Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lillian Mary Smyth is active.

Publication


Featured researches published by Lillian Mary Smyth.


The Journal of Pathology | 2016

Massively parallel sequencing of phyllodes tumours of the breast reveals actionable mutations, and TERT promoter hotspot mutations and TERT gene amplification as likely drivers of progression

Salvatore Piscuoglio; Charlotte K.Y. Ng; Melissa P. Murray; Kathleen A. Burke; Marcia Edelweiss; Felipe C. Geyer; Gabriel S. Macedo; Akiko Inagaki; Anastasios D. Papanastasiou; Luciano G. Martelotto; Caterina Marchiò; Raymond S. Lim; Rafael A. Ioris; Pooja K. Nahar; Ino de Bruijn; Lillian Mary Smyth; Muzaffar Akram; Dara S. Ross; John H.J. Petrini; Larry Norton; David B. Solit; José Baselga; Edi Brogi; Marc Ladanyi; Britta Weigelt; Jorge S. Reis-Filho

Phyllodes tumours (PTs) are breast fibroepithelial lesions that are graded based on histological criteria as benign, borderline or malignant. PTs may recur locally. Borderline PTs and malignant PTs may metastasize to distant sites. Breast fibroepithelial lesions, including PTs and fibroadenomas, are characterized by recurrent MED12 exon 2 somatic mutations. We sought to define the repertoire of somatic genetic alterations in PTs and whether these may assist in the differential diagnosis of these lesions. We collected 100 fibroadenomas, 40 benign PTs, 14 borderline PTs and 22 malignant PTs; six, six and 13 benign, borderline and malignant PTs, respectively, and their matched normal tissue, were subjected to targeted massively parallel sequencing (MPS) using the MSK‐IMPACT sequencing assay. Recurrent MED12 mutations were found in 56% of PTs; in addition, mutations affecting cancer genes (eg TP53, RB1, SETD2 and EGFR) were exclusively detected in borderline and malignant PTs. We found a novel recurrent clonal hotspot mutation in the TERT promoter (−124 C>T) in 52% and TERT gene amplification in 4% of PTs. Laser capture microdissection revealed that these mutations were restricted to the mesenchymal component of PTs. Sequencing analysis of the entire cohort revealed that the frequency of TERT alterations increased from benign (18%) to borderline (57%) and to malignant PTs (68%; p < 0.01), and TERT alterations were associated with increased levels of TERT mRNA (p < 0.001). No TERT alterations were observed in fibroadenomas. An analysis of TERT promoter sequencing and gene amplification distinguished PTs from fibroadenomas with a sensitivity and a positive predictive value of 100% (CI 95.38–100%) and 100% (CI 85.86–100%), respectively, and a sensitivity and a negative predictive value of 39% (CI 28.65–51.36%) and 68% (CI 60.21–75.78%), respectively. Our results suggest that TERT alterations may drive the progression of PTs, and may assist in the differential diagnosis between PTs and fibroadenomas. Copyright


Journal of Clinical Oncology | 2017

AKT Inhibition in Solid Tumors With AKT1 Mutations

David M. Hyman; Lillian Mary Smyth; Mark T.A. Donoghue; Shannon N. Westin; Philippe L. Bedard; Emma Dean; H Bando; Anthony B. El-Khoueiry; J. A Perez-Fidalgo; A Mita; Jan H. M. Schellens; Matthew T. Chang; J. B Reichel; Nancy Bouvier; S. D Selcuklu; Tara Soumerai; J Torrisi; Joseph P. Erinjeri; Helen Ambrose; J. C Barrett; B Dougherty; Andrew Foxley; Justin Lindemann; R McEwen; Martin Pass; Gaia Schiavon; Michael F. Berger; Sarat Chandarlapaty; David B. Solit; Udai Banerji

Purpose AKT1 E17K mutations are oncogenic and occur in many cancers at a low prevalence. We performed a multihistology basket study of AZD5363, an ATP-competitive pan-AKT kinase inhibitor, to determine the preliminary activity of AKT inhibition in AKT-mutant cancers. Patients and Methods Fifty-eight patients with advanced solid tumors were treated. The primary end point was safety; secondary end points were progression-free survival (PFS) and response according to Response Evaluation Criteria in Solid Tumors (RECIST). Tumor biopsies and plasma cell-free DNA (cfDNA) were collected in the majority of patients to identify predictive biomarkers of response. Results In patients with AKT1 E17K-mutant tumors (n = 52) and a median of five lines of prior therapy, the median PFS was 5.5 months (95% CI, 2.9 to 6.9 months), 6.6 months (95% CI, 1.5 to 8.3 months), and 4.2 months (95% CI, 2.1 to 12.8 months) in patients with estrogen receptor-positive breast, gynecologic, and other solid tumors, respectively. In an exploratory biomarker analysis, imbalance of the AKT1 E17K-mutant allele, most frequently caused by copy-neutral loss-of-heterozygosity targeting the wild-type allele, was associated with longer PFS (hazard ratio [HR], 0.41; P = .04), as was the presence of coincident PI3K pathway hotspot mutations (HR, 0.21; P = .045). Persistent declines in AKT1 E17K in cfDNA were associated with improved PFS (HR, 0.18; P = .004) and response ( P = .025). Responses were not restricted to patients with detectable AKT1 E17K in pretreatment cfDNA. The most common grade ≥ 3 adverse events were hyperglycemia (24%), diarrhea (17%), and rash (15.5%). Conclusion This study provides the first clinical data that AKT1 E17K is a therapeutic target in human cancer. The genomic context of the AKT1 E17K mutation further conditioned response to AZD5363.


Molecular Cancer Therapeutics | 2015

Abstract B109: AZD5363, a catalytic pan-Akt inhibitor, in Akt1 E17K mutation positive advanced solid tumors

David M. Hyman; Lillian Mary Smyth; Philippe L. Bedard; Amit M. Oza; Emma Dean; Anne C Armstrong; Joao Paulo Lima; Hideaki Bando; Peter Kabos; J. Alejandro Pérez-Fidalgo; Kathleen N. Moore; Shannon N. Westin; Benoit You; Sarat Chandarlapaty; Leila Alland; Helen Ambrose; Andrew Foxley; Justin Lindemann; Martin Pass; Paul Rugman; Shaista Salim; Gaia Schiavon; Kenji Tamura; José Baselga; Udai Banerji

This abstract has been withheld from publication due to its inclusion in the AACR-NCI-EORTC Molecular Targets Conference 2015 Official Press Program. It will be posted online at the time of its presentation in a press conference or in a session: 10:00 AM ET Saturday, November 7. Citation Format: David M. Hyman, Lillian Smyth, Philippe L. Bedard, Amit Oza, Emma Dean, Anne Armstrong, Joao Lima, Hideaki Bando, Peter Kabos, J. Alejandro Perez-Fidalgo, Kathleen Moore, Shannon N. Westin, Benoit You, Sarat Chandarlapaty, Leila Alland, Helen Ambrose, Andrew Foxley, Justin Lindemann, Martin Pass, Paul Rugman, Shaista Salim, Gaia Schiavon, Kenji Tamura, Jose Baselga, Udai Banerji. AZD5363, a catalytic pan-Akt inhibitor, in Akt1 E17K mutation positive advanced solid tumors. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B109.


Cancer Cell | 2018

The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers

Pedram Razavi; Matthew T. Chang; Guotai Xu; Chaitanya Bandlamudi; Dara S. Ross; Neil Vasan; Yanyan Cai; Craig M. Bielski; Mark T.A. Donoghue; Philip Jonsson; Alexander Penson; Ronglai Shen; Fresia Pareja; Ritika Kundra; Sumit Middha; Michael L. Cheng; Ahmet Zehir; Cyriac Kandoth; Ruchi Patel; Kety Huberman; Lillian Mary Smyth; Komal Jhaveri; Shanu Modi; Tiffany A. Traina; Chau Dang; Wen Zhang; Britta Weigelt; Bob T. Li; Marc Ladanyi; David M. Hyman

We integrated the genomic sequencing of 1,918 breast cancers, including 1,501 hormone receptor-positive tumors, with detailed clinical information and treatment outcomes. In 692 tumors previously exposed to hormonal therapy, we identified an increased number of alterations in genes involved in the mitogen-activated protein kinase (MAPK) pathway and in the estrogen receptor transcriptional machinery. Activating ERBB2 mutations and NF1 loss-of-function mutations were more than twice as common in endocrine resistant tumors. Alterations in other MAPK pathway genes (EGFR, KRAS, among others) and estrogen receptor transcriptional regulators (MYC, CTCF, FOXA1, and TBX3) were also enriched. Altogether, these alterations were present in 22% of tumors, mutually exclusive with ESR1 mutations, and associated with a shorter duration of response to subsequent hormonal therapies.


Cancer Research | 2017

Abstract LB-103: Landscape of somatic ERBB2 Mutations: Findings from AACR GENIE and comparison to ongoing ERBB2 mutant basket study

Alison M. Schram; Helen H. Won; Fabrice Andre; Monica Arnedos; Funda Meric Bernstam; Philippe L. Bedard; Kenna R. Shaw; Hugo M. Horlings; Christine M. Micheel; Ben Ho Park; Grace Mann; Alshad S. Lalani; Lillian Mary Smyth; David B. Solit; Deborah Schrag; Mia A. Levy; Barrett J. Rollins; Mark Routbort; Charles L. Sawyers; Eva Lepisto; Michael F. Berger; David M. Hyman

Background: AACR GENIE is an international data-sharing project that aggregates clinical-grade cancer genomic data. As a demonstration of utility, we evaluated the landscape of ERBB2 mutations in the first 18,486 patients included in this registry and compared it to the first 100 patients enrolled in an ongoing international Phase 2 SUMMIT ‘basket’ study of the pan-HER inhibitor neratinib in ERBB2 mutant solid tumors (NCT01953926). Results: ERBB2 mutations were identified in 2.8% (519/18,486) of patients in the GENIE cohort and observed at all participating centers. In total, there were 482 missense, 66 indels, 19 truncating mutations, and 14 structural variants. A total of 263 unique missense mutations were observed including 12 at previously identified hotspots which accounted for 69.2% of all missense mutations. 35 unique cancer types were represented. The tumor types with the highest proportion of ERBB2 mutations were bladder (12.8%, 82/641), breast (3.9%, 87/2230), colorectal (3.3%, 70/2102), and NSCLC (3%, 90/3006). Among patients with copy number data available (91%) 11% had concurrent ERBB2 amplification, most often in breast cancer. The most frequently observed alterations in ERBB2, adjusted for differing exon coverage between panels, was S310F/Y in 0.46% of the GENIE cohort (12.6% of samples with ERBB2 alterations), Y772_A775dup in 0.21% (6.9%), R678Q in 0.17% (4.5%), L755S in 0.16% (5.2%), V777L in 0.12% (3.8%), and V842I in 0.09% (3.1%). The distribution of specific ERBB2 variants differed significantly by tumor type with exon 20 insertions being most common in NSCLC (44.4%, 40/90), L755S (18.9%, 11/92) in breast, S310F/Y (26.9%, 28/104) in bladder, and V842I (13.9%, 10/72) in colorectal cancer. Structural variants included intragenic deletions (n=4) and fusions involving various partners including GRB7 (n=2), and one each of C1orf87, PPIL6, HEXIM2, THRA, ASIC2, BCA3, WIPF2. The frequencies of ERBB2 mutant cancer types observed in the GENIE cohort were generally comparable to those enrolled to the neratinib basket study including NSCLC (17 vs 22%, respectively), breast (16.4 vs 24%), bladder (15.5 vs 14%), colorectal (13.2 vs 17%), and endometrial (4.2 vs 6%). At the variant level, S310F/Y was less prevalent in GENIE compared to the neratinib study (12.6 vs 24%) while all other mutations were generally similar including L755S (5.2 vs 9%), R678Q (4.5 vs 2%), Y772_A775dup (6.9 vs 13%), V777L (3.8 vs 9%), and V842I (3.1 vs 6%). Conclusion: GENIE confirms that a diversity of ERBB2 mutations are prevalent across a variety of tumor types in patients with advanced cancer. The genomic landscape of ERBB2 mutations was largely similar in the population based GENIE cohort and the neratinib SUMMIT study, providing the first direct evidence that basket study enrollment accurately reflects the true landscape of the target alteration. Citation Format: Alison Schram, Helen H. Won, Fabrice Andre, Monica Arnedos, Funda Meric - Bernstam, Philippe L. Bedard, Kenna R. Shaw, Hugo Horlings, Christine Micheel, Ben Ho Park, Grace Mann, Alshad S. Lalani, Lillian Smyth, David B. Solit, Deborah Schrag, Mia A. Levy, Barrett J. Rollins, Mark Routbort, Charles L. Sawyers, Eva Lepisto, Michael F. Berger, David M. Hyman, on behalf of the AACR Project GENIE Consortium. Landscape of somatic ERBB2 Mutations: Findings from AACR GENIE and comparison to ongoing ERBB2 mutant basket study [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-103. doi:10.1158/1538-7445.AM2017-LB-103


Cancer Research | 2016

Abstract 4509: Clinical genomic profiling of 1000 metastatic breast cancer patients: actionable targets, novel alterations, and clinical correlations

Pedram Razavi; Matthew T. Chang; Sumit Middha; Dara S. Ross; Ahmet Zehir; Tracy Ann Proverbs-Singh; Cyriac Kandoth; Sarat Chandarlapaty; Maura N. Dickler; Jorge S. Reis-Filho; Sujata Patil; Venkatraman E. Seshan; Lillian Mary Smyth; Neil M. Iyengar; Komal Jhaveri; Shanu Modi; Chau T. Dang; Mark E. Robson; Larry Norton; Clifford A. Hudis; Marc Ladanyi; Maurizio Scaltriti; Nikolaus Schultz; David M. Hyman; Michael F. Berger; Barry S. Taylor; David B. Solit; José Baselga

Most large genomic profiling efforts in breast cancer have focused on primary breast tumors. Profiling of metastatic breast cancer (MBC) could more accurately define actionable genomic alterations and reveal novel alterations that arise under the selective pressure and clonal evolution of prior therapy. Utilizing a 410-gene targeted capture-based sequencing platform (MSK-IMPACT), we analyzed 920 tumors (62% metastatic, 38% primary) from 874 MBC patients for somatic mutations, DNA copy number alterations, and structural rearrangements (planned final analysis 1000 patients). Detailed clinical data including treatment outcomes was collected for all patients. The cohort was representative of the well characterized clinical subtypes of breast cancer with 71% ER+ or PR+ and HER2-, 17% HER2+, and 12% triple negative breast cancer (TNBC). Our analysis revealed recurrent alterations in multiple pathways including PI3K/AKT/mTOR (56%), cell cycle regulation (42%), RTK signaling (40%), epigenetic regulation (33%) and MAPK/ERK (19%) pathways. The most frequent actionable alterations include: PIK3CA mutation (36%), ERBB2 amplification (15%), FGFR1 amplification (12%), ESR1 mutation (11%), PTEN mutation/deletion (10%), AKT1 mutation (6%), and ERBB2 mutation (5%). The genomic landscape was significantly different across breast cancer subtypes. For example, the PI3K/AKT/mTOR pathway was altered in ER+ MBC mainly through activating PIK3CA mutations whereas PTEN deletion/mutations were most common in TNBC. We also identified significant differences in the genomic profiles of metastatic and primary tumor samples. Gene enrichment analyses revealed a subset of genes more frequently altered in metastatic tumors (STK11: 13 vs 2; ROS1: 14 vs 2; FGFR4: 15 vs 1) suggesting that mutations in these genes may play a role in breast tumor metastasis and/or therapy resistance. ESR1 mutations were predominantly present in metastatic tumors (88%) and were significantly associated with duration of prior hormonal therapy (P In summary, our genomic analyses of this large cohort of MBC patients revealed actionable alterations in over 60% of the patients. 29% of patients with these alterations were enrolled in clinical trials of matched targeted therapies to date (PIK3CA 27%, AKT1 30%, ESR1 23%, ERBB2 39%) suggesting that prospective genomic characterization can accelerate enrollment of patients with MBC onto therapeutic clinical trials. Citation Format: Pedram Razavi, Matthew T. Chang, Sumit Middha, Dara S. Ross, Ahmet Zehir, Tracy A. Proverbs-Singh, Cyriac Kandoth, Sarat Chandarlapaty, Maura N. Dickler, Jorge S. Reis-Filho, Sujata Patil, Venkatraman Seshan, Lillian Smyth, Neil M. Iyengar, Komal Jhaveri, Shanu Modi, Chau T. Dang, Mark E. Robson, Larry Norton, Clifford A. Hudis, Marc Ladanyi, Maurizio Scaltriti, Nikolaus Schultz, David Hyman, Michael F. Berger, Barry S. Taylor, David B. Solit, Jose Baselga. Clinical genomic profiling of 1000 metastatic breast cancer patients: actionable targets, novel alterations, and clinical correlations. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4509.


Cancer Research | 2016

Abstract P1-14-17: Pathologic complete response rate with doxorubicin and cyclophosphamide followed by weekly paclitaxel with trastuzumab and pertuzumab in patients with HER2-positive early stage breast cancer: A single institution experience

Jasmeet Chadha Singh; Steven Sugarman; Lee W. Jones; Camilla Boafo; Sujata Patil; S Schweber; Anthony F. Yu; Daniel F. Argolo; Shanu Modi; Neil M. Iyengar; Lillian Mary Smyth; Larry Norton; José Baselga; C. Hudis; Chau T. Dang

Background: Trastuzumab and pertuzumab (HP) with standard chemotherapy is approved for use in the neoadjuvant setting. We performed a retrospective analysis of patients (pts) treated with dose-dense doxorubicin and cyclophosphamide (AC) → paclitaxel, trastuzumab, pertuzumab (THP) in the neoadjuvant setting. Here we report the pathologic complete response (pCR) rate. Methods: We abstracted medical records of patients who were treated with pertuzumab-based therapy in the neoadjuvant setting from September 1, 2013 to March 1, 2015. Charts were analyzed for pt demographics, stage of breast cancer, pathology reports, surgical data, and information on systemic therapy. Results: Charts from 66 pts were reviewed; 60 pts were evaluable for pCR defined as absence of invasive disease in the breast, and 6 were not (3-no anthracycline, 1-incomplete chart, 1-no surgery yet, 1-metastatic). Median age was 47 years (range 28-68 years). Of 60 pts, 52 (86%) had operable breast cancer (T1-3, N0-1, M0) of which 7 had clinical stage I disease (T1N0)]; 7 (12%) had locally advanced disease (T2-3, N2-3, M0 or T4a-c, any N, M0), and 1 (2%) had inflammatory breast cancer (T4d, any N, M0). 49 (82%) and 11 (18%) had hormone receptor (HR)-positive and negative diseases, respectively. All patients had HER2-positive breast cancer defined as immunohistochemistry (IHC) 3+ and/or fluorescent in-situ hybridization (FISH) of > 2.0. 30 pts (50%) underwent mastectomy and lumpectomy, respectively. Out of 60 evaluable pts, 41 (68%) had pCR; 32/49 (65%) with HR-positive and 9/11 (82%) with HR-negative diseases had pCR, respectively. Overall 58/60 (97%) pts completed neoadjuvant therapy; 2 did not (1 developed Steven Johnson Syndrome after one cycle of AC and 1 developed pneumonitis after third weekly dose of T with HP). Conclusions: At our single center experience the pCR rate of dose dense AC→THP is high at 68 %. These data are similar to results seen in the TRYPHAENA study, and we await the results from the BERENICE trial evaluating pCR as a secondary endpoint. Citation Format: Singh JC, Sugarman S, Jones L, Boafo C, Patil S, Schweber S, Yu A, Argolo D, Modi S, Iyengar N, Smyth L, Norton L, Baselga J, Hudis C, Dang C. Pathologic complete response rate with doxorubicin and cyclophosphamide followed by weekly paclitaxel with trastuzumab and pertuzumab in patients with HER2-positive early stage breast cancer: A single institution experience. [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 P1-14-17.


Cancer Research | 2016

Abstract P4-13-20: Activity of HP-based therapies as third and later lines for the treatment of HER2-positive metastatic breast cancer: A retrospective study from a single institution

Daniel F. Argolo; M Friedman; Lillian Mary Smyth; Neil M. Iyengar; Jasmeet Chadha Singh; Sujata Patil; Larry Norton; José Baselga; C. Hudis; Chau T. Dang

Background: Dual anti-HER2 blockade with trastuzumab and pertuzumab (HP) plus chemotherapy is an effective therapy (Rx) in the 1st-line setting for HER2-positive metastatic breast cancer (MBC). Our single arm phase II study included patients (pts) treated with HP plus paclitaxel in the 2nd-line setting with progression-free survival (PFS) benefit. Recently, we reported results from a retrospective study of pts treated at our institution, suggesting a longer PFS for those who received HP-based Rxs when compared to any other anti-HER2 based Rxs in the 2nd-line setting. To further assess the activity of this combination in later Rx lines, we conducted a retrospective analysis of pts with HER2-positive MBC who had progressive disease after 2nd-line and were treated with HP-based Rxs in the 3rd and later lines at MSKCC. Historically, the median (med) PFS in this setting with trastuzumab-based Rx is about 3-4 months. Methods: Pts diagnosed with HER2-positive MBC and treated with HP-based Rxs at MSKCC between 1-1-2011 and 03-30-2015 and who progressed on 2nd-line Rx were identified through an institutional database. Primary endpoint was PFS in 3rd and later treatment lines. Results: 70 pts who received any HP-based Rx in the 3rd or later lines of treatment were eligible. The med number of prior anti-HER2 Rx was 3. The baseline characteristics and Rxs are summarized in Table 1. The med PFS for the entire cohort was 5.7 months (95% CI, 4.8-6.5). Conclusions: In this retrospective analysis involving heavly pretreated patients, HP-based Rx appears to be an active regimen and compares favorably to historical data. This supports the NCCN endorsement of HP-based Rx in later lines if HP has not been delivered previously. Citation Format: Argolo D, Friedman M, Smyth L, Iyengar N, Singh J, Patil S, Norton L, Baselga J, Hudis C, Dang C. Activity of HP-based therapies as third and later lines for the treatment of HER2-positive metastatic breast cancer: A retrospective study from a single institution. [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 P4-13-20.


Cancer Research | 2016

Abstract OT3-01-09: Phase II study of gemcitabine, trastuzumab, and pertuzumab in the treatment of metastatic HER2-positive breast cancer after prior trastuzumab/pertuzumab- or pertuzumab-based therapy

Neil M. Iyengar; Daniel F. Argolo; Lillian Mary Smyth; Mf Chen; C. Hudis; Chau T. Dang

Background: Despite significant therapeutic advances in the treatment of breast cancers that overexpress human epidermal growth factor receptor-2 (HER2), the large majority of patients (pts) with metastatic disease will experience progression. The development of new HER2 directed therapies has led to improvements in progression free and overall survival; however optimal therapeutic sequencing remains under study. The combination of taxanes with trastuzumab (H) and pertuzumab (P) is active in the first line setting; however the efficacy of continuing dual anti-HER2 therapy with HP after initial progression is unknown. Trial design: This is a single arm phase II trial of gemcitabine (G) with HP for pts with metastatic HER2-positive breast cancer who have had prior HP-based treatment. G is administered at 1200 mg/m2 on days 1 and 8 of an every 3 week cycle. H is given 8 mg/kg load → 6 mg/kg every 3 weeks or 4 mg/kg load → 2 mg/kg every week. P is given 840 mg load → 420 mg every 3 weeks. All agents will be given intravenously. This trial is currently enrolling pts. Eligibility criteria: Eligible pts are adults with stage IV HER2-positive (3+ IHC or FISH ≥ 2.0 of primary or metastatic site) breast cancer. Pts may have received prior treatment with HP- or P-based therapy in the (neo)adjuvant, unresectable, locally advanced, or metastatic setting. In the metastatic setting, ≤ 3 prior chemotherapies are permitted. Prior G is allowed only if it was not combined with P. Bone-only non-measurable disease is permitted. Adequate organ and bone marrow function, left ventricular ejection fraction > 50%, and ECOG status ≤ 1 are required. Pts with previously treated brain metastases stable for ≥ 2 months may be enrolled. Specific aims: The primary objective is to determine the progression free survival (PFS) at 3 months. Secondary objectives include overall survival, safety and tolerability. An exploratory endpoint is to compare PFS determined by RECIST criteria versus 18-F FDG-PET response criteria. Statistical methods: Based on historical data, the anticipated median PFS in this setting is 3 months. The study will be considered positive if the 3-month PFS is ≥ 70%. A Simon optimal 2-stage design will be used. If 12/21 pts in stage 1 are alive and progression free at 3 months, stage 2 will accrue for a total of 45 pts. If at least 27/45 pts are alive and progression free, the trial will be deemed a success. This design assumes a 10% type I and type II error. Present accrual and target accrual: 3 out of a planned 45 pts have been enrolled. Contact information: For more information, please visit clinicaltrials.gov (NCT02252887). Citation Format: Iyengar NM, Argolo D, Smyth L, Chen MF, Hudis CA, Dang CT. Phase II study of gemcitabine, trastuzumab, and pertuzumab in the treatment of metastatic HER2-positive breast cancer after prior trastuzumab/pertuzumab- or pertuzumab-based therapy. [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 OT3-01-09.


Indian Journal of Medical and Paediatric Oncology | 2015

Adjuvant hormonal therapy in premenopausal women with breast cancer.

Lillian Mary Smyth; Clifford A. Hudis

Duration of tamoxifen Tamoxifen use was truncated at 5 years based on the National Surgical Adjuvant Breast and Bowel Project (NSABP) B14 study. While this trial initially showed that 5 years of treatment was effective as compared to no-tamoxifen for hormone receptor positive, node negative breast cancer (mostly postmenopausal), a re-randomization of the treated cohort suggested that 10 years of therapy could be inferior to five.[9,10] This observation guided practice globally.

Collaboration


Dive into the Lillian Mary Smyth's collaboration.

Top Co-Authors

Avatar

José Baselga

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Chau T. Dang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David M. Hyman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Larry Norton

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Neil M. Iyengar

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Clifford A. Hudis

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael F. Berger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sarat Chandarlapaty

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Daniel F. Argolo

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David B. Solit

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge