A. Jo Chien
University of California, San Francisco
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by A. Jo Chien.
Cancer | 2012
J. Letourneau; E. Ebbel; Patricia P. Katz; A. Katz; Wei Z. Ai; A. Jo Chien; Michelle E. Melisko; Marcelle I. Cedars; M.P. Rosen
The post‐treatment quality of life (QOL) impacts of receiving precancer‐treatment infertility counseling and of pursuing fertility preservation have not been described in large‐scale studies of reproductive age women with cancer.
The New England Journal of Medicine | 2016
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.).
Cancer | 2012
J. Letourneau; E. Ebbel; Patricia P. Katz; Kutluk Oktay; Charles E. McCulloch; Wei Z. Ai; A. Jo Chien; Michelle E. Melisko; Marcelle I. Cedars; M.P. Rosen
The authors sought to describe the age‐specific impact of infertility and early menopause after chemotherapy among reproductive age women with cancer.
Seminars in Oncology | 2008
A. Jo Chien; Mark M. Moasser
Taxanes and anthracyclines are two of the most potent and broadly effective classes of chemotherapeutic agents. However, resistance to these agents is common and significantly limits their potential. As such, there is a great need to understand the mechanisms underlying de novo and acquired resistance to these agents. Beyond the resistance barrier lies even greater potential to significantly alter the natural course of human cancer. This review discusses what we currently understand about the mechanisms of resistance to taxanes and anthracyclines. Preclinical models suggest a role for ATP-binding cassette transporters, tubulin isoforms, microtubule-associated proteins, tubulin gene mutations, and mitotic checkpoint signaling proteins in resistance to taxanes. Preclinical models also suggest that drug transport proteins, antioxidant defenses, apoptotic signaling, and topoisomerase modulation may mediate anthracycline resistance. Many of these hypotheses remain untested in appropriately designed clinical studies, but limited clinical evidence will be reviewed. Epothilones represent a novel class of non-taxane microtubule stabilizing agents with distinct drug-resistance profiles. Potential mechanisms behind these differences and their potential role in the treatment of both taxane- and anthracycline-refractory patients are discussed.
Expert Opinion on Drug Safety | 2010
A. Jo Chien; Hope S. Rugo
Importance of the field: Trastuzumab has become a mainstay in the treatment of women with human epidermal growth factor receptor-2 (HER2)-overexpressing breast cancer in the metastatic and adjuvant settings. Although trastuzumab is generally well tolerated, cardiac toxicity has emerged as a rare but potentially serious complication that limits its use in some patients. It is critically important to understand the nature of this cardiac risk when counseling patients, especially as new anti-HER2 agents are developed and tested in combination with trastuzumab. Areas covered in this review: This review describes the incidence, risk factors and natural history of trastuzumab-associated cardiac toxicity reported in updated efficacy and cardiac safety analyses of metastatic and adjuvant trastuzumab clinical trials. Mechanisms of trastuzumab-associated cardiotoxicity are proposed and compared to what is known about anthracycline-induced cardiotoxicity. The existing cardiac safety data for lapatinib and other newer HER2-targeted therapies are also discussed, both as single agents and in combination with trastuzumab. What the reader will gain: The reader will gain a comprehensive understanding of the existing cardiac safety data for trastuzumab including the notable differences in trial design and study populations between each of the major adjuvant trials. Readers will become familiar with the risk factors associated with trastuzumab-induced cardiotoxicity as well as with the natural history of its course. Take home message: The majority of trastuzumab-related cardiac events observed have been asymptomatic declines in left ventricular ejection fraction. The incidence of severe congestive heart failure and cardiac death observed in the large adjuvant trastuzumab trials ranges from 0.6 to 4%. Both symptomatic and asymptomatic events are largely reversible and manageable; however, little is known about the significance of asymptomatic left ventricular ejection fraction decline and longer cardiac follow-up is needed. Close cardiac monitoring must be performed for all patients receiving anti-HER2 agents currently in the clinic or in development.
Breast Cancer Research and Treatment | 2013
A. Jo Chien; Hope S. Rugo
The widespread use of trastuzumab in the past decade has led to a significant and measureable improvement in the survival of patients with human epidermal growth factor receptor-2 (HER2) overexpressing breast cancer, and in many ways has redefined the natural history of this aggressive breast cancer subtype. Historically, survival in patients with HER2-positive disease was dictated by the systemic disease course, and what appears to be the central nervous system (CNS) tropism associated with HER2-amplified tumors was not clinically evident. With improved systemic control and prolonged survival, the incidence of brain metastases has increased, and CNS disease, often in the setting of well-controlled extracranial disease, is proving to be an increasingly important and clinically challenging cause of morbidity and mortality in patients with HER2-positive advanced breast cancer. This review summarizes the known clinical data for the systemic treatment of HER2-positive CNS metastases and includes information about ongoing clinical trials of novel therapies as well as emerging strategies for early detection and prevention.
Cancer Research | 2014
John W. Park; Minetta C. Liu; Douglas Yee; Angela DeMichele; Laura J. van 't Veer; Nola M. Hylton; Fraser Symmans; Meredith Buxton; A. Jo Chien; Amy Wallace; Michelle E. Melisko; Richard Schwab; Judy C. Boughey; Debashish Tripathy; Hank Kaplan; Rita Nanda; Stephen Y. Chui; Kathy S. Albain; Stacy L. Moulder; Anthony Elias; Julie E. Lang; Kirsten Edminston; Donald W. Northfelt; David M. Euhus; Qamar J. Khan; Julia Lyandres; Sarah E. Davis; Christina Yau; Ashish Sanil; Laura Esserman
Background: I-SPY 2 is a multicenter, phase II neoadjuvant trial in women with high-risk stage II/III breast cancer using adaptive randomization within biomarker subtypes to evaluate novel agents added to standard chemotherapy. Primary endpoint is pathologic complete response (pCR). Goal is to identify regimens that meet a high Bayesian predictive probability of statistical significance in a neoadjuvant 300-patient phase III trial defined by hormone-receptor (HR), HER2 status, and MammaPrint (MP). Experimental regimens may “graduate” in 1 of 10 signatures, with a maximum of 120 patients. We report efficacy results for neratinib (N). Methods: Tumors ≥2.5cm by clinical exam & ≥2cm by imaging are eligible for screening. MP low risk/HR+/HER2- tumors are ineligible for randomization. Patients receive chemotherapy (paclitaxel qwk x 12, doxorubicin and cyclophosphamide q2-3 wk x 4, T->AC). HER2- pts were randomized to N+T->AC vs. T->AC and HER2+ pts to N+T->AC vs. trastuzumab+T->AC. Analysis is intent-to-treat with pts who switch to non-protocol therapy regarded as non-pCRs. We provide estimated pCR rates (95% Bayesian probability intervals), probabilities of superiority of neratinib over control, and Bayesian predictive probabilities of success in an equally randomized phase III trial. Results: Neratinib met the predictive probability criterion in HR-/HER2+, “graduated”, and accrual ceased [115 N patients (65 HER2+), 78 concurrently randomized controls (22 HER2+)]. The table shows results for all 10 signatures. Two patients (1 N and 1 control) withdrew consent and are not included. Conclusion: I-SPY 29s standing trial mechanism efficiently evaluates agents in biomarker-defined patient subsets. In a modest number of patients, adaptive randomization successfully identified a biomarker signature (HR-/HER2+) for neratinib9s further development. All HER2+ and MP+ tumors may also benefit from this regimen, consistent with preclinical data. Evaluation in I-SPY 3, a phase III registration trial, is planned. Citation Format: John W. Park, Minetta C. Liu, Douglas Yee, Angela DeMichele, Laura van 9t Veer, Nola Hylton, Fraser Symmans, Meredith B. Buxton, A. Jo Chien, Amy Wallace, Michelle Melisko, Richard Schwab, Judy Boughey, Debashish Tripathy, Hank Kaplan, Rita Nanda, Stephen Chui, Kathy S. Albain, Stacy Moulder, Anthony Elias, Julie E. Lang, Kirsten Edminston, Donald Northfelt, David Euhus, Qamar Khan, Julia Lyandres, Sarah E. Davis, Christina Yau, Ashish Sanil, Laura J. Esserman, Donald A. Berry. Neratinib plus standard neoadjuvant therapy for high-risk breast cancer: Efficacy results from the I-SPY 2 TRIAL. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr CT227. doi:10.1158/1538-7445.AM2014-CT227
Clinical Cancer Research | 2013
Angela DeMichele; Donald A. Berry; JoAnne Zujewski; Sally Hunsberger; Larry Rubinstein; Joseph E. Tomaszewski; Gary J. Kelloff; Jane Perlmutter; Meredith Buxton; Julia Lyandres; Kathy S. Albain; Christopher C. Benz; A. Jo Chien; Paul Haluska; Brian Leyland-Jones; Minetta C. Liu; Pamela N. Munster; Olufunmilayo I. Olopade; John W. Park; Barbara A. Parker; Lajos Pusztai; Debu Tripathy; Hope S. Rugo; Douglas Yee; Laura Esserman
New approaches to drug development are critically needed to lessen the time, cost, and resources necessary to identify and optimize active agents. Strategies to accelerate drug development include testing drugs earlier in the disease process, such as the neoadjuvant setting. The U.S. Food and Drug Administration (FDA) has issued guidance designed to accelerate drug approval through the use of neoadjuvant studies in which the surrogate short-term endpoint, pathologic response, can be used to identify active agents and shorten the time to approval of both efficacious drugs and biomarkers identifying patients most likely to respond. However, this approach has unique challenges. In particular, issues of patient safety are paramount, given the exposure of potentially curable patients to investigational agents with limited safety experience. Key components to safe drug development in the neoadjuvant setting include defining a study population at sufficiently poor prognosis with standard therapy to justify exposure to investigational agents, defining the extent and adequacy of safety data from phase I, detecting potentially harmful interactions between investigational and standard therapies, improving study designs, such as adaptive strategies, that limit patient exposure to ineffective agents, and intensifying safety monitoring in the course of the trial. The I-SPY2 trial is an example of a phase II neoadjuvant trial of novel agents for breast cancer in which these issues have been addressed, both in the design and conduct of the trial. These adaptations of phase II design enable acceleration of drug development by reducing time and cost to screen novel therapies for activity without compromising safety. Clin Cancer Res; 19(11); 2817–23. ©2013 AACR.
Journal of Clinical Oncology | 2014
A. Jo Chien; Pamela N. Munster; Michelle E. Melisko; Hope S. Rugo; John W. Park; Andrei Goga; Glenna M. Auerback; Elham Khanafshar; Karen Ordovas; Kevin M. Koch; Mark M. Moasser
PURPOSE The highly effective treatment of human epidermal growth factor receptor (HER) 2-amplified breast cancer has proven challenging because of a signal buffering capacity inherent in the functionally relevant HER2-HER3 target. HER2-HER3 signaling can be inactivated by doses of lapatinib that fully inactivate the HER2 kinase. In mouse models, such doses are not tolerable in continuous administration, but they are tolerable and highly effective in intermittent dosing. We pursued the clinical translation of this treatment hypothesis. PATIENTS AND METHODS We conducted a phase I dose-escalation study in women with advanced HER2-overexpressing breast cancer. Lapatinib was administered on days 1 through 5 of repeating 14-day cycles. Dose escalation was conducted using a 3+3 design with plasma lapatinib level monitoring. RESULTS Forty patients were evaluable for toxicity, and 34 patients were evaluable for dose-limiting toxicity (DLT). Lapatinib dose was escalated to 7,000 mg per day in twice-daily dosing with no DLTs; however, plasma lapatinib concentrations plateaued in this dose range. Additional cohorts evaluated strategies to increase lapatinib exposure, including the food effect, CYP3A4 inhibition, and dose fractionation. Of these, only ketoconazole was able to increase lapatinib exposure, despite highly variable lapatinib bioavailability. Intolerable exposure levels were not encountered. Eight patients (20%) experienced grade 3 diarrhea. Six patients achieved a response, and dramatic responses were seen in three patients with lapatinib concentrations approaching 10,000 ng/mL. CONCLUSION Lapatinib exposure can be safely and significantly increased through intermittent dosing but reaches a ceiling that currently impedes clinical translation of the treatment hypothesis. Preliminary efficacy data suggest that exposures approaching those seen in mouse models can result in highly significant tumor responses.
The New England Journal of Medicine | 2016
John W. Park; Minetta C. Liu; Douglas Yee; Christina Yau; Laura J. van 't Veer; W. Fraser Symmans; Melissa Paoloni; Jane Perlmutter; Nola M. Hylton; Michael Hogarth; Angela DeMichele; Meredith Buxton; A. Jo Chien; Anne M. Wallace; Judy C. Boughey; Tufia C. Haddad; Stephen Y. Chui; Kathleen A. Kemmer; Henry G. Kaplan; Claudine Isaacs; Rita Nanda; Debasish Tripathy; Kathy S. Albain; Kirsten K. Edmiston; Anthony Elias; Donald W. Northfelt; Lajos Pusztai; Stacy L. Moulder; Julie E. Lang; Rebecca K. Viscusi