Network


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

Hotspot


Dive into the research topics where Stacy L. Moulder is active.

Publication


Featured researches published by Stacy L. Moulder.


Journal of Clinical Oncology | 2012

PI3K/AKT/mTOR Inhibitors in Patients With Breast and Gynecologic Malignancies Harboring PIK3CA Mutations

Filip Janku; Jennifer J. Wheler; Shannon N. Westin; Stacy L. Moulder; Aung Naing; Apostolia M. Tsimberidou; Siqing Fu; Gerald S. Falchook; David S. Hong; Ignacio Garrido-Laguna; Rajyalakshmi Luthra; J. Jack Lee; Karen H. Lu; Razelle Kurzrock

PURPOSE Mutations of the PIK3CA gene may predict response to phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) inhibitors. Concomitant mutations in the mitogen-activated protein kinase (MAPK) pathway may mediate resistance. PATIENTS AND METHODS Tumors from patients with breast, cervical, endometrial, and ovarian cancer referred to the Clinical Center for Targeted Therapy (Phase I Program) were analyzed for PIK3CA, KRAS, NRAS, and BRAF mutations. Patients with PIK3CA mutations were treated, whenever feasible, with agents targeting the PI3K/AKT/mTOR pathway. RESULTS Of 140 patients analyzed, 25 (18%) had PIK3CA mutations, including five of 14 patients with squamous cell cervical, seven of 29 patients with endometrial, six of 29 patients with breast, and seven of 60 patients with ovarian cancers. Of the 25 patients with PIK3CA mutations, 23 (median of two prior therapies) were treated on a protocol that included a PI3K/AKT/mTOR pathway inhibitor. Two (9%) of 23 patients had stable disease for more than 6 months, and seven patients (30%) had a partial response. In comparison, only seven (10%) of 70 patients with the same disease types but with wild-type PIK3CA treated on the same protocols responded (P = .04). Seven patients (30%) with PIK3CA mutations had coexisting MAPK pathway (KRAS, NRAS, BRAF) mutations (ovarian cancer, n = 5; endometrial cancer, n = 2), and two of these patients (ovarian cancer) achieved a response. CONCLUSION PIK3CA mutations were detected in 18% of tested patients. Patients with PIK3CA mutations treated with PI3K/AKT/mTOR inhibitors demonstrated a higher response rate than patients without mutations. A subset of patients with ovarian cancer with simultaneous PIK3CA and MAPK mutations responded to PI3K/AKT/mTOR inhibitors, suggesting that not all patients demonstrate resistance when the MAPK pathway is concomitantly activated.


Molecular Cancer Therapeutics | 2011

PIK3CA mutations in patients with advanced cancers treated with PI3K/AKT/mTOR axis inhibitors

Filip Janku; Apostolia M. Tsimberidou; Ignacio Garrido-Laguna; Xuemei Wang; Rajyalakshmi Luthra; David S. Hong; Aung Naing; Gerald S. Falchook; John Moroney; Sarina Anne Piha-Paul; Jennifer J. Wheler; Stacy L. Moulder; Siqing Fu; Razelle Kurzrock

Preclinical data suggest that PIK3CA mutations predict response to PI3K/AKT/mTOR inhibitors. Concomitant KRAS or BRAF mutations may mediate resistance. Therefore, tumors from patients referred to the phase I program for targeted therapy starting in October 2008 were analyzed for PIK3CA mutations using PCR-based DNA sequencing of exons 9 and 20. Consecutive patients with diverse tumor types and PIK3CA mutation were treated whenever possible with agents targeting the PI3K/AKT/mTOR pathway. Overall, PIK3CA mutations were detected in 25 of 217 patients (11.5%; exon 9, n = 11; exon 20, n = 14). In tumor types with more than 10 patients tested, PIK3CA mutations were most frequent in endometrial (3 of 14, 21%), ovarian (5 of 30, 17%), colorectal (9 of 54, 17%), breast (2 of 14, 14%), cervical (2 of 15, 13%), and squamous cell cancer of the head and neck (1 of 11, 9%). Of the 25 patients with PIK3CA mutations, 17 (68%) were treated on a protocol that included a PI3K/AKT/mTOR pathway inhibitor, and 6 (35%) achieved a partial response. In contrast, only 15 of 241 patients (6%) without documented PIK3CA mutations treated on the same protocols responded (P = 0.001). Of the 17 patients with PIK3CA mutations, 6 (35%) had simultaneous KRAS or BRAF mutations (colorectal, n = 4; ovarian, n = 2). Colorectal cancer patients with PIK3CA and KRAS mutations did not respond to therapy, whereas both ovarian cancer patients with PIK3CA and KRAS or BRAF mutations did. In conclusion, PIK3CA mutations were detected in 11.5% of patients with diverse solid tumors. The response rate was significantly higher for patients with PIK3CA mutations treated with PI3K/AKT/mTOR pathway inhibitors than for those without documented mutations. Mol Cancer Ther; 10(3); 558–65. ©2011 AACR.


Annals of Oncology | 2009

Prognostic impact of discordance between triple-receptor measurements in primary and recurrent breast cancer

Cornelia Liedtke; Kristine Broglio; Stacy L. Moulder; Limin Hsu; Shu-Wan Kau; W. F. Symmans; Constance Albarracin; Funda Meric-Bernstam; Wendy A. Woodward; Richard L. Theriault; Ludwig Kiesel; Gabriel N. Hortobagyi; Lajos Pusztai; A. M. Gonzalez-Angulo

BACKGROUND We evaluated discordance in expression measurements for estrogen receptor (ER), progesterone receptor (PR), and HER2 between primary and recurrent tumors in patients with recurrent breast cancer and its effect on prognosis. METHODS A total of 789 patients with recurrent breast cancer were studied. ER, PR, and HER2 status were determined by immunohistochemistry (IHC) and/or FISH. Repeat markers for ER, PR, and HER2 were available in 28.9%, 27.6%, and 70.0%, respectively. Primary and recurrent tumors were classified as triple receptor-negative breast cancer (TNBC) or receptor-positive breast cancer (RPBC, i.e. expressing at least one receptor). Discordance was correlated with clinical/pathological parameters. RESULTS Discordance for ER, PR, and HER2 was 18.4%, 40.3%, and 13.6%, respectively. Patients with concordant RPBC had significantly better post-recurrence survival (PRS) than discordant cases; patients with discordant receptor status had similarly unfavorable survival as patients with concordant TNBC. IHC scores for ER and PR showed weak concordance between primary and recurrent tumors. Concordance of HER2-FISH scores was higher. CONCLUSIONS Concordance of quantitative hormone receptor measurements between primary and recurrent tumors is modest consistent with suboptimal reproducibility of measurement methods, particularly for IHC. Discordant cases have poor survival probably due to inappropriate use of targeted therapies. However, biological change in clinical phenotype cannot be completely excluded.


The New England Journal of Medicine | 2016

Adaptive Randomization of Veliparib–Carboplatin Treatment in Breast Cancer

Hope S. Rugo; Olufunmilayo I. Olopade; Angela DeMichele; Christina Yau; Laura J. van 't Veer; Meredith Buxton; Michael Hogarth; Nola M. Hylton; Melissa Paoloni; Jane Perlmutter; W. Fraser Symmans; Douglas Yee; A. Jo Chien; Anne M. Wallace; Henry G. Kaplan; Judy C. Boughey; Tufia C. Haddad; Kathy S. Albain; Minetta C. Liu; Claudine Isaacs; Qamar J. Khan; Julie E. Lang; Rebecca K. Viscusi; Lajos Pusztai; Stacy L. Moulder; Stephen Y. Chui; Kathleen A. Kemmer; Anthony Elias; Kirsten K. Edmiston; David M. Euhus

BACKGROUND The genetic and clinical heterogeneity of breast cancer makes the identification of effective therapies challenging. We designed I-SPY 2, a phase 2, multicenter, adaptively randomized trial to screen multiple experimental regimens in combination with standard neoadjuvant chemotherapy for breast cancer. The goal is to match experimental regimens with responding cancer subtypes. We report results for veliparib, a poly(ADP-ribose) polymerase (PARP) inhibitor, combined with carboplatin. METHODS In this ongoing trial, women are eligible for participation if they have stage II or III breast cancer with a tumor 2.5 cm or larger in diameter; cancers are categorized into eight biomarker subtypes on the basis of status with regard to human epidermal growth factor receptor 2 (HER2), hormone receptors, and a 70-gene assay. Patients undergo adaptive randomization within each biomarker subtype to receive regimens that have better performance than the standard therapy. Regimens are evaluated within 10 biomarker signatures (i.e., prospectively defined combinations of biomarker subtypes). Veliparib-carboplatin plus standard therapy was considered for HER2-negative tumors and was therefore evaluated in 3 signatures. The primary end point is pathological complete response. Tumor volume changes measured by magnetic resonance imaging during treatment are used to predict whether a patient will have a pathological complete response. Regimens move on from phase 2 if and when they have a high Bayesian predictive probability of success in a subsequent phase 3 neoadjuvant trial within the biomarker signature in which they performed well. RESULTS With regard to triple-negative breast cancer, veliparib-carboplatin had an 88% predicted probability of success in a phase 3 trial. A total of 72 patients were randomly assigned to receive veliparib-carboplatin, and 44 patients were concurrently assigned to receive control therapy; at the completion of chemotherapy, the estimated rates of pathological complete response in the triple-negative population were 51% (95% Bayesian probability interval [PI], 36 to 66%) in the veliparib-carboplatin group versus 26% (95% PI, 9 to 43%) in the control group. The toxicity of veliparib-carboplatin was greater than that of the control. CONCLUSIONS The process used in our trial showed that veliparib-carboplatin added to standard therapy resulted in higher rates of pathological complete response than standard therapy alone specifically in triple-negative breast cancer. (Funded by the QuantumLeap Healthcare Collaborative and others; I-SPY 2 TRIAL ClinicalTrials.gov number, NCT01042379.).


Cell Reports | 2014

Assessing PIK3CA and PTEN in early-phase trials with PI3K/AKT/mTOR inhibitors

Filip Janku; David S. Hong; Siqing Fu; Sarina Anne Piha-Paul; Aung Naing; Gerald S. Falchook; Apostolia M. Tsimberidou; Vanda M. Stepanek; Stacy L. Moulder; J. Jack Lee; Rajyalakshmi Luthra; Ralph Zinner; Russell Broaddus; Jennifer J. Wheler; Razelle Kurzrock

Despite a wealth of preclinical studies, it is unclear whether PIK3CA or phosphatase and tensin homolog (PTEN) gene aberrations are actionable in the clinical setting. Of 1,656 patients with advanced, refractory cancers tested for PIK3CA or PTEN abnormalities, PIK3CA mutations were found in 9% (146/1,589), and PTEN loss and/or mutation was found in 13% (149/1,157). In multicovariable analysis, treatment with a phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) inhibitor was the only independent factor predicting response to therapy in individuals harboring a PIK3CA or PTEN aberration. The rate of stable disease ≥6 months/partial response reached 45% in a subgroup of individuals with H1047R PIK3CA mutations. Aberrations in the PI3K/AKT/mTOR pathway are common and potentially actionable in patients with diverse advanced cancers. This work provides further important clinical validation for continued and accelerated use of biomarker-driven trials incorporating rational drug combinations.


Clinical Pharmacology & Therapeutics | 2008

Advances in the Treatment of Breast Cancer

Stacy L. Moulder; Gabriel N. Hortobagyi

Breast cancer is the most commonly diagnosed cancer in American women and is the second leading cause of cancer‐related deaths in this population. This review highlights how the use of a multidisciplinary approach to the management of diseases of the breast and the introduction of novel systemic therapies have improved the quality of life and survival in patients with breast cancer.


Seminars in Oncology | 2002

HER (erbB) tyrosine kinase inhibitors in the treatment of breast cancer.

Carlos L. Arteaga; Stacy L. Moulder; F. Michael Yakes

Protein tyrosine kinases are tightly regulated enzymes that play an important role in the control of most fundamental cellular processes, including cell proliferation, differentiation, metabolism, migration, and survival. These signaling proteins are the frequent target of oncogenic mutations or other genetic alterations leading to dysregulated tyrosine kinase activity, cellular transformation, and subsequent tumor progression. Many of the known dominant oncogenes encode aberrant protein tyrosine kinases and are causally associated with a significant fraction of human neoplasms, including breast carcinoma. The epidermal growth factor receptor and HER2/neu are two transmembrane tyrosine kinases that are members of the HER (erbB) signaling network. Aberrant signaling by this network is present in a cohort of breast carcinomas. Structure/function studies of these kinases have led to the identification of molecular approaches aimed at disabling signaling by this transforming network. Trastuzumab, a monoclonal antibody that binds the ectodomain of HER2, was recently shown to induce regression of HER2-overexpressing breast cancers, confirming the role of HER2 in tumor maintenance and progression. A rational therapeutic approach that builds on these results with trastuzumab and expands the targeting of the HER network will be presented.


Journal of Clinical Oncology | 2006

Targeted Inhibition of Farnesyltransferase in Locally Advanced Breast Cancer: A Phase I and II Trial of Tipifarnib Plus Dose-Dense Doxorubicin and Cyclophosphamide

Joseph A. Sparano; Stacy L. Moulder; Aslamuzzaman Kazi; Linda T. Vahdat; Tianhong Li; Christine M. Pellegrino; Pam Munster; Mokenge P. Malafa; David Lee; Shira Hoschander; Una Hopkins; Dawn L. Hershman; John J. Wright; Said M. Sebti

PURPOSE To determine the recommended phase II dose (RPTD) of the farnesyltransferase (FTase) inhibitor tipifarnib when combined with doxorubicin and cyclophosphamide (AC) in patients with advanced breast cancer, the pathologic complete response (pCR) rate after preoperative treatment with four cycles of the combination in locally advanced breast cancer (LABC), and the effect of tipifarnib on primary tumor FTase enzyme activity in vivo. PATIENTS AND METHODS Thirty-two patients with metastatic breast cancer (n = 11) or LABC (n = 21) received AC (doxorubicin 60 mg/m2 and cyclophosphamide 600 mg/m2) administered intravenously on day 1 plus tipifarnib (100, 200, or 300 mg bid for 6 to 14 days) without (n = 2) or with (n = 30) granulocyte colony-stimulating factor (G-CSF) for up to four cycles. Patients with LABC underwent surgery after up to four cycles of the combination. RESULTS When combined with AC every 2 weeks plus G-CSF, the RPTD of tipifarnib was 200 mg bid administered on days 2 to 7. Seven (33%) of 21 patients (95% CI, 15% to 55%) with LABC treated with up to four cycles of the combination at the RPTD had a pCR in the breast at surgery. The five patients had serial biopsies that demonstrated at least 50% FTase enzyme inhibition in the primary tumor (median, 100%; range, 55% to 100%) after tipifarnib. CONCLUSION Tipifarnib may be safely combined with dose-dense AC using a dose and schedule that significantly inhibits FTase enzyme activity in human breast cancer in vivo and may enhance the pCR rate after four cycles of preoperative dose-dense AC.


Molecular Cancer Therapeutics | 2012

Safety, Pharmacokinetics, and Activity of GRN1005, a Novel Conjugate of Angiopep-2, a Peptide Facilitating Brain Penetration, and Paclitaxel, in Patients with Advanced Solid Tumors

Razelle Kurzrock; Nashat Y. Gabrail; Chandtip Chandhasin; Stacy L. Moulder; Carrie Smith; Andrew Brenner; Kamalesh Kumar Sankhala; Alain C. Mita; Kelly Elian; Danielle Bouchard; John Sarantopoulos

GRN1005 is a novel peptide–drug conjugate composed of paclitaxel covalently linked to a peptide, angiopep-2, that targets the low-density lipoprotein receptor-related protein 1. This first-in-human study evaluated the safety, tolerability, pharmacokinetics, and efficacy of GRN1005 in patients with advanced solid tumors. Patients in sequential cohorts (one patient per cohort until grade 2 toxicity, then 3 + 3 design) received intravenous GRN1005 at escalating doses between 30 and 700 mg/m2 once in every 21 days. In the maximum tolerated dose (MTD) expansion group, patients were required to have brain metastases. Fifty-six patients received GRN1005, including 41 with brain metastases (median number of prior therapies = 4). MTD was 650 mg/m2; the main dose-limiting toxicity was myelosuppression. Sixteen of 20 patients dosed at the MTD had brain metastases. Pharmacokinetics was dose linear and the mean terminal-phase elimination half-life was 3.6 hours. No evidence of accumulation was observed after repeat dosing. No anti-GRN1005 antibodies were detected. Five of the 20 patients (25%) dosed at 650 mg/m2 (MTD), three of whom had previous taxane therapy, achieved an overall partial response (breast, n = 2; non–small cell lung cancer, n = 2; and ovarian cancer, n = 1); responses in all five patients were also accompanied by shrinkage of brain lesions (−17% to −50%). In addition, six patients (11%; doses 30–700 mg/m2) experienced stable disease that lasted 4 months or more. GRN1005 was well tolerated and showed activity in heavily pretreated patients with advanced solid tumors, including those who had brain metastases and/or failed prior taxane therapy. Mol Cancer Ther; 11(2); 308–16. ©2011 AACR.


Clinical Cancer Research | 2009

Phase I Trial of a Combination of the Multikinase Inhibitor Sorafenib and the Farnesyltransferase Inhibitor Tipifarnib in Advanced Malignancies

David S. Hong; Said M. Sebti; Robert A. Newman; Michelle A. Blaskovich; Lei Ye; Robert F. Gagel; Stacy L. Moulder; Jennifer J. Wheler; Aung Naing; Nizar M. Tannir; Chaan S. Ng; Steven I. Sherman; Adel El Naggar; Rabia Khan; J. C. Trent; John J. Wright; Razelle Kurzrock

Purpose: We evaluated the safety, maximum tolerated dose, pharmacokinetics, and biological effects of the combination of the Raf-1, RET, KIT, platelet-derived growth factor receptor, and vascular endothelial growth factor receptor 2 kinase inhibitor sorafenib and the farnesyltransferase inhibitor tipifarnib. Experimental Design: A standard 3 + 3 phase I dose-escalation design was used with a 28-day cycle (sorafenib daily and tipifarnib for 21 days, by mouth). Results: Fifty patients were treated; 43 reached restaging evaluation after cycle 2. The most common side effects were grade 1 to 2 rash, hyperglycemia, and diarrhea. Dose-limiting toxicity was rash, and the recommended phase II dose is sorafenib 400 mg p.o. qam/200 mg p.o. qpm and tipifarnib p.o. 100 mg bd. Despite the low doses of tipifarnib, one quarter of patients had 50 reduction in farnesyltransferase levels. Interestingly, six of eight patients with medullary thyroid cancer had durable stable disease (n = 3) or partial remissions (n = 3), lasting 12 to 26+ months. Five of the six responders had available tissue, and RET gene mutations were identified in them. Prolonged (6 months) stable disease was also seen in nine patients as follows: papillary thyroid cancer (n = 4; 18+ to 27+ months), adrenocortical cancer (n = 2; 7 and 11 months), and one each of melanoma (platelet-derived growth factor receptor mutation positive; 14 months), renal (6 months), and pancreatic cancer (6 months). Conclusions: Our study shows that the combination of tipifarnib and sorafenib is well tolerated. Activity was seen, especially in patients with medullary thyroid cancer, a tumor characterized by RET mutations. (Clin Cancer Res 2009;15(22):70618)

Collaboration


Dive into the Stacy L. Moulder's collaboration.

Researchain Logo
Decentralizing Knowledge