breast Cancer; a Companion Study to the NCIC CTG MAP.3 Trial
- Author
-
A Hiltz, Angela M. Cheung, Judy Garber, JN Ingle, D. Tu, Carol J. Fabian, Gloria E. Sarto, Rowan T. Chlebowski, Harriet Richardson, and Paul E. Goss
- Subjects
musculoskeletal diseases ,Bone mineral ,Gynecology ,Cancer Research ,medicine.medical_specialty ,Randomization ,business.industry ,Osteoporosis ,medicine.disease ,Confidence interval ,chemistry.chemical_compound ,Breast cancer ,Oncology ,Exemestane ,chemistry ,Internal medicine ,Medicine ,Population study ,Young adult ,business - Abstract
Background Exemestane significantly reduced invasive and preinvasive breast cancers in postmenopausal women at increased risk for breast cancer in the NCIC CTG MAP3 trial with no serious toxicities, including excess fractures or osteoporosis. Purpose: To provide additional information on the effect of exemestane on bone loss in women at high risk for breast cancer, within a subset of women participating on the NCIC CTG MAP.3B study. The primary hypothesis is that exemestane does not induce clinically significant bone loss in postmenopausal women at increased risk of developing breast cancer at 2 years. The primary objective of this companion study is to examine the effect of exemestane on lumbar spine and total hip BMD by DEXA at 2 years in women participating in the MAP3 trial. Methods: The MAP.3B bone sub-study registered women from the main MAP. 3 trial from May 2008 to March 2010. Eligible women had to have an acceptable quality BMD scan by DEXA taken within 12 months prior to randomization to MAP.3. A BMD T-score >-2.0 SD (i.e. better than 2 standard deviations below the average peak BMD of a young adult woman) was established as the study population cutoff. A questionnaire including information on height, falls, fractures, lifestyle information including physical activity, tobacco and alcohol use was completed at baseline, 12 months, 24 months and at last visit. Fasting serum for bone biomarkers was collected at 12 months and total hip and L1-L4 (postero-anterior) spine BMD were measured 2 years after randomization on the same Lunar or Hologic scanner. The primary objective was to determine differences in hip and spine BMD at 2 years. Secondary outcomes include number of skeletal fractures and development of osteoporosis 2 years after randomization and changes in bone biomarkers at 1 year after randomization. For the analysis of the primary endpoints, the upper limit of a one sided 95% confidence interval for the difference in mean percentage changes between placebo and exemestane will be calculated for the BMD by DEXA at each site. We will conclude that exemestane does not induce significant bone loss in postmenopausal women at increased risk of developing breast cancer at 2 years when the upper limit is less than 3% for both sites. Similar confidence interval approach will be used to analyze the secondary endpoints. Results: Between May 2008 and March 2010, 238 postmenopausal women were recruited. Median age was 61.8 years, and the majority of women were Caucasian (91%), with approximately 20% of the participants reporting a recent fall (within past 12 months) and another 13% reporting a recent fracture prior to randomization. We will report results from the primary as well as the secondary endpoints at the SABCS meeting. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P4-11-13.
- Published
- 2011
- Full Text
- View/download PDF
43. P5-14-01: Differences in Efficacy by Assessment Method: NCIC CTG Adjuvant Breast Cancer Trials MA.5, MA.12, MA.14, MA.21, MA.27 Meta-Analysis
- Author
-
Karen A. Gelmon, Vivien H.C. Bramwell, Mark Levine, Timothy J. Whelan, B. Dong, Wendy R. Parulekar, Lois E. Shepherd, Rinat Yerushalmi, J-Aw Chapman, Paul E. Goss, Michael Pollak, Margot J. Burnell, JN Ingle, and Kathleen I. Pritchard
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Randomization ,business.industry ,Proportional hazards model ,Cancer ,ECOG Performance Status ,medicine.disease ,Surgery ,Breast cancer ,Meta-analysis ,Internal medicine ,medicine ,Clinical endpoint ,business ,Survival analysis - Abstract
Background: Based on recent breast cancer literature, we hypothesized that there could be substantive differences in apparent efficacy estimates using a log-normal (LN) survival model rather than with standard Kaplan-Meier (K-M) or Cox model methods. While both Cox and LN survival analyses offer greater specification by individual patient characteristics, the LN model may more robustly estimate survival under model misspecification. Methods: We recently pooled data for 5 NCIC CTG primary breast cancer trials: MA.5, MA.12, MA.14, MA.21, and MA.27. The total patient count for patients who received at least 1 dose of trial therapy is 11,253. Compilation included definition of STEEP endpoints (C Hudis, JCO, 2008) and standardized factor categorizations. The primary endpoint is Breast Cancer Free Interval (BCFI) defined as the time from randomization until recurrence: first local invasive or DCIS; regional, or distant; contralateral invasive or DCIS; or death from breast cancer. We found substantive evidence of non-proportionality for 7 factors compiled for the meta-analyses. In this work, we fit multivariate Cox and LN models with these 7 factors, lymph node status and pathologic T status. We then compare BCFI efficacy estimates for patient and tumour characteristics at 1-, 3-, and 5-years obtained with K-M, Cox, and LN models. Results: There was evidence that the Cox assumption of proportional hazards was violated for 7 factors: age, menopausal status, hormone receptor status, anthracycline use, chemotherapy use, race, and ECOG performance status. Differences between models were intrinsically affected by timing and extent of non-proportionality; there was no consistent pattern. In particular, investigations to date indicate efficacy estimates with absolute differences between K-M, Cox and LN estimates which varied by time of assessment: at 1-year 0.0 to 6.7%, at 3-years 0.4 to 18.6%, and at 5-years 0.2 to 17.0%. BCFI estimates with the K-M were inconsistently closer to those with the LN or Cox model: for K-M to Cox at 1-year 0.4 to 5.2%, at 3-years 0.4 to 15%, at 5-years 0.4 to 14.3%; for K-M to LN at 1-year 0.0 to 6.7%, at 3-years 0.5 to 18.6%, at 5-years 0.2 to 17.0%; for Cox to LN at 1-year 0.8 to 1.8%, at 3-years 1.9 to 6.0%, at 5-years 0.6 to 5.7%. K-M and Cox models have step-wise adjustments at events for K-M and Cox, rather than smooth modeling with the LN. Discussion: Even with reasonably large population subgroups, there were substantive differences in apparent survival (0.0 to 18.6%) between K-M, Cox and LN model types. The magnitude of differences in survival estimates was large enough to be clinically relevant and warrant further consideration as we evaluate new therapies and prognostic/predictive factors. We will be statistically investigating framework robustness under differing levels of model misspecification. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P5-14-01.
- Published
- 2011
- Full Text
- View/download PDF
44. P2-10-05: Cryo-Microcomputed Tomography (CT) with Synchrotron Radiation: Visualization of Human Breast Cancer Tissue and Comparison with Their Histopathologic Findings
- Author
-
S-H Park, H-T Kim, Y-J Jeong, J-Y Huang, H-Y Jeong, JN Kim, and J-G Bong
- Subjects
Cancer Research ,medicine.medical_specialty ,Materials science ,Breast imaging ,Synchrotron radiation ,Visualization ,law.invention ,Oncology ,Beamline ,Optical microscope ,law ,medicine ,Radiology ,Tomography ,Image resolution ,Surface reconstruction ,Biomedical engineering - Abstract
Background: A synchrotron radiation (SR)-based X-ray source offers a powerful tool for diagnosis of breast disease due to the energy spectrum properties and the peculiar laminar beam geometry. The aims of this study were to estimate the visualization of the human breast cancer tissue with SR cryo-micro CT and to compare the results with histopathological examinations. Material and Methods: The cancerous breast tissue samples were routinely fixed in 10% neutral buffered formalin, and each specimen was cut down to a cylindrical sample with 2 mm diameter and 10 mm height. Each breast cancer sample was rapidly frozen with dry ice, mounted on a computer-controlled precision stage and maintained at cryogenic temperature throughout data collection. Experiments were performed at the bending magnet beamline 7B2 of Pohang Light Source (PLS) in Accelerator Laboratory (PAL) which is a third-generation SR facility with 2.5 GeV operating energy. The white beam imaging system developed for synchrotron tomography consists of a 1 mm Si attenuator, 100 μm-thick CdWO4 scintillator and a full-frame charge-coulped device camera. The detector is placed 10 mm downstream from the sample on an optical table which can be rotated in the fan beam about a vertical axis for tomography. For tomography, images were collected at 0.18° increments through 180°. The visual image was magnified using a 20x microscope objective and captured using a digital CCD camera. The spatial resolution determined by standard sample was about 1.5 μm. Three-dimensional volume images of the specimen were obtained by applying a filtered back-projection algorithm to the projection images using a software package OCTOPUS. Surface reconstruction and volume segmentation and rendering were performed using Amira software. After imaging the samples were split into several sections, processed and embedded in paraffin. Obtained tomography images were compared with corresponding histopathological findings in optical microscopy. Results: A total of 1000 synchrotron tomography images were acquired from the samples and only a small number of different typical cases were selected for a detailed analysis. The correspondence between tomography images and histopathological findings were determined. Synchrotron tomography images yield high contrast from smoothly varying internal structures corresponding to information on actual structures seen at histopathological analysis. Discussion: Since the projection of 3D anatomic information onto 2D image will complicate subtle difference in X-ray transmission and objects at different depths will be superimposed on each other, the subtle difference in subject attenuation, diffraction and contrast may not be clearly visible or completely lost. Tomography with SR has superior visualization of subject contrast, together with depth localization, so it is useful for visualizing anatomical structures in the sample. In this study, the obtained SR cryo-micro CT images of human breast cancer tissue were comparable with standard histopathologic findings. The results suggest that tomography with SR has a great potential as a diagnostic tool and also its clinical application is feasible, especially in breast imaging. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P2-10-05.
- Published
- 2011
- Full Text
- View/download PDF
45. Abstract P2-09-03: Mammographic Density Response to Aromatase Inhibitor Therapy
- Author
-
Kathy R. Brandt, Aman U. Buzdar, Richard M. Weinshilboum, Celine M. Vachon, Janet E. Olson, Paul E. Goss, Daniel J. Serie, Fang Fang Wu, VJ Suman, Lois E. Shepherd, JN Ingle, and Matthew L. Kosel
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Aromatase inhibitor ,medicine.diagnostic_test ,business.industry ,medicine.drug_class ,Anastrozole ,Cancer ,medicine.disease ,chemistry.chemical_compound ,Breast cancer ,Exemestane ,chemistry ,Internal medicine ,Adjuvant therapy ,Medicine ,Mammography ,business ,Tamoxifen ,medicine.drug - Abstract
Background: Mammographic density, the variation in fat, epithelial and stromal tissues seen on screening mammography, is a strong risk factor for breast cancer and can be modified by hormonal agents. Changes in density from tamoxifen or postmenopausal hormone (PMH) use are associated with risk, suggesting that density may be a surrogate marker of therapeutic efficacy. Aromatase inhibitors (AIs) are given as adjuvant therapy in hormone receptor positive postmenopausal breast cancer and are known to decrease levels of estrone and estradiol in both serum and breast tissue. Our goal here was to examine the influence of AIs on mammographic density in women with early breast cancer. Methods: We conducted a case-control study of postmenopausal breast cancer patients initiating adjuvant AI therapy (anastrozole or exemestane) on protocols NCIC CTG MA27, NCCTG N063I and MC (Mayo Clinic) 0532. Eligibility included; an intact contralateral breast with no prior surgery; a screening mammogram within twelve months before AI initiation and at 9-15 months on therapy; no prior endocrine therapy and informed consent. Controls were sampled from the Mayo Mammography Health Study, a cohort of 19,924 receiving screening mammography at the Mayo Clinic, and matched to cases on age, prior PMH use, baseline body mass index (BMI) and interval between mammograms. Pre-treatment and on-study mammograms for cases (corresponding mammograms for controls) were digitized. Change in percent density was estimated on the craniocaudal view of the non-cancerous breast using two methods: a subjective assessment of change by an expert radiologist (within 5%; 5-10% increase, 10-25% increase, 25%+ increase, 5-10% decrease, 10-25% decrease and 25%+ decrease) and a quantitative assessment of absolute change using a computer-assisted thresholding program (Cumulus). Analyses compared magnitude of change in density by both the subjective and quantitative methods between cases and matched controls. Results: 574 pairs were eligible for analyses (MA27-505 cases; N063I-12 cases; MC0532-57 cases). Characteristics of the two groups are shown in the table below. Using either density estimation method, there was a greater decrease in density among women on AI therapy vs. matched controls. In 33% (95% CI: 29-37%) of pairs, there was at least a one greater category decrease for the case relative to her control by subjective estimation. In 14% (95% CI: 11-18%) of the pairs, there was at least a 5% greater decrease for the case relative to her control by quantitative estimation. Data will be available according to AI class (non-steroidal versus steroidal) in November. Conclusions: In the largest report to date to examine the influence of AI therapy on mammographic density, we provide evidence that AI is associated with decreases in density in a small subgroup of women. We are currently examining factors that influence these AI-associated decreases in density and whether these differences are unique to one class of AI. (Supported in part by NIH grants P50CA116201, U01GM61388, U10CA77202, U10CA25224) Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P2-09-03.
- Published
- 2010
- Full Text
- View/download PDF
46. Abstract PD05-11: Development, Characterization, and Effective In Vitro Treatment of an Endoxifen Resistant Breast Cancer Cell Line
- Author
-
C Muzaffer, Thomas C. Spelsberg, Matthew P. Goetz, John R. Hawse, Vivian Negron, Malayannan Subramaniam, JN Ingle, Xianglin Wu, and Wilma L. Lingle
- Subjects
Cancer Research ,medicine.medical_specialty ,Programmed cell death ,Cell growth ,medicine.drug_class ,CD24 ,Biology ,Endocrinology ,Oncology ,Apoptosis ,Estrogen ,Internal medicine ,medicine ,Cancer research ,Gene silencing ,Epithelial–mesenchymal transition ,Tamoxifen ,medicine.drug - Abstract
Background: A major issue surrounding the use of therapeutic drugs for the treatment of breast cancer is the eventual development of resistance. Endoxifen, the most potent tamoxifen metabolite, is being developed as a novel endocrine therapy for the treatment of endocrine responsive breast cancer patients. While numerous studies have investigated the process of tamoxifen resistance, no such data exist regarding the mechanism by which cells develop resistance to endoxifen. Here, we describe the development and characterization of a novel endoxifen resistant MCF7 breast cancer cell line and the identification of a specific treatment to effectively target these resistant cells. Methods: MCF7 cells were chronically exposed to concentrations of endoxifen previously demonstrated to be associated with the greatest reductions in estrogen stimulated proliferation and transcription (1000 nM). Changes in the physiological and molecular properties of these cells were monitored during the course of resistance using a wide range of techniques. Results: Following 15 months of endoxifen exposure, an epithelial to mesenchymal transition (EMT) was induced in MCF7 cells, characterized by loss of E-cadherin expression, up-regulation of fibronectin and vimentin expression and increased responsiveness to TGFβ. Resistant cells exhibit a 7-fold increase in their proliferation rates relative to parental cells and display basal like properties (triple negative) due to silencing of ERα , progesterone receptor and CD24 expression. Resistant cells were confirmed to be estrogen insensitive through the use of cell proliferation and gene expression studies. Wound healing and cell migration assays revealed that resistant cells are highly aggressive with a significant level of metastatic potential. Microarray analysis revealed over 7500 genes to be differentially expressed in the resistant cell line relative to parental cells using a 2-fold cutoff, of which only 52 genes (0.7%) were determined to be up-regulated. Interestingly, a number of the most highly altered genes have previously been implicated in the development of EMT and/or resistance including Twist2, IGF binding protein 5, GATA and β-tubulin. Based on the identification of P-tubulin as the most up-regulated gene in resistant cells, 2-methoxyestradiol (2ME2) was recognized as a candidate drug to specifically target this resistant cell line due to it known roles in blocking tubulin polymerization. Indeed, 2ME2 exposure resulted in the induction of apoptosis and significant cell death in vitro. Conclusions: Chronic exposure of MCF7 cells to high concentrations of endoxifen led to induction of EMT with cells that are basal like, estrogen and SERM insensitive, highly TGFP responsive with significantly increased proliferation rates and metastatic potential relative to parental cells. These findings suggest that the mechanisms of resistance to endoxifen may differ from those observed with long term exposure to tamoxifen and have identified 2ME2 as a potentially successful alternative therapy for endoxifen resistant breast cancer cells. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr PD05-11.
- Published
- 2010
- Full Text
- View/download PDF
47. Abstract P3-12-11: Combining Genotype at Low Penetrance Breast Cancer Loci with Family History Risk Leads to Significant Risk Reclassification
- Author
-
AJ Martin, Lee Baker, A Onen, P. Pharoah, J Gale, Jn. Berg, Alastair M. Thompson, S White, and Jacqueline Dunlop
- Subjects
Oncology ,Cancer Research ,education.field_of_study ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Population ,medicine.disease ,Bioinformatics ,Penetrance ,Breast cancer screening ,Breast cancer ,Internal medicine ,Relative risk ,medicine ,Population Risk ,Family history ,education ,business ,Genetic testing - Abstract
Background: A significant proportion of women are at increased genetic risk of breast cancer. Mutations in high penetrance genes such as BRCA1 and BRCA2 are only responsible in the minority of cases, with low penetrance polymorphisms in other genes expected to account for the majority of the remaining genetic risk. An increasing number of such low penetrance polymorphisms are being identified, but each polymorphism only contributes a small amount to overall risk. Currently, in clinical practice, women who are at increased risk of breast cancer are identified by their family history, and the role of genetic testing for multifactorial risk remains uncertain. We have taken the population frequency and genotype relative risk information for the 18 most established low penetrance breast cancer risk loci and explored the effect of combining information from these loci, with risk derived from family history. Results: Genotyping at these 18 loci could provide significant risk information for an individual. The top 1% of women in the genotype risk distribution would have a risk of breast cancer of 2.11 times the general population. At this level of risk, they would qualify for breast cancer screening from age 40 according to evidence based guidelines issued by the UK National Institute of Clinical Excellence (NICE). In addition, the top 5% of the population are at 1.67 times risk of breast cancer and would have the same risk at age 40 as a 50 year old at population risk who would qualify for breast screening according to UK and US National Screening Guidelines. To investigate whether low penetrance genotype has greater potential if combined with other risk factors, we used a simple multiplicative model to combine family history risk of cancer derived using BOADICEA with genotype information. Our data suggest that genotype would result in a significant reclassification of individual risk. For example, 10% of women who only had a sister affected with breast cancer at 55 would qualify for additional screening under NICE criteria if genotype were taken into account. Extending this approach with 160 complex family histories from the Tayside family history breast clinic, we have shown that genotyping could result in reclassification and change of management for 19.1% of women being assessed in this clinic, with 12.4% of women moving into a higher risk category, and 6.7% of women moving into a lower category. Discussion: These data suggest that genotyping for low penetrance breast cancer risk loci is clinically relevant, and that it will be more powerful if it can be combined with other established risk factors. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P3-12-11.
- Published
- 2010
- Full Text
- View/download PDF
48. Abstract P3-10-26: Quantitative Protein and Gene Expression Biomarkers of Tamoxifen and Letrozole Recurrence in the NCIC CTG MA.17 Cohort
- Author
-
JN Ingle, Lois E. Shepherd, David L. Rimm, Soonmyung Paik, M Pins, Dennis C. Sgroi, Dongsheng Tu, Dianne M. Finkelstein, Hironobu Sasano, A Ristimaki, Peggy L. Porter, Kathleen I. Pritchard, and Paul E. Goss
- Subjects
Gynecology ,Oncology ,Cancer Research ,medicine.medical_specialty ,Tissue microarray ,biology ,business.industry ,Letrozole ,Cancer ,medicine.disease ,Clinical trial ,Breast cancer ,Internal medicine ,biology.protein ,medicine ,Aromatase ,Stage (cooking) ,business ,Tamoxifen ,medicine.drug - Abstract
Background: The MA.17 study showed that extended adjuvant endocrine therapy with letrozole (LET) after completing 5 years of tamoxifen (TAM) markedly reduced the risk of recurrence in women with ER+ early stage breast cancer and improved overall survival in women presenting with node +ve disease. The HOXB 13:IL17BR gene expression ratio (signature) has been shown to predict outcome in breast cancer patients treated with adjuvant tamoxifen monotherapy and provides additional information beyond that from known positive (ER and PR) and negative (Her-1 and Her-2) predictors of responsiveness to tamoxifen in node-ve women. We report a case control evaluation of the Breast Cancer Index (BCI; bioTheranostics, Inc.), which combines the HOXB13 and IL17BR twogene and the molecular grade index (MGI) gene expression signatures, with respect to distinguishing which patients are at risk of late recurrences and who would respond to extended endocrine therapy with LET. The prognostic and predictive utility of quantitative immunofluorescence of ER, PR, Her-2, tumor aromatase, COX-2, GATA3 and Nat1 in the TAM-PLACEBO and the TAM-LET cohorts will also be evaluated and compared to results derived by standard immunohistochemistry. Methods: FFPE tumor blocks were collected from patients who experienced a breast cancer recurrence up to unblinding of MA.17. Controls were matched 2:1 for age, tumor size, lymph node status, and prior chemotherapy, and were all disease free for longer than cases. All cases were reviewed for standard histopathology by two independent pathologists. RNA was extracted, amplified, converted to cDNA and subjected to RT-PCR with primers and probes to HOXB13, IL17BR, BUB1A, CENPA, NEK2, RACGAP1 and RRM2. ER, PR HER1, HER2, COX2, Aromatase, GATA3 and NAT1 will be analyzed by routine IHC techniques and by immunoflourescent Automated Quantitative Analysis (AQuA). Results: 105 cases and 210 matched controls are available for evaluation. All sections are under review and tissue microarrays have been performed on all cases and controls. Detailed results on the BCI and ER, PR, Her-2 will be available at the SABCS. Discussion: MA.17 has shown that extended adjuvant endocrine therapy after tamoxifen is effective at preventing disease recurrence given for an additional 5 years. Numerous clinical trials are exploring whether extending AIs will show this benefit, and there is an increasing need to improve the therapeutic index by distinguishing those at risk from those who are not. It is also important to determine which patients will benefit from the therapy and which will recur without benefit. The latter patients could be triaged to clinical trials of novel therapies to overcome endocrine resistance. This study will help to define these issues and pave the way for more effective selection of specific patients for adjuvant endocrine strategies. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P3-10-26.
- Published
- 2010
- Full Text
- View/download PDF
49. Abstract P2-09-25: ERα Expression in Breast Cancer: A Conundrum of Antibody Specificity?
- Author
-
Thomas C. Spelsberg, JN Ingle, Jr. Hawse, Malayannan Subramaniam, Vivian Negron, Matthew P. Goetz, Wilma L. Lingle, and Xianglin Wu
- Subjects
Cancer Research ,biology ,medicine.diagnostic_test ,medicine.drug_class ,Cancer ,Immunofluorescence ,Monoclonal antibody ,medicine.disease ,Molecular biology ,Epitope ,Breast cancer ,Oncology ,biology.protein ,medicine ,Immunohistochemistry ,Antibody ,Estrogen receptor alpha - Abstract
Background: The role of estrogen receptor alpha (ERα) in breast cancer has been studied extensively, and its protein expression is prognostic and a primary determinant of endocrine sensitivity; however, much less is known about the role of ERß. In vitro studies demonstrate a tumor suppressive function for ERß, and we have recently implicated a role for ERα in sensitizing ERα expressing breast cancer cells to the anti-estrogenic effects of endoxifen. However, the in vivo relevance of ERα remains unclear due to conflicting reports. Here, we provide evidence that some of this controversy may be explained by variability in antibody specificity. In addition, we describe the development and characterization of a novel, highly specific monoclonal antibody and provide data regarding ERα expression in human breast cancers. Methods: Five commercially available ERα antibodies were screened for their sensitivity and specificity using western blotting, immunoprecipitation, immunofluorescence and immunohistochemistry in known ERα negative and positive cell lines as well as in normal human tissue samples. A novel monoclonal ERα antibody (C10) was developed and characterized in the same manner. Following identification of two specific antibodies, ERα expression was assessed in 66 breast tumors collected prior to adjuvant therapy. Samples were scored separately for nuclear and cytoplasmic staining. Results: In depth analysis of commercially available ERα antibodies reveled that the majority were non-specific with substantial cross-reactivity to ERα . Only one commercial antibody (PPG5/10), which solely recognizes full-length ERß, and our newly developed monoclonal antibody, which recognizes full-length and all 4 ERα variants, were determined to be sensitive and specific for ERα expression. These same two antibodies resulted in strong staining for endogenous levels of ERα protein in normal prostate tissue by immunohistochemistry. We further assessed these two antibodies in a set of breast tumors. Preliminary analysis revealed significant differences for ERα positivity between these two antibodies. Based on nuclear staining, 92% of tumors were ERα positive using the PPG5/10 antibody while only 34% were positive with C10. Approximately 50% of all tumors exhibited cytoplasmic staining with both antibodies. Conclusions: Our studies demonstrate that the majority of commercially available ERα antibodies are either non-specific or insensitive for the detection of ERα via immunohistochemistry. The present data call into question the relevance of prior studies which tested the association between clinical outcome and ERα expression and demonstrate the need to further analyze the role of ERα in breast cancer using highly specific and validated antibodies. While both the PPG5/10 and C10 antibodies are highly specific for ERß, the significant discrepancy in nuclear staining between them in breast tumors may be due to changes in epitope availability as a result of post-translational processing. Our newly developed C10 antibody could provide additional discriminatory features which may be useful in predicting response to therapy and/or associations with other clinicopathological factors and such studies are currently underway. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P2-09-25.
- Published
- 2010
- Full Text
- View/download PDF
50. Abstract GS4-03: Randomized comparison of adjuvant tamoxifen (T) plus ovarian function suppression (OFS) versus tamoxifen in premenopausal women with hormone receptor-positive (HR+) early breast cancer (BC): Update of the SOFT trial
- Author
-
G Fleming, PA Francis, I Láng, EM Ciruelos, M Bellet, HR Bonnefoi, MA Climent, L Pavesi, HJ Burstein, S Martino, NE Davidson, CE Geyer, BA Walley, RE Coleman, P Kerbrat, S Buchholz, JN Ingle, M Rabaglio-Poretti, M Colleoni, and MM Regan
- Subjects
Cancer Research ,Oncology - Abstract
Background: The primary results of SOFT at 5.6 years median follow-up found adding OFS to T did not provide a significant benefit in the overall study population of premenopausal women with HR+ BC (Francis et al, NEJM 2015). For those women at sufficient risk for recurrence to warrant adjuvant chemotherapy (CT) and who remained premenopausal, the addition of OFS improved disease outcomes. Follow-up was immature for overall survival (OS). We report a planned update with visit cut-off of 31Dec16 after 8 yrs median follow-up. Methods: SOFT randomized premenopausal women with HR+ BC from Nov 2003 to Jan 2011 to 5 yrs of T vs T+OFS vs Exemestane(E)+OFS. OFS was by choice of GnRH agonist triptorelin, oophorectomy or ovarian irradiation. SOFT was stratified by the use of prior CT; 47% received no CT and 53% remained premenopausal after prior CT, determined by premenopausal estradiol level within 8 months of CT completion. The primary endpoint was invasive disease-free survival (DFS; randomization until invasive local, regional, distant recurrence or contralateral breast; invasive second malignancy; death). Secondary endpoints included invasive breast cancer-free interval (BCFI), distant recurrence-free interval (DRFI) and OS. NCT00066690. Results: DFS for patients assigned T+OFS (n=1015) was significantly improved over T (n=1018; HR=0.76 [95%CI 0.62-0.93]) and 8yr DFS was 83.2% vs 78.9%, respectively; BCFI and DRFI results were supportive (see Table). Hazard ratios for these 3 endpoints showed no heterogeneity by use of prior CT. For patients with prior CT, 8yr DFS was 76.7% with T+OFS vs 71.4% with T (Δ=5.3%); in those without CT, 8yr DFS was 90.6% vs 87.4% (Δ=3.2%). E+OFS (n=1014) improved outcomes relative to T (Table); 8yr DFS for E+OFS was 85.9% (80.4% with use of prior CT and 92.5% for those without CT). OS was improved with T+OFS vs T (8yr OS 93.3% vs 91.5%). 8yr OS was 92.1% with E+OFS. 201/225 deaths occurred in women with prior CT. For women without CT there have been 10, 5 and 9 deaths in the T+OFS, T and E+OFS groups (total n=1419), respectively, only half of these deaths after breast cancer event. N. EventsHazard Ratio (95% CI)Endpoint(3 arms)T+OFS vs TE+OFS vs TDFS5180.76 (0.62-0.93) P=0.0090.65 (0.53-0.81)BCFI4370.76 (0.61-0.95)0.64 (0.51-0.81)DRFI3060.86 (0.66-1.13)0.73 (0.55-0.96)OS2250.67 (0.48-0.92)0.85 (0.62-1.15) Overall toxicity was worse with T+ OFS than with T, including 32% vs 25% grade 3+ targeted AEs. Early cessation of tamoxifen occurred for 19% assigned T+OFS and 22% of women assigned T; the cumulative incidence of early cessation of triptorelin on the T+OFS arm was 23% by 4yrs. Early cessation of exemestane occurred for 28% and of triptorelin for 21% by 4yrs on the E+OFS arm. Conclusions: With additional follow-up to a median of 8yrs, SOFT further supports the value of OFS for some premenopausal women. Follow-up continues, which will further clarify the safety and the benefit of OFS for late recurrence and overall survival. Oncologists appear to be able to select a low risk group (no chemotherapy) for whom treatment escalation is unlikely to improve survival. Citation Format: Fleming G, Francis PA, Láng I, Ciruelos EM, Bellet M, Bonnefoi HR, Climent MA, Pavesi L, Burstein HJ, Martino S, Davidson NE, Geyer Jr CE, Walley BA, Coleman RE, Kerbrat P, Buchholz S, Ingle JN, Rabaglio-Poretti M, Colleoni M, Regan MM. Randomized comparison of adjuvant tamoxifen (T) plus ovarian function suppression (OFS) versus tamoxifen in premenopausal women with hormone receptor-positive (HR+) early breast cancer (BC): Update of the SOFT trial [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr GS4-03.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.