Network


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

Hotspot


Dive into the research topics where Antoinette R. Tan is active.

Publication


Featured researches published by Antoinette R. Tan.


Journal of Clinical Oncology | 2010

Neratinib, an Irreversible ErbB Receptor Tyrosine Kinase Inhibitor, in Patients With Advanced ErbB2-Positive Breast Cancer

Harold J. Burstein; Yan Sun; Luc Dirix; Z Jiang; Robert Paridaens; Antoinette R. Tan; Ahmad Awada; Anantbhushan Ranade; Shunchang Jiao; Gary E. Schwartz; Richat Abbas; Christine Powell; Kathleen Turnbull; Jennifer Vermette; Charles Zacharchuk; Rajendra A. Badwe

PURPOSE Neratinib is an oral, irreversible pan-ErbB receptor tyrosine kinase inhibitor. The efficacy and safety of neratinib were evaluated in two cohorts of patients with advanced ErbB2-positive breast cancer-those with and those without prior trastuzumab treatment-in an open-label, multicenter, phase II trial. PATIENTS AND METHODS Patients in the two cohorts (prior trastuzumab, n = 66; no prior trastuzumab, n = 70) received oral neratinib 240 mg once daily. The primary end point was the 16-week progression-free survival (PFS) rate for the evaluable population (prior trastuzumab, n = 63; no prior trastuzumab, n = 64), as assessed by independent review. RESULTS The 16-week PFS rates were 59% for patients with prior trastuzumab treatment and 78% for patients with no prior trastuzumab treatment. Median PFS was 22.3 and 39.6 weeks, respectively. Objective response rates were 24% among patients with prior trastuzumab treatment and 56% in the trastuzumab-naïve cohort. The most common adverse events were diarrhea, nausea, vomiting, and fatigue. Diarrhea was the most frequent grades 3 to 4 adverse event, occurring in 30% of patients with prior trastuzumab treatment and in 13% of patients with no prior trastuzumab treatment, which prompted dose reductions in 29% and 4% of patients, respectively, but treatment discontinuation in only one patient. No neratinib-related, grades 3 or 4 cardiotoxicity was reported. CONCLUSION Oral neratinib showed substantial clinical activity and was reasonably well tolerated among both heavily pretreated and trastuzumab-naïve patients who had advanced, ErbB2-positive breast cancer. Diarrhea was the most common adverse effect but was manageable with antidiarrheal agents and dose modification.


Journal of Clinical Oncology | 2009

Phase II study of eribulin mesylate, a halichondrin B analog, in patients with metastatic breast cancer previously treated with an anthracycline and a taxane.

Linda T. Vahdat; Brian Pruitt; Carol J. Fabian; Ragene Rivera; David A. Smith; Elizabeth Tan-Chiu; Jonathan L. Wright; Antoinette R. Tan; Noshir Anthony Dacosta; Ellen Chuang; John W. Smith; Joyce O'Shaughnessy; Dale Shuster; Nicole Meneses; Kumari Chandrawansa; Fang Fang; Patricia E. Cole; Simon Ashworth; Joanne L. Blum

PURPOSE Eribulin mesylate (E7389), a nontaxane microtubule dynamics inhibitor, is a structurally simplified, synthetic analog of the marine natural product halichondrin B. This open-label, single-arm, phase II study evaluated efficacy and tolerability of eribulin in heavily pretreated patients with metastatic breast cancer (MBC). METHODS MBC patients who were previously treated with an anthracycline and a taxane received eribulin mesylate (1.4 mg/m(2)) as a 2- to 5-minute intravenous (IV) infusion on days 1, 8, and 15 of a 28-day cycle. Because of neutropenia (at day 15), an alternative regimen of eribulin on days 1 and 8 of a 21-day cycle was administered. The primary end point was overall response rate. RESULTS Of the 103 patients treated, the median number of prior chemotherapy regimens was four (range, one to 11 regimens). In the per-protocol population (n = 87), eribulin achieved an independently reviewed objective response rate (all partial responses [PRs]) of 11.5% (95% CI, 5.7 to 20.1) and a clinical benefit rate (PR plus stable disease > or = 6 months) of 17.2% (95% CI, 10.0 to 26.8). The median duration of response was 171 days (5.6 months; range, 44 to 363 days), the median progression-free survival was 79 days (2.6 months; range, 1 to 453 days), and the median overall survival was 275 days (9.0 months; range, 15 to 826 days). The most common drug-related grades 3 to 4 toxicities were as follows: neutropenia, 64%; leukopenia, 18%; fatigue, 5%; peripheral neuropathy, 5%; and febrile neutropenia, 4%. CONCLUSION Eribulin demonstrated activity with manageable tolerability (including infrequent grade 3 and no grade 4 neuropathy) in heavily pretreated patients with MBC when dosed as a short IV infusion on days 1 and 8 of a 21-day cycle.


Journal of Clinical Oncology | 2004

Evaluation of Biologic End Points and Pharmacokinetics in Patients With Metastatic Breast Cancer After Treatment With Erlotinib, an Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor

Antoinette R. Tan; Xiaowei Yang; Stephen M. Hewitt; Arlene Berman; Erin R. Lepper; Alex Sparreboom; Allyson Parr; William D. Figg; Catherine Chow; Seth M. Steinberg; Stephen L. Bacharach; Millie Whatley; Jorge A. Carrasquillo; Jaime S. Brahim; Seth Ettenberg; Stan Lipkowitz; Sandra M. Swain

PURPOSE To evaluate changes in epidermal growth factor receptor (EGFR) phosphorylation and its downstream signaling in tumor and surrogate tissue biopsies in patients with metastatic breast cancer treated with erlotinib, an EGFR tyrosine kinase inhibitor, and to assess relationships between biomarkers in tumor and normal tissues and between biomarkers and pharmacokinetics. PATIENTS AND METHODS Eighteen patients were treated orally with 150 mg/d of erlotinib. Ki67, EGFR, phosphorylated EGFR (pEGFR), phosphorylated mitogen-activated protein kinase (pMAPK), and phosphorylated AKT (pAKT) in 15 paired tumor, skin, and buccal mucosa biopsies (at baseline and after 1 month of therapy) were examined by immunohistochemistry and analyzed quantitatively. Pharmacokinetic sampling was also obtained. RESULTS The stratum corneum layer and Ki67 in keratinocytes of the epidermis in 15 paired skin biopsies significantly decreased after treatment (P = .0005 and P = .0003, respectively). No significant change in Ki67 was detected in 15 tumors, and no responses were observed. One was EGFR-positive and displayed heterogeneous expression of the receptor, and 14 were EGFR-negative. In the EGFR-positive tumor, pEGFR, pMAPK, and pAKT were reduced after treatment. Paradoxically, pEGFR was increased in EGFR-negative tumors post-treatment (P = .001). Although markers were reduced in surrogate and tumor tissues in the patient with EGFR-positive tumor, no apparent associations were observed in patients with EGFR-negative tumor. CONCLUSION Erlotinib has inhibitory biologic effects on normal surrogate tissues and on an EGFR-positive tumor. The lack of reduced tumor proliferation may be attributed to the heterogeneous expression of receptor in the EGFR-positive patient and absence of target in this cohort of heavily pretreated patients.


Journal of Clinical Oncology | 2002

Phase I Clinical and Pharmacokinetic Study of Flavopiridol Administered as a Daily 1-Hour Infusion in Patients With Advanced Neoplasms

Antoinette R. Tan; Donna Headlee; Richard A. Messmann; Edward A. Sausville; Susan G. Arbuck; Anthony J. Murgo; Giovanni Melillo; Suoping Zhai; William D. Figg; Sandra M. Swain; Adrian M. Senderowicz

PURPOSE To define the maximum-tolerated dose (MTD), dose-limiting toxicity, and pharmacokinetics of the cyclin-dependent kinase inhibitor flavopiridol administered as a daily 1-hour infusion every 3 weeks. PATIENTS AND METHODS Fifty-five patients with advanced neoplasms were treated with flavopiridol at doses of 12, 17, 24, 30, 37.5, and 52.5 mg/m(2)/d for 5 days; doses of 50 and 62.5 mg/m(2)/d for 3 days; and doses of 62.5 and 78 mg/m(2)/d for 1 day. Plasma sampling was performed to characterize the pharmacokinetics of flavopiridol with these schedules. RESULTS Dose-limiting neutropenia developed at doses >/= 52.5 mg/m(2)/d. Nonhematologic toxicities included nausea, vomiting, diarrhea, hypotension, and a proinflammatory syndrome characterized by anorexia, fatigue, fever, and tumor pain. The median peak concentrations of flavopiridol achieved at the MTDs on the 5-day, 3-day, and 1-day schedule were 1.7 micro mol/L (range, 1.3 to 4.2 micro mol/L), 3.2 micro mol/L (range, 1.7 to 4.8 micro mol/L), and 3.9 micro mol/L (1.8 to 5.1 micro mol/L), respectively. Twelve patients had stable disease for >/= 3 months, with a median duration of 6 months (range, 3 to 11 months). CONCLUSION The recommended phase II doses of flavopiridol as a 1-hour infusion are 37.5 mg/m(2)/d for 5 days, 50 mg/m(2)/d for 3 days, and 62.5 mg/m(2)/d for 1 day. Flavopiridol as a daily 1-hour infusion can be safely administered and can achieve concentrations in the micromolar range, sufficient to inhibit cyclin-dependent kinases in preclinical models. Further studies to determine the optimal schedule of flavopiridol as a single agent and in combination with chemotherapeutic agents are underway.


PLOS ONE | 2014

Phase I Study of GC1008 (Fresolimumab): A Human Anti-Transforming Growth Factor-Beta (TGFβ) Monoclonal Antibody in Patients with Advanced Malignant Melanoma or Renal Cell Carcinoma

John C. Morris; Antoinette R. Tan; Thomas Olencki; Geoffrey I. Shapiro; Bruce J. Dezube; Michael Reiss; Frank J. Hsu; Jay A. Berzofsky; Donald P. Lawrence

Background In advanced cancers, transforming growth factor-beta (TGFβ) promotes tumor growth and metastases and suppresses host antitumor immunity. GC1008 is a human anti-TGFβ monoclonal antibody that neutralizes all isoforms of TGFβ. Here, the safety and activity of GC1008 was evaluated in patients with advanced malignant melanoma and renal cell carcinoma. Methods In this multi-center phase I trial, cohorts of patients with previously treated malignant melanoma or renal cell carcinoma received intravenous GC1008 at 0.1, 0.3, 1, 3, 10, or 15 mg/kg on days 0, 28, 42, and 56. Patients achieving at least stable disease were eligible to receive Extended Treatment consisting of 4 doses of GC1008 every 2 weeks for up to 2 additional courses. Pharmacokinetic and exploratory biomarker assessments were performed. Results Twenty-nine patients, 28 with malignant melanoma and 1 with renal cell carcinoma, were enrolled and treated, 22 in the dose-escalation part and 7 in a safety cohort expansion. No dose-limiting toxicity was observed, and the maximum dose, 15 mg/kg, was determined to be safe. The development of reversible cutaneous keratoacanthomas/squamous-cell carcinomas (4 patients) and hyperkeratosis was the major adverse event observed. One malignant melanoma patient achieved a partial response, and six had stable disease with a median progression-free survival of 24 weeks for these 7 patients (range, 16.4–44.4 weeks). Conclusions GC1008 had no dose-limiting toxicity up to 15 mg/kg. In patients with advanced malignant melanoma and renal cell carcinoma, multiple doses of GC1008 demonstrated acceptable safety and preliminary evidence of antitumor activity, warranting further studies of single agent and combination treatments. Trial Registration Clinicaltrials.gov NCT00356460


Clinical Cancer Research | 2009

Phase I Study of Eribulin Mesylate Administered Once Every 21 Days in Patients with Advanced Solid Tumors

Antoinette R. Tan; Eric H. Rubin; Diana C. Walton; Dale Shuster; Y. Nancy Wong; Fang Fang; Simon Ashworth; Lee S. Rosen

Purpose: To evaluate the maximum tolerated dose (MTD), dose-limiting toxicities (DLT), and pharmacokinetics of eribulin mesylate (E7389), a halichondrin B analogue, administered every 21 days in patients with advanced solid tumors. Experimental Design: Eribulin mesylate was given as a 1-hour infusion every 21 days at doses of 0.25, 0.5, 1, 2, 2.8, and 4 mg/m2. The MTD was identified using an accelerated titration design. The pharmacokinetics of eribulin were evaluated in the plasma and urine with the first dose. Results: Twenty-one patients were enrolled. At 4 mg/m2, three patients experienced a DLT of febrile neutropenia on day 7. The dose level was reduced to 2.8 mg/m2 where two of three patients experienced dose-limiting febrile neutropenia. Six additional patients were enrolled at 2 mg/m2 (seven patients in total received this dose) and one of these patients experienced a neutropenic DLT. The MTD of eribulin mesylate was therefore 2 mg/m2. Nonhematologic toxicities included alopecia, fatigue, anorexia, and nausea. Pharmacokinetic analysis showed linear kinetics for eribulin over the dose range studied and a terminal half-life of 2 days. The plasma-concentration-time profile exhibited a rapid distribution phase followed by a slow elimination phase. Drug clearance was nonrenal. One patient with non–small cell lung cancer achieved an unconfirmed partial response and 12 patients had stable disease. Conclusions: Eribulin mesylate administered as a 1-hour infusion every 21 days has a manageable toxicity profile at 2 mg/m2, with further dose escalation limited by neutropenia.


Breast Cancer Research and Treatment | 2009

Transforming growth factor-β signaling: emerging stem cell target in metastatic breast cancer?

Antoinette R. Tan; Gabriela Alexe; Michael Reiss

In most human breast cancers, lowering of TGFβ receptor- or Smad gene expression combined with increased levels of TGFβs in the tumor microenvironment is sufficient to abrogate TGFβs tumor suppressive effects and to induce a mesenchymal, motile and invasive phenotype. In genetic mouse models, TGFβ signaling suppresses de novo mammary cancer formation but promotes metastasis of tumors that have broken through TGFβ tumor suppression. In mouse models of “triple-negative” or basal-like breast cancer, treatment with TGFβ neutralizing antibodies or receptor kinase inhibitors strongly inhibits development of lung- and bone metastases. These TGFβ antagonists do not significantly affect tumor cell proliferation or apoptosis. Rather, they de-repress anti-tumor immunity, inhibit angiogenesis and reverse the mesenchymal, motile, invasive phenotype characteristic of basal-like and HER2-positive breast cancer cells. Patterns of TGFβ target genes upregulation in human breast cancers suggest that TGFβ may drive tumor progression in estrogen-independent cancer, while it mediates a suppressive host cell response in estrogen-dependent luminal cancers. In addition, TGFβ appears to play a key role in maintaining the mammary epithelial (cancer) stem cell pool, in part by inducing a mesenchymal phenotype, while differentiated, estrogen receptor-positive, luminal cells are unresponsive to TGFβ because the TGFBR2 receptor gene is transcriptionally silent. These same cells respond to estrogen by downregulating TGFβ, while antiestrogens act by upregulating TGFβ. This model predicts that inhibiting TGFβ signaling should drive the differentiation of mammary stem cells into ductal cells. Consequently, TGFβ antagonists may convert basal-like or HER2-positive cancers to a more epithelioid, non-proliferating (and, perhaps, non-metastatic) phenotype. Conversely, these agents might antagonize the therapeutic effects of anti-estrogens in estrogen-dependent luminal cancers. These predictions need to be addressed prospectively in clinical trials and should inform the selection of patient populations most likely to benefit from this novel anti-metastatic therapeutic approach.


Clinical Cancer Research | 2006

Pharmacokinetics of Cetuximab After Administration of Escalating Single Dosing and Weekly Fixed Dosing in Patients with Solid Tumors

Antoinette R. Tan; Dirk F. Moore; Manuel Hidalgo; James H. Doroshow; Elizabeth Poplin; Susan Goodin; David J. Mauro; Eric H. Rubin

Purpose: Previous studies of cetuximab pharmacokinetics did not fully characterize its elimination phase. The purpose of this trial was to evaluate the pharmacokinetics of cetuximab given as a single dose followed by weekly fixed repeated dosing in patients with solid tumors. Experimental Design: Patients were randomly assigned to treatment with a single 2-hour infusion of cetuximab at doses of 50, 100, 250, 400, or 500 mg/m2 followed 3 weeks later by weekly 1-hour infusions of cetuximab at a fixed dose of 250 mg/m2. Extended pharmacokinetic sampling was collected through 504 hours after the first drug administration. Trough samples were obtained before each fixed weekly dose. Single and multidose pharmacokinetic variables were correlated with clinical outcomes. Results: Forty patients were enrolled. Pharmacokinetic analysis confirmed previous reports of nonlinear pharmacokinetics for cetuximab. Modeling studies predicted a 90% saturation of clearance at a dose of 260 mg/m2. Analyses of weekly trough concentrations indicated a slight accumulation of drug concentrations following repeated weekly dosing. Correlative studies indicated a significant association between cetuximab clearance and both body surface area (P = 0.002) and weight (P = 0.002). The occurrence of rash was significantly associated with disease stability (P < 0.002) but not with cetuximab pharmacokinetic variables. Conclusions: Pharmacokinetic results support using body surface area or weight in calculating individual cetuximab doses. A weekly dose of 250 mg/m2 is predicted to nearly fully saturate cetuximab clearance and, by inference, epidermal growth factor receptors. The association between rash and disease stability supports further prospective studies of this relationship.


Cancer Research | 2016

Abstract S5-07: Preliminary efficacy and safety of pembrolizumab (MK-3475) in patients with PD-L1–positive, estrogen receptor-positive (ER+)/HER2-negative advanced breast cancer enrolled in KEYNOTE-028

Hope S. Rugo; J-P Delord; S-A Im; Patrick A. Ott; Sarina Anne Piha-Paul; Philippe L. Bedard; Jasgit C. Sachdev; C. Le Tourneau; E. van Brummelen; A. Varga; Sanatan Saraf; Dina Pietrangelo; Vassiliki Karantza; Antoinette R. Tan

Background: The programmed cell death 1 (PD-1) pathway is used by tumors to evade immune surveillance. Pembrolizumab is a humanized anti–PD-1 monoclonal antibody that blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2. Pembrolizumab has shown robust antitumor activity against several advanced malignancies, including triple-negative breast cancer. We assessed the safety and efficacy of pembrolizumab in patients with PD-L1–positive, ER+/HER2-negative advanced breast cancer. Methods: KEYNOTE-028 (ClinicalTrials.gov, NCT02054806) is an ongoing multicohort, open-label phase 1b study evaluating the safety and efficacy of pembrolizumab in patients with PD-L1–positive advanced solid tumors. Key eligibility criteria for this cohort include ER+ and HER2-negative tumor status defined by institutional standards, locally advanced or metastatic disease, ECOG performance status of 0 or 1, failure of or inability to receive standard therapy, and PD-L1 expression in stroma or in ≥1% of tumor cells as assessed at a central laboratory using a prototype immunohistochemistry assay with the 22C3 antibody (Merck). Pembrolizumab was administered at a dose of 10 mg/kg every 2 weeks for up to 24 months or until confirmed progression or intolerable toxicity. Response is based on RECIST v1.1 as assessed by investigator review every 8 weeks for the first 6 months and every 12 weeks thereafter. Primary efficacy end point is overall response rate (ORR). Results: Of the 248 patients with ER+/HER2-negative breast cancer who had evaluable tumor samples screened for PD-L1 expression, 48 (19%) had PD-L1–positive tumors. Of these, 25 patients were enrolled. Median age was 53 years (range, 36-79), and 44% of patients had an ECOG performance status of 1. Patients were heavily pretreated, with 76% having received ≥3 prior lines of therapy for advanced disease, including 48.0% who received ≥5 prior lines. Analyses of ORR, duration of response, and adverse events are ongoing and will be completed by September 4, 2015. Conclusion: Data from this KEYNOTE-028 cohort will provide information on the antitumor activity and safety of pembrolizumab in patients with heavily pretreated, PD-L1–positive, ER+/HER2-negative advanced breast cancer. Citation Format: Rugo HS, Delord J-P, Im S-A, Ott PA, Piha-Paul SA, Bedard PL, Sachdev J, Le Tourneau C, van Brummelen E, Varga A, Saraf S, Pietrangelo D, Karantza V, Tan A. Preliminary efficacy and safety of pembrolizumab (MK-3475) in patients with PD-L1–positive, estrogen receptor-positive (ER+)/HER2-negative advanced breast cancer enrolled in KEYNOTE-028. [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 S5-07.


Cancer Journal | 2008

Therapeutic Strategies for Triple-Negative Breast Cancer

Antoinette R. Tan; Sandra M. Swain

Triple-negative breast cancer (TNBC) is a clinically relevant term referring to breast carcinomas that do not express the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor type 2 and became operational after human epidermal growth factor receptor type 2 testing was introduced. This is a challenging disease to treat because of the absence of a specific target, but these tumors are sensitive to chemotherapy. An improved understanding of the biology of TNBC has led to evaluation of DNA-damaging chemotherapy drugs, specifically, platinum compounds, and several targeted agents, including poly(ADP-ribose) polymerase inhibitors, epidermal growth factor receptor inhibitors, angiogenesis inhibitors, microtubule inhibitors, Src inhibitors, checkpoint kinase I inhibitors, mammalian target of rapamycin inhibitors, androgen receptor blocker, tumor necrosis factor-related apoptosis-inducing ligand receptor agonists, and transforming growth factor-β antagonists, that may lead to improved clinical outcomes. Ongoing clinical trials will further define the optimal chemotherapy regimen and most effective targeted therapeutic strategy for TNBC.

Collaboration


Dive into the Antoinette R. Tan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandra M. Swain

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Norman Wolmark

Allegheny Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alice P. Chen

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge